Selective κ Opioid Antagonists nor-BNI, GNTI and JDTic Have Low Affinities for Non-Opioid Receptors and Transporters

Background Nor-BNI, GNTI and JDTic induce selective κ opioid antagonism that is delayed and extremely prolonged, but some other effects are of rapid onset and brief duration. The transient effects of these compounds differ, suggesting that some of them may be mediated by other targets. Results In binding assays, the three antagonists showed no detectable affinity (K i≥10 µM) for most non-opioid receptors and transporters (26 of 43 tested). There was no non-opioid target for which all three compounds shared detectable affinity, or for which any two shared sub-micromolar affinity. All three compounds showed low nanomolar affinity for κ opioid receptors, with moderate selectivity over μ and δ (3 to 44-fold). Nor-BNI bound weakly to the α2C-adrenoceptor (K i = 630 nM). GNTI enhanced calcium mobilization by noradrenaline at the α1A-adrenoceptor (EC50 = 41 nM), but did not activate the receptor, displace radioligands, or enhance PI hydrolysis. This suggests that it is a functionally-selective allosteric enhancer. GNTI was also a weak M1 receptor antagonist (K B = 3.7 µM). JDTic bound to the noradrenaline transporter (K i = 54 nM), but only weakly inhibited transport (IC50 = 1.1 µM). JDTic also bound to the opioid-like receptor NOP (K i = 12 nM), but gave little antagonism even at 30 µM. All three compounds exhibited rapid permeation and active efflux across Caco-2 cell monolayers. Conclusions Across 43 non-opioid CNS targets, only GNTI exhibited a potent functional effect (allosteric enhancement of α1A-adrenoceptors). This may contribute to GNTI's severe transient effects. Plasma concentrations of nor-BNI and GNTI may be high enough to affect some peripheral non-opioid targets. Nonetheless, κ opioid antagonism persists for weeks or months after these transient effects dissipate. With an adequate pre-administration interval, our results therefore strengthen the evidence that nor-BNI, GNTI and JDTic are highly selective κ opioid antagonists.

[1]  Andrew Coop,et al.  Kappa opioid antagonists: Past successes and future prospects , 2005, The AAPS Journal.

[2]  Graeme Milligan,et al.  Allostery at G Protein-Coupled Receptor Homo- and Heteromers: Uncharted Pharmacological Landscapes , 2010, Pharmacological Reviews.

[3]  Y. Daali,et al.  P‐glycoprotein is not involved in the differential oral potency of naloxone and naltrexone , 2009, Fundamental & clinical pharmacology.

[4]  Allison T. Knoll,et al.  Dynorphin, stress, and depression , 2010, Brain Research.

[5]  Cécile Béguin,et al.  Kappa-opioid ligands in the study and treatment of mood disorders. , 2009, Pharmacology & therapeutics.

[6]  F. Monsma,et al.  Moving from the Orphanin FQ Receptor to an Opioid Receptor Using Four Point Mutations* , 1996, The Journal of Biological Chemistry.

[7]  P. Voorn,et al.  Dynorphin displaces binding at the glycine site of the NMDA receptor in the rat striatum , 2007, Neuroscience Letters.

[8]  B. Cohen,et al.  Medicinal Chemistry of Kappa Opioid Receptor Antagonists , 2009 .

[9]  Maria F. Sassano,et al.  Automated design of ligands to polypharmacological profiles , 2012, Nature.

[10]  S. Inan Pharmacological and neuroanatomical analysis of GNTI-induced repetitive behavior in mice , 2010 .

[11]  N. Dun,et al.  Inhibitory effect of lidocaine on pain and itch using formalin-induced nociception and 5'-guanidinonaltrindole-induced scratching models in mice: behavioral and neuroanatomical evidence. , 2009, European journal of pharmacology.

[12]  Bryan L. Roth,et al.  Structure of the Nociceptin/Orphanin FQ Receptor in Complex with a Peptide Mimetic , 2012, Nature.

[13]  Francesco Berardi,et al.  Perspectives of P-glycoprotein modulating agents in oncology and neurodegenerative diseases: pharmaceutical, biological, and diagnostic potentials. , 2010, Journal of medicinal chemistry.

[14]  S. Husbands,et al.  In vivo and in vitro characterization of naltrindole-derived ligands at the κ-opioid receptor , 2013, Journal of psychopharmacology.

[15]  P. Plenge,et al.  An affinity-modulating site on neuronal monoamine transport proteins. , 1997, Pharmacology & toxicology.

[16]  Y. Kuraishi,et al.  Noradrenergic regulation of itch transmission in the spinal cord mediated by α-adrenoceptors , 2011, Neuropharmacology.

[17]  M. Bruchas,et al.  Long-Acting κ Opioid Antagonists Disrupt Receptor Signaling And Produce Noncompetitive Effects By Activating C-Jun N-Terminal Kinase* , 2007, Journal of Biological Chemistry.

[18]  B. Cohen,et al.  Duration of Action of a Broad Range of Selective κ-Opioid Receptor Antagonists Is Positively Correlated with c-Jun N-Terminal Kinase-1 Activation , 2011, Molecular Pharmacology.

[19]  A. Cowan,et al.  Kappa-Opioid Antagonists as Pruritogenic Agents , 2009 .

[20]  Stephen A. Wring,et al.  Passive Permeability and P-Glycoprotein-Mediated Efflux Differentiate Central Nervous System (CNS) and Non-CNS Marketed Drugs , 2002, Journal of Pharmacology and Experimental Therapeutics.

[21]  N. Dun,et al.  Investigation of gastrin-releasing peptide as a mediator for 5′-guanidinonaltrindole-induced compulsive scratching in mice , 2011, Peptides.

[22]  R. Raffa,et al.  Pilocarpine-induced reciprocal hindlimb scratching in mice , 1987, Pharmacology Biochemistry and Behavior.

[23]  M. Heilig,et al.  The kappa opioid receptor antagonist JDTic attenuates alcohol seeking and withdrawal anxiety , 2012, Addiction biology.

[24]  F. L. Moore,et al.  A subset of kappa opioid ligands bind to the membrane glucocorticoid receptor in an amphibian brain. , 2000, Endocrinology.

[25]  N. Dun,et al.  Nalfurafine prevents 5′-guanidinonaltrindole- and compound 48/80-induced spinal c-fos expression and attenuates 5′-guanidinonaltrindole-elicited scratching behavior in mice , 2009, Neuroscience.

[26]  M. Nitta,et al.  Structure-activity relationship study on alpha1 adrenergic receptor antagonists from beer. , 2009, Bioorganic & medicinal chemistry letters.

[27]  J. Kamei,et al.  Norbinaltorphimine, a selective kappa-opioid receptor antagonist, induces an itch-associated response in mice. , 2001, European journal of pharmacology.

[28]  R. Bodnar,et al.  Reductions in locomotor activity following central opioid receptor subtype antagonists in rats , 1996, Physiology & Behavior.

[29]  R. Lewis,et al.  Conopeptide ρ-TIA Defines a New Allosteric Site on the Extracellular Surface of the α1B-Adrenoceptor*♦ , 2012, The Journal of Biological Chemistry.

[30]  S. Yamada,et al.  Characterization of specific [3H]nociceptin binding in rat brain and spinal cord. , 2001, Biological & pharmaceutical bulletin.

[31]  A. Levine,et al.  Nor-binaltorphimine decreases deprivation and opioid-induced feeding , 1990, Brain Research.

[32]  J. Docherty Subtypes of functional α1-adrenoceptor , 2010, Cellular and Molecular Life Sciences.

[33]  R. Raffa,et al.  Evidence that reciprocal hindlimb scratching elicited in mice by intrathecal administration of muscarinic agonists is mediated through M1 type receptors. , 1987, Life sciences.

[34]  T. Endoh,et al.  Nor-binaltorphimine: a potent and selective kappa-opioid receptor antagonist with long-lasting activity in vivo. , 1992, Archives internationales de pharmacodynamie et de therapie.

[35]  Bryan L. Roth,et al.  Structure of the human kappa opioid receptor in complex with JDTic , 2012, Nature.

[36]  Kristina Luthman,et al.  Caco-2 monolayers in experimental and theoretical predictions of drug transport1PII of original article: S0169-409X(96)00415-2. The article was originally published in Advanced Drug Delivery Reviews 22 (1996) 67–84.1 , 2001 .

[37]  I. Hidalgo,et al.  Assessing the absorption of new pharmaceuticals. , 2001, Current topics in medicinal chemistry.

[38]  B. Cohen,et al.  Long-acting κ opioid antagonists nor-BNI, GNTI and JDTic: pharmacokinetics in mice and lipophilicity , 2012, BMC pharmacology.

[39]  P. Wellman Norepinephrine and the control of food intake. , 2000, Nutrition.

[40]  D. Perez Structure–function of α1-adrenergic receptors , 2007 .

[41]  M. Braude,et al.  Preclinical toxicity studies of naltrexone. , 1976, NIDA research monograph.

[42]  D. Lambert,et al.  Pharmacological characterization of the nociceptin / orphanin FQ receptor antagonist SB-612111: in vivo studies , 2007 .

[43]  Hong Zou,et al.  Effect of GNTI, a kappa opioid receptor antagonist, on MK-801-induced hyperlocomotion and stereotypy in mice , 2006, Acta Pharmacologica Sinica.

[44]  A. Levine,et al.  The kappa-opioid antagonist GNTI reduces U50,488-, DAMGO-, and deprivation-induced feeding, but not butorphanol- and neuropeptide Y-induced feeding in rats , 2001, Brain Research.

[45]  Willem Soudijn,et al.  Allosteric modulation of G-protein-coupled receptors , 2001 .

[46]  Quantification of Functional Selectivity at the Human α1A-Adrenoceptor , 2011, Molecular Pharmacology.

[47]  A. Ford,et al.  Facilitatory Interplay in α1a and β2 Adrenoceptor Function Reveals a Non-Gq Signaling Mode: Implications for Diversification of Intracellular Signal Transduction , 2009, Molecular Pharmacology.

[48]  D. Lambert,et al.  Pharmacological Characterization of the Nociceptin/Orphanin FQ Receptor Antagonist SB-612111 [(–)-cis-1-Methyl-7-[[4-(2,6-dichlorophenyl)piperidin-1-yl]methyl]-6,7,8,9-tetrahydro-5H-benzocyclohepten-5-ol]: In Vitro Studies , 2007, Journal of Pharmacology and Experimental Therapeutics.

[49]  B. Cohen,et al.  Repeated Exposure to the κ-Opioid Receptor Agonist Salvinorin A Modulates Extracellular Signal-Regulated Kinase and Reward Sensitivity , 2011, Biological Psychiatry.

[50]  H. Yamamura,et al.  Extremely long-lasting antagonistic actions of nor-binaltorphimine (nor-BNI) in the mouse tail-flick test. , 1992, The Journal of pharmacology and experimental therapeutics.

[51]  W. Klemm,et al.  Alpha noradrenergic agonists promote catalepsy in the mouse , 1988, Pharmacology Biochemistry and Behavior.

[52]  E. Nielsen,et al.  Validation of a fluorescence-based high-throughput assay for the measurement of neurotransmitter transporter uptake activity , 2008, Journal of Neuroscience Methods.

[53]  R. Westerink,et al.  Caveats and limitations of plate reader-based high-throughput kinetic measurements of intracellular calcium levels. , 2011, Toxicology and applied pharmacology.

[54]  K. Luthman,et al.  Caco-2 monolayers in experimental and theoretical predictions of drug transport. , 2001, Advanced drug delivery reviews.

[55]  S. Lazareno,et al.  Allosteric interactions between the antagonist prazosin and amiloride analogs at the human alpha(1A)-adrenergic receptor. , 2000, Molecular pharmacology.

[56]  N. Mello,et al.  Kappa opioid antagonist effects of the novel kappa antagonist 5′-guanidinonaltrindole (GNTI) in an assay of schedule-controlled behavior in rhesus monkeys , 2002, Psychopharmacology.

[57]  F. Ehlert,et al.  Selectivity of Agonists for the Active State of M1 to M4 Muscarinic Receptor Subtypes , 2009, Journal of Pharmacology and Experimental Therapeutics.

[58]  S. Knapp,et al.  Structural Determinants of G-protein α Subunit Selectivity by Regulator of G-protein Signaling 2 (RGS2)* , 2009, The Journal of Biological Chemistry.

[59]  D. Perez,et al.  Binding, partial agonism, and potentiation of alpha(1)-adrenergic receptor function by benzodiazepines: A potential site of allosteric modulation. , 1999, The Journal of pharmacology and experimental therapeutics.

[60]  R. Stevens,et al.  Structure of the human k-opioid receptor in complex with JDTic , 2012 .