Characterization of a Potential KOR/DOR Dual Agonist with No Apparent Abuse Liability via a Complementary Structure-Activity Relationship Study on Nalfurafine Analogues.

Discovery of analgesics void of abuse liability is critical to battle the opioid crisis in the United States. Among many strategies to achieve this goal, targeting more than one opioid receptor seems promising to minimize this unwanted side effect while achieving a reasonable therapeutic profile. In the process of understanding the structure-activity relationship of nalfurafine, we identified a potential analgesic agent, NMF, as a dual kappa opioid receptor/delta opioid receptor agonist with minimum abuse liability. Further characterizations, including primary in vitro ADMET studies (hERG toxicity, plasma protein binding, permeability, and hepatic metabolism), and in vivo pharmacodynamic and toxicity profiling (time course, abuse liability, tolerance, withdrawal, respiratory depression, body weight, and locomotor activity) further confirmed NMF as a promising drug candidate for future development.

[1]  R. Tallarida,et al.  Modulation of Morphine Analgesia, Antinociceptive Tolerance, and Mu-Opioid Receptor Binding by the Cannabinoid CB2 Receptor Agonist O-1966 , 2022, Frontiers in Pharmacology.

[2]  M. Hajizadeh,et al.  Effect of Methadone Maintenance on Expression of BDNF and CREB Genes in Brain VTA of Male Morphine Treated Rats. , 2021, Central nervous system agents in medicinal chemistry.

[3]  F. Bihel,et al.  Comprehensive overview of biased pharmacology at the opioid receptors: biased ligands and bias factors. , 2021, RSC medicinal chemistry.

[4]  M. Bouvier,et al.  Illuminating the complexity of GPCR pathway selectivity - advances in biosensor development. , 2021, Current opinion in structural biology.

[5]  G. Collins,et al.  Impact of Morphine Dependence and Withdrawal on the Reinforcing Effectiveness of Fentanyl, Cocaine, and Methamphetamine in Rats , 2021, Frontiers in Pharmacology.

[6]  M. Banks,et al.  A drug-vs-food “choice” self-administration procedure in rats to investigate pharmacological and environmental mechanisms of substance use disorders , 2021, Journal of Neuroscience Methods.

[7]  Boshi Huang,et al.  Verifying the role of 3-hydroxy of 17-cyclopropylmethyl-4,5α-epoxy-3,14β-dihydroxy-6β-[(4'-pyridyl) carboxamido]morphinan derivatives via their binding affinity and selectivity profiles on opioid receptors. , 2021, Bioorganic chemistry.

[8]  R. Uprety,et al.  Kratom Alkaloids, Natural and Semi-Synthetic, Show Less Physical Dependence and Ameliorate Opioid Withdrawal , 2021, Cellular and Molecular Neurobiology.

[9]  K. Tan,et al.  Probing biased activation of mu-opioid receptor by the biased agonist PZM21 using all atom molecular dynamics simulation. , 2021, Life sciences.

[10]  J. Miller,et al.  The mixed kappa and delta opioid receptor agonist, MP1104, attenuates chemotherapy-induced neuropathic pain , 2020, Neuropharmacology.

[11]  C. Hartrick,et al.  Dual-Acting Peripherally Restricted Delta/Kappa Opioid (CAV1001) Produces Antinociception in Animal Models of Sub-Acute and Chronic Pain , 2020, Journal of pain research.

[12]  Yan Zhang,et al.  Stereoselective syntheses of 3-dehydroxynaltrexamines and N-methyl-3-dehydroxynaltrexamines. , 2020, Tetrahedron letters.

[13]  F. Hsu,et al.  Antinociceptive, reinforcing, and pruritic effects of the G-protein signalling-biased mu opioid receptor agonist PZM21 in non-human primates. , 2020, British journal of anaesthesia.

[14]  J. V. Aldrich,et al.  Multifunctional opioid receptor agonism and antagonism by a novel macrocyclic tetrapeptide prevents reinstatement of morphine‐seeking behaviour , 2020, British journal of pharmacology.

[15]  S. Foster,et al.  Biased agonism of clinically approved μ-opioid receptor agonists and TRV130 is not controlled by binding and signaling kinetics , 2020, Neuropharmacology.

[16]  S. Schulz,et al.  Morphine‐induced respiratory depression is independent of β‐arrestin2 signalling , 2020, British journal of pharmacology.

[17]  Bing Yu,et al.  Recent Advances in The Rational Drug Design Based on Multi-target Ligands. , 2020, Current medicinal chemistry.

[18]  Joshua S. Beckmann,et al.  Changes in fentanyl demand following naltrexone, morphine, and buprenorphine in male rats. , 2019, Drug and alcohol dependence.

[19]  B. Avery,et al.  Investigation of the Adrenergic and Opioid Binding affinities, Metabolic Stability, Plasma Protein Binding Properties and Functional Effects of Selected Indole-based Kratom Alkaloids. , 2019, Journal of medicinal chemistry.

[20]  W. Dewey,et al.  Fentanyl depression of respiration: comparison with heroin and morphine , 2019, bioRxiv.

[21]  K. Berg,et al.  Signaling characteristics and functional regulation of delta opioid-kappa opioid receptor (DOP-KOP) heteromers in peripheral sensory neurons , 2019, Neuropharmacology.

[22]  Karl T. Schmidt,et al.  Role of RGS12 in the differential regulation of kappa opioid receptor-dependent signaling and behavior , 2019, Neuropsychopharmacology.

[23]  R. V. van Rijn,et al.  A Review of the Therapeutic Potential of Recently Developed G Protein-Biased Kappa Agonists , 2019, Front. Pharmacol..

[24]  S. B. Caine,et al.  Sex differences in opioid reinforcement under a fentanyl vs. food choice procedure in rats , 2019, Neuropsychopharmacology.

[25]  S. Schulz,et al.  Targeting multiple opioid receptors – improved analgesics with reduced side effects? , 2018, British journal of pharmacology.

[26]  Ryan T. Strachan,et al.  Structure of the Nanobody-Stabilized Active State of the Kappa Opioid Receptor , 2018, Cell.

[27]  N. Ozaki,et al.  Pharmacokinetics of Morphine in Rats with Adjuvant-induced Arthritis. , 2017, In vivo.

[28]  Shelley R. Edwards,et al.  Effects of nalfurafine on the reinforcing, thermal antinociceptive, and respiratory-depressant effects of oxycodone: modeling an abuse-deterrent opioid analgesic in rats , 2017, Psychopharmacology.

[29]  Dong-Sheng Cao,et al.  ADME Properties Evaluation in Drug Discovery: Prediction of Caco-2 Cell Permeability Using a Combination of NSGA-II and Boosting , 2016, J. Chem. Inf. Model..

[30]  D. Kell,et al.  Distributed under Creative Commons Cc-by 4.0 the Apparent Permeabilities of Caco-2 Cells to Marketed Drugs: Magnitude, and Independence from Both Biophysical Properties and Endogenite Similarities , 2022 .

[31]  G. Pasternak,et al.  Synthesis and characterization of a dual kappa-delta opioid receptor agonist analgesic blocking cocaine reward behavior. , 2015, ACS chemical neuroscience.

[32]  N. Imamachi,et al.  Effects of Intrathecal &kgr;-Opioid Receptor Agonist on Morphine-Induced Itch and Antinociception in Mice , 2015, Regional Anesthesia & Pain Medicine.

[33]  G. Mikus,et al.  Morphine-6-glucuronide is responsible for the analgesic effect after morphine administration: a quantitative review of morphine, morphine-6-glucuronide, and morphine-3-glucuronide. , 2014, British journal of anaesthesia.

[34]  S. Hirono,et al.  Design, synthesis, and structure-activity relationship of novel opioid κ receptor selective agonists: α-iminoamide derivatives with an azabicyclo[2.2.2]octene skeleton. , 2014, Bioorganic & medicinal chemistry letters.

[35]  A. Kourounakis,et al.  Multi-target drug design approaches for multifactorial diseases: from neurodegenerative to cardiovascular applications. , 2014, Current medicinal chemistry.

[36]  K. Schiene,et al.  Cebranopadol: A Novel Potent Analgesic Nociceptin/Orphanin FQ Peptide and Opioid Receptor Agonist , 2014, The Journal of Pharmacology and Experimental Therapeutics.

[37]  B. Kieffer,et al.  Knockout subtraction autoradiography: a novel ex vivo method to detect heteromers finds sparse KOP receptor/DOP receptor heterodimerization in the brain. , 2014, European journal of pharmacology.

[38]  J. Deruiter,et al.  Synthesis and antinociceptive properties of N-phenyl-N-(1-(2-(thiophen-2-yl)ethyl)azepane-4-yl)propionamide in the mouse tail-flick and hot-plate tests. , 2014, Bioorganic & medicinal chemistry letters.

[39]  James P. Cain,et al.  Novel fentanyl-based dual μ/δ-opioid agonists for the treatment of acute and chronic pain. , 2013, Life sciences.

[40]  G. Pasternak,et al.  Novel 6β-acylaminomorphinans with analgesic activity. , 2013, European journal of medicinal chemistry.

[41]  Suzanne Skolnik,et al.  Permeability diagnosis model in drug discovery: a diagnostic tool to identify the most influencing properties for gastrointestinal permeability. , 2013, Current topics in medicinal chemistry.

[42]  L. Benet,et al.  Drug Discovery and Regulatory Considerations for Improving In Silico and In Vitro Predictions that Use Caco-2 as a Surrogate for Human Intestinal Permeability Measurements , 2013, The AAPS Journal.

[43]  S. Hirono,et al.  Essential structure of opioid κ receptor agonist nalfurafine for binding to the κ receptor 3: synthesis of decahydro(iminoethano)phenanthrene derivatives with an oxygen functionality at the 3-position and their pharmacologies. , 2012, Bioorganic & medicinal chemistry letters.

[44]  H. Nagase,et al.  Essential structure of opioid κ receptor agonist nalfurafine for binding to the κ receptor 2: synthesis of decahydro(iminoethano)phenanthrene derivatives and their pharmacologies. , 2012, Bioorganic & medicinal chemistry letters.

[45]  N. Yamaotsu,et al.  Essential structure of opioid κ receptor agonist nalfurafine for binding to κ receptor 1: synthesis of decahydroisoquinoline derivatives and their pharmacologies. , 2012, Chemical & pharmaceutical bulletin.

[46]  G. Loudos,et al.  Molecular nanomedicine towards cancer: ¹¹¹In-labeled nanoparticles. , 2012, Journal of pharmaceutical sciences.

[47]  S. Hirono,et al.  Synthesis of new opioid derivatives with a propellane skeleton and their pharmacology. Part 2: Propellane derivatives with an amide side chain. , 2012, Bioorganic & medicinal chemistry letters.

[48]  Donna A Volpe,et al.  Drug-permeability and transporter assays in Caco-2 and MDCK cell lines. , 2011, Future medicinal chemistry.

[49]  V. Hruby,et al.  Development of potent μ and δ opioid agonists with high lipophilicity. , 2011, Journal of medicinal chemistry.

[50]  Z. Pan,et al.  Synaptic Mechanism for Functional Synergism between δ- and μ-Opioid Receptors , 2010, The Journal of Neuroscience.

[51]  M. Waldhoer,et al.  Opioid-receptor-heteromer-specific trafficking and pharmacology. , 2010, Current opinion in pharmacology.

[52]  X. Pu,et al.  Chronic morphine administration induces over-expression of aldolase C with reduction of CREB phosphorylation in the mouse hippocampus. , 2009, European journal of pharmacology.

[53]  Yuichi Sugiyama,et al.  Prediction of Hepatic Clearance in Human From In Vitro Data for Successful Drug Development , 2009, The AAPS Journal.

[54]  Richard Morphy,et al.  Designing multiple ligands - medicinal chemistry strategies and challenges. , 2009, Current pharmaceutical design.

[55]  S. Husbands,et al.  Effects of Atypical κ-Opioid Receptor Agonists on Intrathecal Morphine-Induced Itch and Analgesia in Primates , 2009, Journal of Pharmacology and Experimental Therapeutics.

[56]  C. Bertucci,et al.  Species-dependent stereoselective drug binding to albumin: a circular dichroism study. , 2008, Chirality.

[57]  Xiao Chen,et al.  Expression changes of hippocampal energy metabolism enzymes contribute to behavioural abnormalities during chronic morphine treatment , 2007, Cell Research.

[58]  Lawrence X. Yu,et al.  Classification of Drug Permeability with a Caco-2 Cell Monolayer Assay , 2007 .

[59]  G. Trainor,et al.  The importance of plasma protein binding in drug discovery , 2007, Expert opinion on drug discovery.

[60]  W. Dewey,et al.  PKC and PKA inhibitors reinstate morphine-induced behaviors in morphine tolerant mice. , 2006, Pharmacological research.

[61]  L. Jia,et al.  LC-MS/MS assay and dog pharmacokinetics of the dimeric pyrrolobenzodiazepine SJG-136 (NSC 694501). , 2006, Journal of chromatography. B, Analytical technologies in the biomedical and life sciences.

[62]  David J. Daniels,et al.  A Bivalent Ligand (KDN-21) Reveals Spinal δ and κ Opioid Receptors Are Organized as Heterodimers That Give Rise to δ1 and κ2 Phenotypes. Selective Targeting of δ−κ Heterodimers , 2004 .

[63]  K. Rice,et al.  Opioid Interactions in Rhesus Monkeys: Effects of δ + μ and δ + κ Agonists on Schedule-Controlled Responding and Thermal Nociception , 2003, Journal of Pharmacology and Experimental Therapeutics.

[64]  N. Lee,et al.  Opioid receptor interactions: local and nonlocal, symmetric and asymmetric, physical and functional. , 2003, Life sciences.

[65]  B. Fermini,et al.  The impact of drug-induced QT interval prolongation on drug discovery and development , 2003, Nature Reviews Drug Discovery.

[66]  Michael J Banker,et al.  Development and validation of a 96-well equilibrium dialysis apparatus for measuring plasma protein binding. , 2003, Journal of pharmaceutical sciences.

[67]  P. Portoghese,et al.  Identity of the putative δ1-opioid receptor as a δ–κ heteromer in the mouse spinal cord , 2003 .

[68]  M. Narita,et al.  Blockade of mu-opioid receptor-mediated G-protein activation and antinociception by TRK-820 in mice. , 2003, European journal of pharmacology.

[69]  N. Mello,et al.  Mixed kappa agonists and mu agonists/antagonists as potential pharmacotherapeutics for cocaine abuse: synthesis and opioid receptor binding affinity of N-substituted derivatives of morphinan. , 2001, Bioorganic & medicinal chemistry letters.

[70]  R. Hurley,et al.  A cellular mechanism for the antinociceptive effect of a kappa opioid receptor agonist , 2001, Pain.

[71]  J. Kamei,et al.  TRK-820, a selective kappa-opioid agonist, produces potent antinociception in cynomolgus monkeys. , 2001, Japanese journal of pharmacology.

[72]  M. Narita,et al.  The novel kappa-opioid receptor agonist TRK-820 has no affect on the development of antinociceptive tolerance to morphine in mice. , 2000, European journal of pharmacology.

[73]  J. Kamei,et al.  Characterization of the antinociceptive effects of TRK-820 in the rat. , 2000, European journal of pharmacology.

[74]  M. Narita,et al.  Potent antinociceptive effects of TRK-820, a novel κ-opioid receptor agonist , 1999 .

[75]  E. Weber,et al.  ACEA-1328, an NMDA receptor antagonist, increases the potency of morphine and U50,488H in the tail flick test in mice. , 1998, Pharmacological research.

[76]  D. Sawyer,et al.  Kappa Antinociceptive Activity of Spiradoline in the Cold-Water Tail-Flick Assay in Rats , 1998, Pharmacology Biochemistry and Behavior.

[77]  J. Levine,et al.  κ- and δ-opioid agonists synergize to produce potent analgesia , 1990, Brain Research.

[78]  J. W. Gates,et al.  The Replacement of Phenolic Hydroxyl Groups by Hydrogen , 1966 .

[79]  F. Carroll,et al.  Dissociable effects of the kappa opioid receptor agonist nalfurafine on pain/itch-stimulated and pain/itch-depressed behaviors in male rats , 2017, Psychopharmacology.

[80]  M. Kainoh,et al.  Nalfurafine hydrochloride, a selective κ opioid receptor agonist, has no reinforcing effect on intravenous self-administration in rhesus monkeys. , 2016, Journal of pharmacological sciences.

[81]  G. Koob,et al.  Compulsive-Like Responding for Opioid Analgesics in Rats with Extended Access , 2015, Neuropsychopharmacology.