1
|
Rivas-Santisteban R, Lillo J, Garrigós C, Navarro G, Franco R. GPR88 impairs the signaling of kappa opioid receptors in a heterologous system and in primary striatal neurons. Neuropharmacology 2025; 265:110242. [PMID: 39613254 DOI: 10.1016/j.neuropharm.2024.110242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/14/2024] [Accepted: 11/26/2024] [Indexed: 12/01/2024]
Abstract
The physiological role of GPR88, an orphan G protein-coupled receptor (GPCR) predominantly expressed in the striatum, remains unclear, despite its altered expression in parkinsonian animal models. GPR88 is known to interact with other GPCRs. Specifically, GPR88 expression inhibits signaling mediated by the μ-opioid receptor in cells coexpressing both receptors. The effect of GPR88 on the kappa-opioid receptor (KOR) is less understood. In this study, we examine the interaction between GPR88 and KOR, and the impact of GPR88 expression on KOR-mediated signaling in heterologous cells and primary striatal neurons. Bioluminescence resonance energy transfer and proximity ligation assays revealed an interaction between GPR88 and KOR. Functional assays showed that GPR88 antagonized the effects of U69,593, a selective KOR agonist, on forskolin-stimulated cAMP levels, β-arrestin-2 recruitment, and phosphorylation of extracellular signal-regulated kinases (ERK1/2) in HEK-293T cells coexpressing both receptors. In primary striatal neurons, GPR88 and KOR complexes were observed, with KOR activation effects enhanced when GPR88 expression was suppressed using RNA interference. These results suggest that GPR88 and KOR are coexpressed in striatal neurons, where GPR88 inhibits KOR activation. Notably, the GPR88-KOR heteromer was more prevalent in dopamine D1-receptor-containing neurons of the direct pathway of the basal ganglia. Given the roles of KORs in dopamine release, motor function regulation, and pain and reward perception, the GPR88-KOR interaction warrants further investigation in the context of neuropathic pain, Parkinson's disease, and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Rafael Rivas-Santisteban
- Laboratory of Computational Medicine, Biostatistics Unit, Faculty of Medicine, Autonomous University of Barcelona, Campus Bellaterra, 08193, Barcelona, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases. CiberNed., Spanish National Health Institute Carlos iii, Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| | - Jaume Lillo
- Network Center for Biomedical Research in Neurodegenerative Diseases. CiberNed., Spanish National Health Institute Carlos iii, Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain; Molecular Neurobiology Laboratory, Dept. Biochemistry and Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Claudia Garrigós
- Molecular Neurobiology Laboratory, Dept. Biochemistry and Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Gemma Navarro
- Network Center for Biomedical Research in Neurodegenerative Diseases. CiberNed., Spanish National Health Institute Carlos iii, Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain; Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Rafael Franco
- Network Center for Biomedical Research in Neurodegenerative Diseases. CiberNed., Spanish National Health Institute Carlos iii, Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain; Molecular Neurobiology Laboratory, Dept. Biochemistry and Molecular Biomedicine, Facultat de Biologia, Universitat de Barcelona, 08028, Barcelona, Spain; School of Chemistry, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
2
|
Gao Y, Zhu H, Lv L, Xu X, Li W, Fu W. Discovery of N'-benzyl-3-chloro-N-((1S,3R,4R)-3-((dimethylamino)methyl)-4-hydroxy-4-(3-methoxyphenyl)cyclohexyl)benzenesulfonamide as a novel selective KOR ligand. Eur J Med Chem 2024; 276:116643. [PMID: 38986343 DOI: 10.1016/j.ejmech.2024.116643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 06/29/2024] [Accepted: 06/29/2024] [Indexed: 07/12/2024]
Abstract
The effective management of moderate to severe pain often relies on the use of analgesic agents. However, the widespread utility of these medications is hindered by the occurrence of several undesirable side effects. In light of this challenge, there is growing interest in the development of κ opioid receptor (KOR) agonists, which have shown promise in mitigating these adverse effects. In this study, leveraging the structural scaffold of compound D (our previous study), we embarked on the design, synthesis, and evaluation of a series of N'-benzyl-3-chloro-N- ((1S,3R,4R)-3-((dimethylamino)methyl)-4-hydroxy-4-(3-methoxyphenyl)cyclohexyl)benzenesulfonamide derivatives. These compounds were subjected to comprehensive in vitro and in vivo test. Through systematic structure-activity relationship (SAR) exploration, we successfully identified compound 23p (Ki(KOR):1.9 nM) as a highly selective KOR ligand of new chemotype. 23p showed high clearance in vitro PK test, and abdominal contraction test showed potent antinociceptive effect. 23p and its O-demethyl metabolite 25 were both found in the plasma of mouse, 25 also showed potent affinity toward KOR (Ki(KOR): 3.1 nM), both they contribute to the analgesic effect. Moreover, 23p exhibited potent antinociceptive activity in abdominal constriction test, which was effectively abolished by pre-treatment of nor-BNI, a selective KOR antagonist.
Collapse
Affiliation(s)
- Yang Gao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Haoran Zhu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Lunan Lv
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Xiaodi Xu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China.
| |
Collapse
|
3
|
Kansagra KA, Momin T, Patel HB, Shah C, Parmar G, Ghoghari A, Patel HV, Parmar DV. Safety, tolerability, pharmacokinetics, and pharmacodynamics of a novel kappa opioid receptor agonist ZYKR1: a randomized double-blind placebo-control phase 1 study in healthy adult human participants. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4737-4745. [PMID: 38141084 DOI: 10.1007/s00210-023-02912-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
To perform first-in-human single-dose escalation trial of ZYKR1, which is a potent, selective, and peripherally-restricted kappa opioid receptor agonist, is the purpose of this study. This randomized, double-blind, placebo-controlled single ascending dose study conducted at Zydus Research Centre, Ahmedabad, India included healthy male participants aged 18-55 years and weighing > 50 kg. The primary objective was to evaluate the safety and tolerability of ZYKR1. The secondary objective was to evaluate the pharmacokinetics and pharmacodynamics (PD) of ZYKR1. Participants received ZYKR1 (0.5 - 6 mcg/kg) or placebo infused intravenously in 15 ± 1 min. Of total five dose groups (0.5 - 6 mcg/kg), each group included eight participants with six and two randomized to ZYKR1 and placebo, respectively. Three participants experienced six treatment-emergent adverse events (TEAEs); two were gastrointestinal disorders (nausea and vomiting at 2 mcg/kg); and four were related to the nervous system (headache (at 2 mcg/kg) and facial tingling, facial numbness and paresthesia (at 6 mcg/kg)); all TEAEs were mild and resolved without sequelae. The Cmax of ZYKR1 was achieved after 15 - 20 min of start of infusion. The mean exposures (Cmax and AUC0 - t) increased in a dose-proportional manner. The mean t1/2 ranged from 2.20 to 2.98 h across the dose range. Increase in the mean prolactin level was significantly higher in treatment groups compared with that in the placebo group. Intravenous ZYKR1 at doses up to 6 mcg/kg showed acceptable safety and tolerability and demonstrated a short half-life with principal route of excretion as renal. ZYKR1 displayed a potent PD effect reflected by increased prolactin levels, supporting further study in patients. Trial registration Clinical Trial Registry of India: CTRI/2018/07/014927. Date of registration: 18/07/2018.
Collapse
Affiliation(s)
- Kevinkumar A Kansagra
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India.
| | | | - Hardik B Patel
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India
| | - Chintan Shah
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India
| | - Gordhan Parmar
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India
| | - Ashok Ghoghari
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India
| | - Harilal V Patel
- Zydus Research Centre, Clinical R & D, Zydus Lifesciences Limited, Sarkhej-Bavla N. H. No. 8 A, Moraiya, Ahmedabad, 382213, Gujarat, India
| | | |
Collapse
|
4
|
St. Onge C, Pagare PP, Zheng Y, Arriaga M, Stevens DL, Mendez RE, Poklis JL, Halquist MS, Selley DE, Dewey WL, Banks ML, Zhang Y. Systematic Structure-Activity Relationship Study of Nalfurafine Analogues toward Development of Potentially Nonaddictive Pain Management Treatments. J Med Chem 2024; 67:9552-9574. [PMID: 38814086 PMCID: PMC11181328 DOI: 10.1021/acs.jmedchem.4c00646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/08/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
Despite the availability of numerous pain medications, the current array of Food and Drug Administration-approved options falls short in adequately addressing pain states for numerous patients and consequently worsens the opioid crisis. Thus, it is imperative for basic research to develop novel and nonaddictive pain medications. Toward addressing this clinical goal, nalfurafine (NLF) was chosen as a lead and its structure-activity relationship (SAR) systematically studied through design, syntheses, and in vivo characterization of 24 analogues. Two analogues, 21 and 23, showed longer durations of action than NLF in a warm-water tail immersion assay, produced in vivo effects primarily mediated by KOR and DOR, penetrated the blood-brain barrier, and did not function as reinforcers. Additionally, 21 produced fewer sedative effects than NLF. Taken together, these results aid the understanding of NLF SAR and provide insights for future endeavors in developing novel nonaddictive therapeutics to treat pain.
Collapse
Affiliation(s)
- Celsey
M. St. Onge
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, 800 E. Leigh
Street, Richmond, Virginia 23219, United States
| | - Piyusha P. Pagare
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, 800 E. Leigh
Street, Richmond, Virginia 23219, United States
| | - Yi Zheng
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, 800 E. Leigh
Street, Richmond, Virginia 23219, United States
| | - Michelle Arriaga
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - David L. Stevens
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Rolando E. Mendez
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Justin L. Poklis
- Department
of Pharmaceutics, Virginia Commonwealth
University, 410 North
12th Street, Richmond, Virginia 23298, United States
| | - Matthew S. Halquist
- Department
of Pharmaceutics, Virginia Commonwealth
University, 410 North
12th Street, Richmond, Virginia 23298, United States
| | - Dana E. Selley
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - William L. Dewey
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Matthew L. Banks
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
| | - Yan Zhang
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, 800 E. Leigh
Street, Richmond, Virginia 23219, United States
- Department
of Pharmacology and Toxicology, Virginia
Commonwealth University, 410 North 12th Street, Richmond, Virginia 23298, United States
- Institute
for Drug and Alcohol Studies, 203 East Cary Street, Richmond, Virginia 23298, United States
| |
Collapse
|
5
|
De Neve J, Elhabazi K, Gonzalez S, Herby C, Schneider S, Utard V, Fellmann-Clauss R, Petit-Demouliere N, Lecat S, Kremer M, Ces A, Daubeuf F, Martin C, Ballet S, Bihel F, Simonin F. Multitarget μ-Opioid Receptor Agonists─Neuropeptide FF Receptor Antagonists Induce Potent Antinociception with Reduced Adverse Side Effects. J Med Chem 2024. [PMID: 38687204 DOI: 10.1021/acs.jmedchem.4c00442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The design of bifunctional compounds is a promising approach toward the development of strong analgesics with reduced side effects. We here report the optimization of the previously published lead peptide KGFF09, which contains opioid receptor agonist and neuropeptide FF receptor antagonist pharmacophores and is shown to induce potent antinociception and reduced side effects. We evaluated the novel hybrid peptides for their in vitro activity at MOP, NPFFR1, and NPFFR2 and selected four of them (DP08/14/32/50) for assessment of their acute antinociceptive activity in mice. We further selected DP32 and DP50 and observed that their antinociceptive activity is mostly peripherally mediated; they produced no respiratory depression, no hyperalgesia, significantly less tolerance, and strongly attenuated withdrawal syndrome, as compared to morphine and the recently FDA-approved TRV130. Overall, these data suggest that MOP agonist/NPFF receptor antagonist hybrids might represent an interesting strategy to develop novel analgesics with reduced side effects.
Collapse
Affiliation(s)
- Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Khadija Elhabazi
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Simon Gonzalez
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Claire Herby
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Séverine Schneider
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Valérie Utard
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Rosine Fellmann-Clauss
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Nathalie Petit-Demouliere
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Sandra Lecat
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Mélanie Kremer
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (INCI), 67000 Strasbourg, France
| | - Aurelia Ces
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (INCI), 67000 Strasbourg, France
| | - François Daubeuf
- Plateforme de Chimie Biologique Intégrative de Strasbourg, UAR 3286, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Charlotte Martin
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Frédéric Bihel
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| |
Collapse
|
6
|
Huskinson SL, Platt DM, Smith ZR, Doyle WS, Zamarripa CA, Dunaway K, Prisinzano TE, Freeman KB. Quantification of observable behaviors following oral administration of oxycodone and nalfurafine in male rhesus monkeys. Drug Alcohol Depend 2023; 252:110953. [PMID: 37734282 PMCID: PMC10615792 DOI: 10.1016/j.drugalcdep.2023.110953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/08/2023] [Accepted: 08/28/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND Recent preclinical studies have investigated the atypical kappa-opioid receptor (KOR) agonist, nalfurafine, as a co-formulary with mu-opioid receptor (MOR) agonists as a potential deterrent for misuse. However, no study has investigated effects of nalfurafine combined with a MOR agonist using an oral route of administration. The objective of the current study was to measure behavioral effects of orally administered oxycodone and nalfurafine, alone and combined, in rhesus monkeys using a quantitative behavioral observation procedure. METHODS Adult male rhesus monkeys (N=5) were orally administered vehicle, oxycodone (0.56-1.8mg/kg), nalfurafine (0.001-0.0056mg/kg), or mixtures (1.0mg/kg oxycodone/0.001-0.0056mg/kg nalfurafine) in a Jell-O vehicle at multiple timepoints (10-320min). Species-typical and drug-induced behaviors were recorded by observers blinded to conditions. RESULTS Oxycodone alone significantly increased scratch and face-rub behaviors without affecting other behaviors. Nalfurafine decreased baseline levels of scratch without affecting other behaviors, and oxycodone-nalfurafine combinations resulted in reduced oxycodone-induced scratching at a dose (0.001mg/kg) that did not produce sedation-like effects. Oxycodone combined with larger nalfurafine doses (0.0032-0.0056mg/kg) also reduced oxycodone induced scratch that were accompanied with sedation-like effects (i.e., increased lip droop). CONCLUSIONS Nalfurafine was orally active in rhesus monkeys, and it reduced oxycodone-induced pruritus at a dose that did not produce sedation-like effects that are commonly observed with prototypical KOR agonists. Combinations of low doses of nalfurafine with MOR agonists such as oxycodone may be well-tolerated by humans who are prescribed MOR agonists for the treatment of pain.
Collapse
Affiliation(s)
- Sally L Huskinson
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA; Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216.
| | - Donna M Platt
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA; Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216
| | - Zachary R Smith
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - William S Doyle
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216
| | - C Austin Zamarripa
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Kristen Dunaway
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Thomas E Prisinzano
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA
| | - Kevin B Freeman
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA; Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216
| |
Collapse
|
7
|
Santino F, Gentilucci L. Design of κ-Opioid Receptor Agonists for the Development of Potential Treatments of Pain with Reduced Side Effects. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28010346. [PMID: 36615540 PMCID: PMC9822356 DOI: 10.3390/molecules28010346] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/13/2022] [Accepted: 12/25/2022] [Indexed: 01/04/2023]
Abstract
The κ-opioid receptor (KOR) has recently emerged as an alternative therapeutic target for the development of pain medications, without deleterious side effects associated with the μ-opioid receptor (MOR). However, modulation of KOR is currently under investigation for the treatment of depression, mood disorders, psychiatric comorbidity, and specific drug addictions. However, KOR agonists also trigger adverse effects including sedation, dysphoria, and hallucinations. In this respect, there is currently much debate on alternative paradigms. Recent effort has been devoted in search of biased ligands capable of selectively activating favorable signaling over signaling associated with unwanted side effects. On the other hand, the use of partial agonists is expected to allow the analgesia to be produced at dosages lower than those required to produce the adverse effects. More empirically, the unwanted central effects can be also avoided by using peripherally restricted agonists. In this review, we discuss the more recent trends in the design of KOR-selective, biased or partial, and finally, peripherally acting agonists. Special emphasis is given on the discussion of the most recent approaches for controlling functional selectivity of KOR-specific ligands.
Collapse
|
8
|
Khan MIH, Sawyer BJ, Akins NS, Le HV. A systematic review on the kappa opioid receptor and its ligands: New directions for the treatment of pain, anxiety, depression, and drug abuse. Eur J Med Chem 2022; 243:114785. [PMID: 36179400 DOI: 10.1016/j.ejmech.2022.114785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022]
Abstract
Kappa opioid receptor (KOR) is a member of the opioid receptor system, the G protein-coupled receptors that are expressed throughout the peripheral and central nervous systems and play crucial roles in the modulation of antinociception and a variety of behavioral states like anxiety, depression, and drug abuse. KOR agonists are known to produce potent analgesic effects and have been used clinically for the treatment of pain, while KOR antagonists have shown efficacy in the treatment of anxiety and depression. This review summarizes the history, design strategy, discovery, and development of KOR ligands. KOR agonists are classified as non-biased, G protein-biased, and β-arrestin recruitment-biased, according to their degrees of bias. The mechanisms and associated effects of the G protein signaling pathway and β-arrestin recruitment signaling pathway are also discussed. Meanwhile, KOR antagonists are classified as long-acting and short-acting, based on their half-lives. In addition, we have special sections for mixed KOR agonists and selective peripheral KOR agonists. The mechanisms of action and pharmacokinetic, pharmacodynamic, and behavioral studies for each of these categories are also discussed in this review.
Collapse
Affiliation(s)
- Md Imdadul H Khan
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Benjamin J Sawyer
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Nicholas S Akins
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Hoang V Le
- Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, University, MS, 38677, USA.
| |
Collapse
|
9
|
Lee YS. Peptidomimetics and Their Applications for Opioid Peptide Drug Discovery. Biomolecules 2022; 12:biom12091241. [PMID: 36139079 PMCID: PMC9496382 DOI: 10.3390/biom12091241] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Despite various advantages, opioid peptides have been limited in their therapeutic uses due to the main drawbacks in metabolic stability, blood-brain barrier permeability, and bioavailability. Therefore, extensive studies have focused on overcoming the problems and optimizing the therapeutic potential. Currently, numerous peptide-based drugs are being marketed thanks to new synthetic strategies for optimizing metabolism and alternative routes of administration. This tutorial review briefly introduces the history and role of natural opioid peptides and highlights the key findings on their structure-activity relationships for the opioid receptors. It discusses details on opioid peptidomimetics applied to develop therapeutic candidates for the treatment of pain from the pharmacological and structural points of view. The main focus is the current status of various mimetic tools and the successful applications summarized in tables and figures.
Collapse
Affiliation(s)
- Yeon Sun Lee
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
10
|
Anapindi KDB, Romanova EV, Checco JW, Sweedler JV. Mass Spectrometry Approaches Empowering Neuropeptide Discovery and Therapeutics. Pharmacol Rev 2022; 74:662-679. [PMID: 35710134 DOI: 10.1124/pharmrev.121.000423] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The discovery of insulin in the early 1900s ushered in the era of research related to peptides acting as hormones and neuromodulators, among other regulatory roles. These essential gene products are found in all organisms, from the most primitive to the most evolved, and carry important biologic information that coordinates complex physiology and behavior; their misregulation has been implicated in a variety of diseases. The evolutionary origins of at least 30 neuropeptide signaling systems have been traced to the common ancestor of protostomes and deuterostomes. With the use of relevant animal models and modern technologies, we can gain mechanistic insight into orthologous and paralogous endogenous peptides and translate that knowledge into medically relevant insights and new treatments. Groundbreaking advances in medicine and basic science influence how signaling peptides are defined today. The precise mechanistic pathways for over 100 endogenous peptides in mammals are now known and have laid the foundation for multiple drug development pipelines. Peptide biologics have become valuable drugs due to their unique specificity and biologic activity, lack of toxic metabolites, and minimal undesirable interactions. This review outlines modern technologies that enable neuropeptide discovery and characterization, and highlights lessons from nature made possible by neuropeptide research in relevant animal models that is being adopted by the pharmaceutical industry. We conclude with a brief overview of approaches/strategies for effective development of peptides as drugs. SIGNIFICANCE STATEMENT: Neuropeptides, an important class of cell-cell signaling molecules, are involved in maintaining a range of physiological functions. Since the discovery of insulin's activity, over 100 bioactive peptides and peptide analogs have been used as therapeutics. Because these are complex molecules not easily predicted from a genome and their activity can change with subtle chemical modifications, mass spectrometry (MS) has significantly empowered peptide discovery and characterization. This review highlights contributions of MS-based research towards the development of therapeutic peptides.
Collapse
Affiliation(s)
- Krishna D B Anapindi
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois (K.D.B.A., E.V.R., J.V.S.) and Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska (J.W.C.)
| | - Elena V Romanova
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois (K.D.B.A., E.V.R., J.V.S.) and Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska (J.W.C.)
| | - James W Checco
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois (K.D.B.A., E.V.R., J.V.S.) and Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska (J.W.C.)
| | - Jonathan V Sweedler
- Department of Chemistry and the Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois (K.D.B.A., E.V.R., J.V.S.) and Department of Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska (J.W.C.)
| |
Collapse
|
11
|
Brice-Tutt AC, Eans SO, Yakovlev D, Aldrich JV, McLaughlin JP. An analog of [d-Trp]CJ-15,208 exhibits kappa opioid receptor antagonism following oral administration and prevents stress-induced reinstatement of extinguished morphine conditioned place preference. Pharmacol Biochem Behav 2022; 217:173405. [PMID: 35584724 DOI: 10.1016/j.pbb.2022.173405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 03/26/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022]
Abstract
Opioid use disorder (OUD) relapse rates are discouragingly high, underscoring the need for new treatment options. The macrocyclic tetrapeptide natural product CJ-15,208 and its stereoisomer [d-Trp]CJ-15,208 demonstrate kappa opioid receptor (KOR) antagonist activity upon oral administration which prevents stress-induced reinstatement of cocaine-seeking behavior. In order to further explore the structure-activity relationships and expand the potential therapeutic applications of KOR antagonism for the treatment of OUD, we screened a series of 24 analogs of [d-Trp]CJ-15,208 with the goal of enhancing KOR antagonist activity. From this screening, analog 22 arose as a compound of interest, demonstrating dose-dependent KOR antagonism after central and oral administration lasting at least 2.5 h. In further oral testing, analog 22 lacked respiratory, locomotor, or reinforcing effects, consistent with the absence of opioid agonism. Pretreatment with analog 22 (30 mg/kg, p.o.) prevented stress-induced reinstatement of extinguished morphine conditioned place preference and reduced some signs of naloxone-precipitated withdrawal in mice physically dependent on morphine. Collectively, these data support the therapeutic potential of KOR antagonists to support abstinence in OUD and ameliorate opioid withdrawal.
Collapse
Affiliation(s)
- Ariana C Brice-Tutt
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Shainnel O Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Dmitry Yakovlev
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Jane V Aldrich
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, United States of America.
| |
Collapse
|
12
|
Effect of Preventive Analgesia with Nalbuphine and Dexmedetomidine in Endoscopic Sinus Surgery. Pain Res Manag 2022; 2022:2344733. [PMID: 35685676 PMCID: PMC9173995 DOI: 10.1155/2022/2344733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/22/2022] [Indexed: 11/29/2022]
Abstract
Background The study was to assess the efficacy and safety of nalbuphine combined with dexmedetomidine for preventive analgesia in endoscopic sinus surgery. Methods 110 patients with deviation of the nasal septum were randomized into the nalbuphine group (group N), dexmedetomidine combined with nalbuphine group (group DN), and saline group (group C). Fifteen minutes before the induction of anesthesia, patients in group N were injected nalbuphine 0.2 mg/kg intravenously; patients in group DN received intravenous infusion of dexmedetomidine 0.5 μg/kg and injection of nalbuphine 0.2 mg/kg; patients in group C received 0.9% saline. Mean arterial pressure (MAP), heart rate (HR), numerical rating scale (NRS) scores, quality of recovery-40 (QoR-40) scores, the need for remedial analgesia, the consumption of remifentanil and propofol, and the incidence of adverse reactions were recorded. Results MAP, HR, and NRS scores of the DN group were significantly lower and the QoR-40 scores were higher than those of groups N and C (P < 0.001). The need for remedial analgesia, the consumption of remifentanil and propofol, and the incidence of nausea in the DN group were the lowest among the three groups (P < 0.001). Conclusion Preventive analgesia with nalbuphine and dexmedetomidine in endoscopic sinus surgery can not only maintain hemodynamic stability but also reduce intraoperative anesthetic dosage, postoperative pain, and improve the quality of postoperative recovery without affecting the revival and extubation time.
Collapse
|
13
|
Huskinson SL, Platt DM, Zamarripa CA, Dunaway K, Brasfield M, Prisinzano TE, Blough BE, Freeman KB. The G-protein biased kappa opioid agonists, triazole 1.1 and nalfurafine, produce non-uniform behavioral effects in male rhesus monkeys. Pharmacol Biochem Behav 2022; 217:173394. [DOI: 10.1016/j.pbb.2022.173394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/07/2022] [Accepted: 04/28/2022] [Indexed: 11/26/2022]
|
14
|
Aldrich JV, McLaughlin JP. Peptide Kappa Opioid Receptor Ligands and Their Potential for Drug Development. Handb Exp Pharmacol 2022; 271:197-220. [PMID: 34463847 DOI: 10.1007/164_2021_519] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ligands for kappa opioid receptors (KOR) have potential uses as non-addictive analgesics and for the treatment of pruritus, mood disorders, and substance abuse. These areas continue to have major unmet medical needs. Significant advances have been made in recent years in the preclinical development of novel opioid peptides, notably ones with structural features that inherently impart stability to proteases. Following a brief discussion of the potential therapeutic applications of KOR agonists and antagonists, this review focuses on two series of novel opioid peptides, all-D-amino acid tetrapeptides as peripherally selective KOR agonists for the treatment of pain and pruritus without centrally mediated side effects, and macrocyclic tetrapeptides based on CJ-15,208 that can exhibit different opioid profiles with potential applications such as analgesics and treatments for substance abuse.
Collapse
Affiliation(s)
- Jane V Aldrich
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
15
|
McCarthy EA, Dischino D, Maguire C, Leon S, Talbi R, Cheung E, Schteingart CD, Rivière PJM, Reed SD, Steiner RA, Navarro VM. Inhibiting Kiss1 Neurons With Kappa Opioid Receptor Agonists to Treat Polycystic Ovary Syndrome and Vasomotor Symptoms. J Clin Endocrinol Metab 2022; 107:e328-e347. [PMID: 34387319 PMCID: PMC8684497 DOI: 10.1210/clinem/dgab602] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT Recent evidence suggests that vasomotor symptoms (VMS) or hot flashes in the postmenopausal reproductive state and polycystic ovary syndrome (PCOS) in the premenopausal reproductive state emanate from the hyperactivity of Kiss1 neurons in the hypothalamic infundibular/arcuate nucleus (KNDy neurons). OBJECTIVE We demonstrate in 2 murine models simulating menopause and PCOS that a peripherally restricted kappa receptor agonist (PRKA) inhibits hyperactive KNDy neurons (accessible from outside the blood-brain barrier) and impedes their downstream effects. DESIGN Case/control. SETTING Academic medical center. PARTICIPANTS Mice. INTERVENTIONS Administration of peripherally restricted kappa receptor agonists and frequent blood sampling to determine hormone release and body temperature. MAIN OUTCOME MEASURES LH pulse parameters and body temperature. RESULTS First, chronic administration of a PRKA to bilaterally ovariectomized mice with experimentally induced hyperactivity of KNDy neurons reduces the animals' elevated body temperature, mean plasma LH level, and mean peak LH per pulse. Second, chronic administration of a PRKA to a murine model of PCOS, having elevated plasma testosterone levels and irregular ovarian cycles, suppresses circulating levels of LH and testosterone and restores normal ovarian cyclicity. CONCLUSION The inhibition of kisspeptin neuronal activity by activation of kappa receptors shows promise as a novel therapeutic approach to treat both VMS and PCOS in humans.
Collapse
Affiliation(s)
- Elizabeth A McCarthy
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Daniel Dischino
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Caroline Maguire
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Silvia Leon
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Rajae Talbi
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Eugene Cheung
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | | | | | - Susan D Reed
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Robert A Steiner
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98195, USA
- Department of Physiology & Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Victor M Navarro
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Harvard Program in Neuroscience, Boston, MA 02115, USA
- Correspondence: Victor M. Navarro PhD, Brigham and Women’s Hospital, Division of Endocrinology, Diabetes and Hypertension, 221 Longwood Ave, Boston, MA 02115, USA.
| |
Collapse
|
16
|
Ghoghari A, Bhatt C, Patel K, Jha A, Patel H, Jain M, Momin T, Parmar D, Patel P. Estimation of ZYKR1 in human urine and plasma utilizing LC-MS/MS positive electrospray ionization; a kappa opioid receptor (KOR) agonist. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1185:122982. [PMID: 34731743 DOI: 10.1016/j.jchromb.2021.122982] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 09/30/2021] [Accepted: 10/02/2021] [Indexed: 10/20/2022]
Abstract
ZYKR1, a short chain novel peptide with selective kappa opioid receptor agonist activity used as analgesics for the treatment of pain management. A sensitive and selective LC-MS/MS assay was developed and validated for estimation of ZYKR1 in human urine and plasma. ZY17258, an analogue compound was used as an internal standard. ZYKR1 was quantified using a selective reaction monitoring in electrospray ionization positive mode. The chromatographic separation was performed using mobile phase consisted of 0.05% v/v formic acid in water and methanol in gradient elution by analytical column Kinetex C8, 100 A°, 5 µm, 100 × 4.6 mm with 8.0 min analytical run time. Solid Phase extraction technique was used for purification of ZYKR1 and IS from human urine and plasma. The calibration curves were linear over range of 0.300 ng/mL to 300 ng/mL and 0.500 ng/mL to 500 ng/mL for human urine and plasma, respectively. No matrix effect and no significant carryover were observed. The extraction recovery was consistent and ranged from about 85% to 93% in human urine and in plasma respectively. Inter-day and intra-day accuracy (bias, %) and precision (CV, %) was -11.11 to 5.91 % and -2.25 to 6.65 % in human urine and -2.74 to 7.17 % and 2.24 to 15.18 % in plasma respectively were well within the acceptance criteria. Both the assays were devoid of endogenous matrix interference and commonly used concomitant drug interference. The validated assays were used for estimation of ZYKR1 from clinical pharmacokinetic study sample bioanalysis in healthy human subjects.
Collapse
Affiliation(s)
- Ashok Ghoghari
- Bioanalytical Laboratory, Zydus Research Centre, Ahmedabad 382 210, India.
| | - Chandrakant Bhatt
- Bioanalytical Laboratory, Zydus Research Centre, Ahmedabad 382 210, India
| | - Kuldip Patel
- Bioanalytical Laboratory, Zydus Research Centre, Ahmedabad 382 210, India
| | - Anil Jha
- Bioanalytical Laboratory, Zydus Research Centre, Ahmedabad 382 210, India
| | - Harilal Patel
- Bioanalytical Laboratory, Zydus Research Centre, Ahmedabad 382 210, India
| | - Mukul Jain
- Bioanalytical Laboratory, Zydus Research Centre, Ahmedabad 382 210, India
| | - Taufik Momin
- Clinical Research, Zydus Research Centre, Ahmedabad 382 210, India
| | - Deven Parmar
- Clinical Research, Zydus Research Centre, Ahmedabad 382 210, India
| | | |
Collapse
|
17
|
Shram MJ, Spencer RH, Qian J, Munera CL, Lewis ME, Henningfield JE, Webster L, Menzaghi F. Evaluation of the abuse potential of difelikefalin, a selective kappa-opioid receptor agonist, in recreational polydrug users. Clin Transl Sci 2021; 15:535-547. [PMID: 34708917 PMCID: PMC8841457 DOI: 10.1111/cts.13173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/10/2021] [Accepted: 09/09/2021] [Indexed: 01/23/2023] Open
Abstract
Difelikefalin, a selective kappa‐opioid receptor agonist with limited central nervous system penetration, is being developed for the treatment of chronic pruritic conditions. This randomized, double‐blind, active‐ and placebo‐controlled, four‐way crossover study was designed to evaluate the abuse potential of difelikefalin in healthy recreational polydrug users. Using a 4 × 4 Williams design, nondependent adult users of opioids and hallucinogens (N = 44) were randomized to receive single intravenous (i.v.) injections of difelikefalin at supratherapeutic doses (5 and 15 mcg/kg); pentazocine (0.5 mg/kg), a schedule IV mu‐opioid partial agonist and kappa‐opioid receptor agonist; and placebo. The abuse potential of difelikefalin was compared with pentazocine and placebo using the maximal score (maximum effect [Emax]) of the Drug Liking visual analog scale (VAS; primary end point), along with multiple secondary end points of subject‐rated measures and pupillometry. Difelikefalin produced significantly lower Drug Liking VAS Emax, and lower peak positive, sedative, and perceptual effects compared with pentazocine. These effects of difelikefalin were small, brief, and not dose‐dependent, although marginally greater than those observed with placebo. Neither dose of difelikefalin elicited significant negative or hallucinogenic effects. On end‐of‐session measures of overall drug liking and willingness to take the drug again, difelikefalin did not differ from placebo, indicating subjects neither liked nor disliked the effects overall and did not feel motivated to take the drug again. Consistent with its lack of mu agonist activity, difelikefalin did not induce miosis compared with pentazocine. All treatments were generally well‐tolerated. This study indicates that difelikefalin presents a low potential for abuse.
Collapse
Affiliation(s)
- Megan J Shram
- Altreos Research Partners, Inc., Toronto, ON, Canada
| | | | - Jenny Qian
- Cara Therapeutics, Inc., Stamford, Connecticut, USA
| | | | - Michael E Lewis
- BioDiligence Partners, Inc., Kennett Square, Pennsylvania, USA
| | | | | | | |
Collapse
|
18
|
Zhang M, Xu B, Li N, Zhang R, Zhang Q, Shi X, Xu K, Xiao J, Chen D, Niu J, Shi Y, Fang Q. Development of Multifunctional and Orally Active Cyclic Peptide Agonists of Opioid/Neuropeptide FF Receptors that Produce Potent, Long-Lasting, and Peripherally Restricted Antinociception with Diminished Side Effects. J Med Chem 2021; 64:13394-13409. [PMID: 34465090 DOI: 10.1021/acs.jmedchem.1c00694] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
We previously reported that a multifunctional opioid/neuropeptide FF receptor agonist, DN-9, achieved peripherally restricted analgesia with reduced side effects. To develop stable and orally bioavailable analogues of DN-9, eight lactam-bridged cyclic analogues of DN-9 between positions 2 and 5 were designed, synthesized, and biologically evaluated. In vitro cAMP assays revealed that these analogues, except 7, were multifunctional ligands that activated opioid and neuropeptide FF receptors. Analogue 1 exhibited improved potency for κ-opioid and NPFF2 receptors. All analogues exhibited potent, long-lasting, and peripherally restricted antinociception in the tail-flick test without tolerance development after subcutaneous administration and produced oral analgesia. Oral administration of the optimized compound analogue 1 exhibited powerful, peripherally restricted antinociceptive effects in mouse models of acute, inflammatory, and neuropathic pain. Remarkably, orally administered analogue 1 had no significant side effects, such as tolerance, dependence, constipation, or respiratory depression, at effective analgesic doses.
Collapse
Affiliation(s)
- Mengna Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, Gansu Province 730000, PR China
| | - Biao Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, Gansu Province 730000, PR China
| | - Ning Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, Gansu Province 730000, PR China
| | - Run Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, Gansu Province 730000, PR China
| | - Qinqin Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, Gansu Province 730000, PR China
| | - Xuerui Shi
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, Gansu Province 730000, PR China
| | - Kangtai Xu
- School of Life Sciences, Lanzhou University, 222 Tianshui South Road, Lanzhou, Gansu Province 730000, PR China
| | - Jian Xiao
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, Gansu Province 730000, PR China
| | - Dan Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, Gansu Province 730000, PR China
| | - Jiandong Niu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, Gansu Province 730000, PR China
| | - Yonghang Shi
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, Gansu Province 730000, PR China
| | - Quan Fang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, Gansu Province 730000, PR China
| |
Collapse
|
19
|
Zhao Y, Joshi AA, Aldrich JV, Murray TF. Quantification of kappa opioid receptor ligand potency, efficacy and desensitization using a real-time membrane potential assay. Biomed Pharmacother 2021; 143:112173. [PMID: 34536757 PMCID: PMC8516733 DOI: 10.1016/j.biopha.2021.112173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/02/2021] [Accepted: 09/05/2021] [Indexed: 10/25/2022] Open
Abstract
We explored the utility of the real-time FLIPR Membrane Potential (FMP) assay as a method to assess kappa opioid receptor (KOR)-induced hyperpolarization. The FMP Blue dye was used to measure fluorescent signals reflecting changes in membrane potential in KOR expressing CHO (CHO-KOR) cells. Treatment of CHO-KOR cells with kappa agonists U50,488 or dynorphin [Dyn (1-13)NH2] produced rapid and concentration-dependent decreases in FMP Blue fluorescence reflecting membrane hyperpolarization. Both the nonselective opioid antagonist naloxone and the κ-selective antagonists nor-binaltorphimine (nor-BNI) and zyklophin produced rightward shifts in the U50,488 concentration-response curves, consistent with competitive antagonism of the KOR mediated response. The decrease in fluorescent emission produced by U50,488 was blocked by overnight pertussis toxin pretreatment, indicating the requirement for PTX-sensitive G proteins in the KOR mediated response. We directly compared the potency of U50,488 and Dyn (1-13)NH2 in the FMP and [35S]GTPγS binding assays, and found that both were approximately 10 times more potent in the cellular fluorescence assay. The maximum responses of both U50,488 and Dyn (1-13)NH2 declined following repeated additions, reflecting receptor desensitization. We assessed the efficacy and potency of structurally distinct KOR small molecule and peptide ligands. The FMP assay reliably detected both partial agonists and stereoselectivity. Using KOR-selective peptides with varying efficacies, we found that the FMP assay allowed high throughput quantification of peptide efficacy. These data demonstrate that the FMP assay is a sensitive method for assessing κ-opioid receptor induced hyperpolarization, and represents a useful approach for quantification of potency, efficacy and desensitization of KOR ligands.
Collapse
Affiliation(s)
- Yuanzi Zhao
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, USA
| | - Anand A Joshi
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA.
| | - Jane V Aldrich
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, USA; Department of Medicinal Chemistry, University of Florida, Gainesville, FL, USA
| | - Thomas F Murray
- Department of Pharmacology and Neuroscience, School of Medicine, Creighton University, Omaha, NE, USA
| |
Collapse
|
20
|
Analogs of the κ opioid receptor antagonist arodyn cyclized by ring-closing metathesis retain κ opioid receptor affinity, selectivity and κ opioid receptor antagonism. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02758-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
21
|
Lee YS, Remesic M, Ramos-Colon C, Wu Z, LaVigne J, Molnar G, Tymecka D, Misicka A, Streicher JM, Hruby VJ, Porreca F. Multifunctional Enkephalin Analogs with a New Biological Profile: MOR/DOR Agonism and KOR Antagonism. Biomedicines 2021; 9:biomedicines9060625. [PMID: 34072734 PMCID: PMC8229567 DOI: 10.3390/biomedicines9060625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 11/25/2022] Open
Abstract
In our previous studies, we developed a series of mixed MOR/DOR agonists that are enkephalin-like tetrapeptide analogs with an N-phenyl-N-piperidin-4-ylpropionamide (Ppp) moiety at the C-terminus. Further SAR study on the analogs, initiated by the findings from off-target screening, resulted in the discovery of LYS744 (6, Dmt-DNle-Gly-Phe(p-Cl)-Ppp), a multifunctional ligand with MOR/DOR agonist and KOR antagonist activity (GTPγS assay: IC50 = 52 nM, Imax = 122% cf. IC50 = 59 nM, Imax = 100% for naloxone) with nanomolar range of binding affinity (Ki = 1.3 nM cf. Ki = 2.4 nM for salvinorin A). Based on its unique biological profile, 6 is considered to possess high therapeutic potential for the treatment of chronic pain by modulating pathological KOR activation while retaining analgesic efficacy attributed to its MOR/DOR agonist activity.
Collapse
Affiliation(s)
- Yeon Sun Lee
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA; (J.L.); (G.M.); (J.M.S.); (F.P.)
- Correspondence: ; Tel.: +1-520-626-2820
| | - Michael Remesic
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA; (M.R.); (C.R.-C.); (V.J.H.)
| | - Cyf Ramos-Colon
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA; (M.R.); (C.R.-C.); (V.J.H.)
| | - Zhijun Wu
- ABC Resource, Plainsboro, NJ 08536, USA;
| | - Justin LaVigne
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA; (J.L.); (G.M.); (J.M.S.); (F.P.)
| | - Gabriella Molnar
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA; (J.L.); (G.M.); (J.M.S.); (F.P.)
| | - Dagmara Tymecka
- Faculty of Chemistry, University of Warsaw, Pasteura, PL-02-093 Warsaw, Poland; (D.T.); (A.M.)
| | - Aleksandra Misicka
- Faculty of Chemistry, University of Warsaw, Pasteura, PL-02-093 Warsaw, Poland; (D.T.); (A.M.)
| | - John M. Streicher
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA; (J.L.); (G.M.); (J.M.S.); (F.P.)
| | - Victor J. Hruby
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ 85721, USA; (M.R.); (C.R.-C.); (V.J.H.)
| | - Frank Porreca
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA; (J.L.); (G.M.); (J.M.S.); (F.P.)
| |
Collapse
|
22
|
Ilari D, Maskri S, Schepmann D, Köhler J, Daniliuc CG, Koch O, Wünsch B. Diastereoselective synthesis of conformationally restricted KOR agonists. Org Biomol Chem 2021; 19:4082-4099. [PMID: 33978027 DOI: 10.1039/d1ob00398d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In order to analyze the bioactive conformation of flexible KOR agonists the ethylenediamine KOR pharmacophore was conformationally constrained by incorporation into a bicyclic system. For this purpose, 2-azabicyclo[3.2.1.]octan-7-amines were designed, synthesized and pharmacologically evaluated. The primary amine 14 as first key intermediate was prepared in a six-step synthesis starting with methyl cyclopent-3-enecarboxylate 9. Whereas phenylacetamides failed to provide bicyclic compounds, the intramolecular nucleophilic substitution of the sulfonamide 25 was initiated by deprotonation with NaH affording the bicyclic compound 26 in 72% yield. The three-step introduction of the pharmacophoric pyrrolidine ring started with nucleophilic substitution of exo-configured tosylate 26 with NaN3, which unexpectedly occurred under retention of configuration leading to exo-configured azide 31. The final KOR agonists 35 and 36 with exo-configured amino moieties were obtained by removal of the N-tosyl moiety of 33 and introduction of the second pharmacophoric element by acylation with dihalophenylacetyl chlorides. The KOR affinity of the pyrrolidine 35a is in the high nanomolar range (Ki = 862 nM). The low KOR affinity is explained by a non-appropriate dihedral angle of 137°/141° of the N(pyrrolidine)-C-C-N(acyl) system. As observed for stereoisomers of potent KOR agonists, phenylacetamide 35a and more importantly sulfonamides 33a and 33b show moderate affinity at σ1 receptors (Ki = 109-208 nM).
Collapse
Affiliation(s)
- Denise Ilari
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany. and Graduate School of Chemistry, Westfälische Wilhelms-Universität Münster, Wilhelm-Klemm-Straße 10, D-48149 Münster, Germany
| | - Sarah Maskri
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany. and GRK 2515, Chemical biology of ion channels (Chembion), Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany. and GRK 2515, Chemical biology of ion channels (Chembion), Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| | - Jens Köhler
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany.
| | - Constantin G Daniliuc
- Organisch-Chemisches Institut, Westfälische Wilhelms-Universität Münster, Corrensstraße 40, D-48149 Münster, Germany
| | - Oliver Koch
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany. and GRK 2515, Chemical biology of ion channels (Chembion), Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany. and Graduate School of Chemistry, Westfälische Wilhelms-Universität Münster, Wilhelm-Klemm-Straße 10, D-48149 Münster, Germany and GRK 2515, Chemical biology of ion channels (Chembion), Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| |
Collapse
|
23
|
Antinociceptive Activity of the Skin Secretion of Phyllomedusa rohdei (Amphibia, Anura). Toxins (Basel) 2020; 12:toxins12090589. [PMID: 32933013 PMCID: PMC7551137 DOI: 10.3390/toxins12090589] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/01/2020] [Accepted: 03/03/2020] [Indexed: 11/16/2022] Open
Abstract
Pain is a distressful experience that can have a major impact on an individual’s quality of life. The need for new and better analgesics has been further intensified in light of the current opioid epidemic. Substances obtained from amphibians have been shown to contain bioactive peptides that exert analgesic effects. The genus Phyllomedusa represents an important source of peptides and bioactive components. The aim of this study was to investigate the antinociceptive effects of the skin secretion of Phyllomedusa rohdei in rodent models of pain. The crude skin extract of P. rohdei was tested in different pain models: acetic acid-induced writhing test (mice), formalin test (rats), Von Frey electronic test for hypernociception induced by PGE2 (rats), and hot plate test (mice). Motor-impairing effects were tested using the rota-rod test. The results showed that the skin extract of P. rohdei exerted antinociceptive effects in all pain models tested. Particularly, the highest dose tested of the skin extract decreased acetic acid-induced writhing by 93%, completely blocked formalin-induced nociception both during the acute and inflammatory phases of the test, PGE2-induced hypernociception by 73% and increased latency to paw withdrawal in the hot plate test by 300%. The effects observed in the hot plate test were reversed by pretreatment with selective µ and κ, but not δ, opioid receptor antagonists, indicating a mechanism of action dependent on µ and κ opioid receptors. The results were not influenced by sedative effects. Further studies remain necessary to reveal the specific compounds involved in the antinociceptive effects of P. rohdei skin extract as a new therapeutic tool in pain management.
Collapse
|
24
|
Brice‐Tutt AC, Wilson LL, Eans SO, Stacy HM, Simons CA, Simpson GG, Coleman JS, Ferracane MJ, Aldrich JV, McLaughlin JP. Multifunctional opioid receptor agonism and antagonism by a novel macrocyclic tetrapeptide prevents reinstatement of morphine-seeking behaviour. Br J Pharmacol 2020; 177:4209-4222. [PMID: 32562259 PMCID: PMC7443475 DOI: 10.1111/bph.15165] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 05/16/2020] [Accepted: 06/03/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE The macrocyclic tetrapeptide natural product CJ-15,208 (cyclo[Phe-d-Pro-Phe-Trp]) is a multifunctional μ-opioid receptor and κ-opioid receptor agonist and κ-opioid receptor antagonist that produces antinociception and prevents stress-induced reinstatement of extinguished cocaine-conditioned place preference (CPP). We hypothesized that an analogue of CJ-15,208, cyclo[Pro-Sar-Phe-d-Phe], would demonstrate multifunctional μ-opioid receptor and κ-opioid receptor ligand activity, producing potent antinociception with fewer liabilities than selective μ-opioid receptor agonists, while preventing both drug- and stress-induced reinstatement of morphine-induced CPP. EXPERIMENTAL APPROACH The opioid receptor agonist and antagonist activity of cyclo[Pro-Sar-Phe-d-Phe] was characterized after i.c.v. and i.p. administration to C57BL/6J or transgenic opioid receptor "knockout" mice using the 55°C warm-water tail-withdrawal assay. Liabilities of locomotor coordination, respiration and spontaneous ambulation, and direct rewarding or aversive properties were assessed. Finally, the ability of cyclo[Pro-Sar-Phe-d-Phe] to block morphine- and stress-induced reinstatement of extinguished CPP was determined. KEY RESULTS cyclo[Pro-Sar-Phe-d-Phe] demonstrated dose-dependent, short-lasting antinociception, with an ED50 (and 95% confidence interval) of 0.15 (0.05-0.21) nmol i.c.v. and 1.91 (0.40-3.54) mg·kg-1 i.p., mediated by μ- and κ-opioid receptors. The macrocyclic tetrapeptide also demonstrated potent dose-dependent κ-opioid receptor antagonist-like activity at 2.5, but not at 4.5, h after administration. cyclo[Pro-Sar-Phe-d-Phe] displayed reduced liabiities compared with morphine, attributed to its additional activity at κ-receptors. Pretreatment with cyclo[Pro-Sar-Phe-d-Phe] prevented stress- and drug-induced reinstatement of extinguished morphine-place preference responses in a time-dependent manner. CONCLUSIONS AND IMPLICATIONS These data suggest that cyclo[Pro-Sar-Phe-d-Phe] is a promising lead compound for both the treatment of pain with reduced sideeffects and preventing both drug- and stress-induced relapse in morphine-abstinent subjects.
Collapse
Affiliation(s)
- Ariana C. Brice‐Tutt
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Lisa L. Wilson
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Shainnel O. Eans
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Heather M. Stacy
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Chloe A. Simons
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Grant G. Simpson
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Jeremy S. Coleman
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Michael J. Ferracane
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Jane V. Aldrich
- Department of Medicinal Chemistry, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
25
|
Mercadante S, Romualdi P. The Therapeutic Potential of Novel Kappa Opioid Receptor-based Treatments. Curr Med Chem 2020; 27:2012-2020. [PMID: 30666905 DOI: 10.2174/0929867326666190121142459] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 12/13/2018] [Accepted: 12/29/2018] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Similarly to the μ opioid receptor, kappa opioid receptor (KOR), is present either in the central nervous system or in peripheral tissues. In the last years, several molecules, able to interact with KOR, have been the focus of basic research for their therapeutic potential in the field of chronic pain, as well as in depression, autoimmune disorders and neurological diseases. DISCUSSION The role of KOR system and the consequent clinical effects derived by its activation or inhibition are discussed. Their potential therapeutic utilization in conditions of stress and drug relapse, besides chronic pain, is presented here, including the possible use of KORagonists in drug addiction. Moreover, the potential role of KOR-antagonists, KOR agonistantagonists and peripheral KOR agonists is proposed. CONCLUSION Other than pain, KORs have a role in regulating reward and mood. Due to its location, KORs seem to mediate interactions between psychiatric disorders, addiction and depression. Experimental studies in animal models have identified brain mechanisms that may contribute to clarify specific pathophysiological processes.
Collapse
Affiliation(s)
- Sebastiano Mercadante
- Pain Relief & Palliative/Supportive Care Unit, La Maddalena Cancer Center, Palermo, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
26
|
Huskinson SL, Platt DM, Brasfield M, Follett ME, Prisinzano TE, Blough BE, Freeman KB. Quantification of observable behaviors induced by typical and atypical kappa-opioid receptor agonists in male rhesus monkeys. Psychopharmacology (Berl) 2020; 237:2075-2087. [PMID: 32372348 PMCID: PMC7308209 DOI: 10.1007/s00213-020-05519-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 04/01/2020] [Indexed: 12/20/2022]
Abstract
RATIONALE Kappa-opioid receptor (KOR) agonists are antinociceptive but have side effects that limit their therapeutic utility. New KOR agonists have been developed that are fully efficacious at the KOR but may produce fewer or reduced side effects that are typical of KOR agonists. OBJECTIVES We determined behavioral profiles for typical and atypical KOR agonists purported to differ in intracellular-signaling profiles as well as a mu-opioid receptor (MOR) agonist, oxycodone, using a behavioral scoring system based on Novak et al. (Am J Primatol 28:124-138, 1992, Am J Primatol 46:213-227, 1998) and modified to quantify drug-induced effects (e.g., Duke et al. J Pharmacol Exp Ther 366:145-157, 2018). METHODS Six adult male rhesus monkeys were administered a range of doses of the typical KOR agonists, U50-488H (0.0032-0.1 mg/kg) and salvinorin A (0.00032-0.01 mg/kg); the atypical KOR agonists, nalfurafine (0.0001-0.001 mg/kg) and triazole 1.1 (0.01-0.32 mg/kg); the MOR agonist, oxycodone (0.0032-0.32 mg/kg); and as controls, cocaine (0.032-0.32 mg/kg) and ketamine (0.32-10 mg/kg). For time-course determinations, the largest dose of each KOR agonist or MOR agonist was administered across timepoints (10-320 min). In mixture conditions, oxycodone (0.1 mg/kg) was followed by KOR-agonist administration. RESULTS Typical KOR agonists produced sedative-like and motor-impairing effects. Nalfurafine was similar to typical KOR agonists on most outcomes, and triazole 1.1 produced no effects on its own except for reducing scratch during time-course determinations. In the mixture, all KOR agonists reduced oxycodone-induced scratching, U50-488H and nalfurafine reduced species-typical activity, and U50-488H increased rest/sleep posture. CONCLUSIONS Atypical "biased" KOR agonists produce side-effect profiles that are relatively benign (triazole 1.1) or reduced (nalfurafine) compared to typical KOR agonists.
Collapse
MESH Headings
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
- Analgesics, Opioid/pharmacology
- Animals
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Diterpenes, Clerodane/pharmacology
- Dose-Response Relationship, Drug
- Macaca mulatta
- Male
- Morphinans/pharmacology
- Motor Activity/drug effects
- Motor Activity/physiology
- Oxycodone/pharmacology
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/physiology
- Spiro Compounds/pharmacology
Collapse
Affiliation(s)
- S L Huskinson
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
| | - D M Platt
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - M Brasfield
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - M E Follett
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - T E Prisinzano
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, 40536, USA
| | - B E Blough
- Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - K B Freeman
- Division of Neurobiology and Behavior Research, Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| |
Collapse
|
27
|
Ferracane MJ, Brice-Tutt AC, Coleman JS, Simpson GG, Wilson LL, Eans SO, Stacy HM, Murray TF, McLaughlin JP, Aldrich JV. Design, Synthesis, and Characterization of the Macrocyclic Tetrapeptide cyclo[Pro-Sar-Phe-d-Phe]: A Mixed Opioid Receptor Agonist-Antagonist Following Oral Administration. ACS Chem Neurosci 2020; 11:1324-1336. [PMID: 32251585 DOI: 10.1021/acschemneuro.0c00086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Substance abuse remains a serious public health crisis, affecting millions of people worldwide. Macrocyclic tetrapeptides like CJ-15,208 and [d-Trp]CJ-15,208 demonstrate opioid activity shown to attenuate the rewarding effects of cocaine in conditioned place preference assays in mice, making them promising lead compounds for treating substance abuse. In the present study, we report the rational design, synthesis, conformational analysis, and continued pharmacological evaluation of the novel macrocyclic tetrapeptide cyclo[Pro-Sar-Phe-d-Phe] to further explore this unique molecular scaffold. This peptide was rationally designed based on X-ray and NMR structures of related macrocyclic tetrapeptides. Following synthesis, its solution-phase conformations were determined by NMR and molecular modeling. The peptide adopted multiple conformations in polar solvents, but a single conformation in chloroform that is stabilized by intramolecular hydrogen bonding. The peptide is orally bioavailable, producing antinociception and antagonism of kappa opioid receptor (KOR) stimulation following oral administration in a mouse 55 °C warm-water tail-withdrawal assay. Notably, cyclo[Pro-Sar-Phe-d-Phe] blocked both stress- and drug-induced reinstatement of cocaine and morphine conditioned place preference in mice following oral administration, and displayed a decreased side-effect profile compared to morphine. Thus, cyclo[Pro-Sar-Phe-d-Phe] is a promising lead compound for the treatment of substance abuse.
Collapse
Affiliation(s)
- Michael J. Ferracane
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
- Department of Chemistry, University of Redlands, Redlands, California 92373, United States
| | - Ariana C. Brice-Tutt
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Jeremy S. Coleman
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Grant G. Simpson
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Lisa L. Wilson
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Shainnel O. Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Heather M. Stacy
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Thomas F. Murray
- Department of Pharmacology, School of Medicine, Creighton University, Omaha, Nebraska 68178, United States
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Jane V. Aldrich
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
28
|
Gisemba SA, Aldrich JV. Optimized Ring Closing Metathesis Reaction Conditions To Suppress Desallyl Side Products in the Solid-Phase Synthesis of Cyclic Peptides Involving Tyrosine( O-allyl). J Org Chem 2020; 85:1407-1415. [PMID: 31880448 PMCID: PMC8018726 DOI: 10.1021/acs.joc.9b02345] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We are exploring constraining aromatic residues in the kappa opioid receptor selective antagonist arodyn (Ac[Phe1,2,3,Arg4,d-Ala8]dynorphin A(1-11)-NH2) by ring closing metathesis (RCM) involving tyrosine(O-allyl) (Tyr(All)), but desallyl products limited the yields of the desired cyclic peptide. The model dipeptide Fmoc-Tyr(All)-Tyr(All) was used to explore different reaction conditions, including the use of isomerization suppressants, to minimize formation of the desallyl products and enhance formation of the desired RCM product. Reaction conditions were identified that enhanced the RCM product yield while suppressing desallyl products using both second-generation Grubbs and second-generation Hoveyda-Grubbs catalysts. These optimized reaction conditions were then applied to the cyclization of a tripeptide and an arodyn analog resulting in ≥70% conversion to the desired cyclic peptides. These strategies should be applicable to RCM involving Tyr(All) and similar residues in peptide and peptidomimetic cyclizations performed on solid phase.
Collapse
Affiliation(s)
- Solomon A. Gisemba
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS 66045
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32610
| | - Jane V. Aldrich
- Department of Medicinal Chemistry, The University of Kansas, Lawrence, KS 66045
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32610
| |
Collapse
|
29
|
Ong JJY, Wei DYT, Goadsby PJ. Recent Advances in Pharmacotherapy for Migraine Prevention: From Pathophysiology to New Drugs. Drugs 2019; 78:411-437. [PMID: 29396834 DOI: 10.1007/s40265-018-0865-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Migraine is a common and disabling neurological disorder, with a significant socioeconomic burden. Its pathophysiology involves abnormalities in complex neuronal networks, interacting at different levels of the central and peripheral nervous system, resulting in the constellation of symptoms characteristic of a migraine attack. Management of migraine is individualised and often necessitates the commencement of preventive medication. Recent advancements in the understanding of the neurobiology of migraine have begun to account for some parts of the symptomatology, which has led to the development of novel target-based therapies that may revolutionise how migraine is treated in the future. This review will explore recent advances in the understanding of migraine pathophysiology, and pharmacotherapeutic developments for migraine prevention, with particular emphasis on novel treatments targeted at the calcitonin gene-related peptide (CGRP) pathway.
Collapse
Affiliation(s)
- Jonathan Jia Yuan Ong
- Headache Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,NIHR-Wellcome Trust King's Clinical Research Facility, King's College Hospital, Wellcome Foundation Building, London, SE5 9PJ, UK.,Division of Neurology, Department of Medicine, National University Health System, University Medicine Cluster, Singapore, Singapore
| | - Diana Yi-Ting Wei
- Headache Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.,NIHR-Wellcome Trust King's Clinical Research Facility, King's College Hospital, Wellcome Foundation Building, London, SE5 9PJ, UK
| | - Peter J Goadsby
- Headache Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK. .,NIHR-Wellcome Trust King's Clinical Research Facility, King's College Hospital, Wellcome Foundation Building, London, SE5 9PJ, UK.
| |
Collapse
|
30
|
Spetea M, Rief SB, Haddou TB, Fink M, Kristeva E, Mittendorfer H, Haas S, Hummer N, Follia V, Guerrieri E, Asim MF, Sturm S, Schmidhammer H. Synthesis, Biological, and Structural Explorations of New Zwitterionic Derivatives of 14- O-Methyloxymorphone, as Potent μ/δ Opioid Agonists and Peripherally Selective Antinociceptives. J Med Chem 2019; 62:641-653. [PMID: 30571123 PMCID: PMC6348443 DOI: 10.1021/acs.jmedchem.8b01327] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Herein, the synthesis and pharmacological characterization of an extended library of differently substituted N-methyl-14- O-methylmorphinans with natural and unnatural amino acids and three dipeptides at position 6 that emerged as potent μ/δ opioid receptor (MOR/DOR) agonists with peripheral antinociceptive efficacy is reported. The current study adds significant value to our initial structure-activity relationships on a series of zwitterionic analogues of 1 (14- O-methyloxymorphone) by targeting additional amino acid residues. The new derivatives showed high binding and potent agonism at MOR and DOR in vitro. In vivo, the new 6-amino acid- and 6-dipeptide-substituted derivatives of 1 were highly effective in inducing antinociception in the writhing test in mice after subcutaneous administration, which was antagonized by naloxone methiodide demonstrating activation of peripheral opioid receptors. Such peripheral opioid analgesics may represent alternatives to presently available drugs for a safer pain therapy.
Collapse
MESH Headings
- Analgesics, Opioid/chemical synthesis
- Analgesics, Opioid/metabolism
- Analgesics, Opioid/therapeutic use
- Animals
- Cell Membrane/metabolism
- Dipeptides/chemistry
- Humans
- Male
- Mice
- Morphine/therapeutic use
- Oxymorphone/analogs & derivatives
- Oxymorphone/chemistry
- Oxymorphone/metabolism
- Oxymorphone/therapeutic use
- Pain/chemically induced
- Pain/drug therapy
- Pain/pathology
- Protein Binding
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Structure-Activity Relationship
Collapse
|
31
|
Ferré G, Czaplicki G, Demange P, Milon A. Structure and dynamics of dynorphin peptide and its receptor. VITAMINS AND HORMONES 2019; 111:17-47. [DOI: 10.1016/bs.vh.2019.05.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
32
|
Peckham AM, De La Cruz A, Dufresne RL. Kappa opioid receptor antagonism: Are opioids the answer for treatment resistant depression? Ment Health Clin 2018; 8:175-183. [PMID: 30155392 PMCID: PMC6063454 DOI: 10.9740/mhc.2018.07.175] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Introduction: Past trials of buprenorphine (BUP) in the treatment of major depressive disorder (MDD) have displayed favorable results, although its clinical utility was limited by the risk of abuse or physical dependence. By combining BUP with samidorphan (SAM), the euphoric high is negated by an opposing mechanism, which theoretically reduces addictive-like properties while allowing the antidepressant properties to remain. As such, the objective of this article is to analyze the results of BUP/SAM premarketing clinical trials as adjunctive treatment for treatment-resistant MDD. Methods: A comprehensive PubMed/MEDLINE search was conducted through November 9, 2017, using the following search terms: depression, samidorphan, buprenorphine, ALKS-5461. Additional data were obtained from Clinicaltrials.gov and resources included in the present study. All English-language clinical trials evaluating the combination of BUP/SAM in the treatment of MDD were included. Results: A few premarketing studies have evaluated the efficacy and safety of BUP/SAM combination as adjunctive treatment in patients with treatment-resistant MDD. The FORWARD-1 through FORWARD-5 trials concluded (1) the most effective dosing ratio of BUP/SAM to reduce abuse potential was 1:1; (2) statistically significant changes in scores from baseline on the Montgomery-Asberg Depression Rating Scale were noted for the 2 mg/2 mg dose compared with placebo; and (3) the most commonly reported adverse effects were nausea, dizziness, and fatigue. Discussion: Buprenorphine/samidorphan has shown favorable results for efficacy and tolerability in premarketing studies evaluating its use as adjunctive therapy for treatment-resistant MDD. Its novel mechanism targeting the opioid pathway may serve as a promising antidepressant devoid of abuse potential.
Collapse
Affiliation(s)
- Alyssa M Peckham
- (Corresponding author) Clinical Assistant Professor, Northeastern University, Bouve College of Health Sciences, School of Pharmacy; Clinical Addiction Pharmacist, Massachusetts General Hospital, Boston, Massachusetts,
| | - Austin De La Cruz
- Clinical Assistant Professor, Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas
| | - Robert L Dufresne
- Professor of Pharmacy and INBRE Behavioral Science Coordinator, Department of Pharmacy Practice, University of Rhode Island College of Pharmacy, Kingston, Rhode Island
| |
Collapse
|
33
|
Farmer AD, Holt CB, Downes TJ, Ruggeri E, Del Vecchio S, De Giorgio R. Pathophysiology, diagnosis, and management of opioid-induced constipation. Lancet Gastroenterol Hepatol 2018; 3:203-212. [DOI: 10.1016/s2468-1253(18)30008-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/11/2017] [Accepted: 11/08/2017] [Indexed: 12/12/2022]
|
34
|
Fang WJ, Murray TF, Aldrich JV. Design, synthesis, and opioid activity of arodyn analogs cyclized by ring-closing metathesis involving Tyr(allyl). Bioorg Med Chem 2017; 26:1157-1161. [PMID: 29273415 DOI: 10.1016/j.bmc.2017.11.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 01/31/2023]
Abstract
Kappa (κ) opioid receptor selective antagonists are useful pharmacological tools in studying κ opioid receptors and have potential to be used as therapeutic agents for the treatment of a variety of diseases including mood disorders and drug addiction. Arodyn (Ac[Phe1-3,Arg4,d-Ala8]Dyn A-(1-11)NH2) is a linear acetylated dynorphin A (Dyn A) analog that is a potent and selective κ opioid receptor antagonist (Bennett et al. J Med Chem 2002;45:5617-5619) and prevents stress-induced reinstatement of cocaine-seeking behavior following central administration (Carey et al. Eur J Pharmacol 2007;569:84-89). To restrict its conformational mobility, explore possible bioactive conformations and potentially increase its metabolic stability we synthesized cyclic arodyn analogs on solid phase utilizing a novel ring-closing metathesis (RCM) reaction involving allyl-protected Tyr (Tyr(All)) residues. This approach preserves the aromatic functionality and directly constrains the side chains of one or more of the Phe residues. The novel cyclic arodyn analog 4 cyclized between Tyr(All) residues incorporated in positions 2 and 3 exhibited potent κ opioid receptor antagonism in the [35S]GTPγS assay (KB = 3.2 nM) similar to arodyn. Analog 3 cyclized between Tyr(All) residues in positions 1 and 2 also exhibited nanomolar κ opioid receptor antagonist potency (KB = 27.5 nM) in this assay. These are the first opioid peptides cyclized via RCM involving aromatic residues, and given their promising pharmacological activity represent novel lead peptides for further exploration.
Collapse
Affiliation(s)
- Wei-Jie Fang
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Thomas F Murray
- Department of Pharmacology, Creighton University, Omaha, NE 68178, USA
| | - Jane V Aldrich
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA; Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
35
|
Erli F, Guerrieri E, Ben Haddou T, Lantero A, Mairegger M, Schmidhammer H, Spetea M. Highly Potent and Selective New Diphenethylamines Interacting with the κ-Opioid Receptor: Synthesis, Pharmacology, and Structure-Activity Relationships. J Med Chem 2017; 60:7579-7590. [PMID: 28825813 PMCID: PMC5601360 DOI: 10.1021/acs.jmedchem.7b00981] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Indexed: 12/26/2022]
Abstract
We previously reported on a series of small molecules targeting the κ-opioid (KOP) receptor featuring a diphenethylamine scaffold and showed the promise of these ligands as effective analgesics with reduced liability for adverse effects. This study expands the structure-activity relationships on our original series by presenting several modifications in the lead compounds 1 (HS665) and 2 (HS666). A library of new diphenethylamines was designed, synthesized, and pharmacologically evaluated. In comparison with 1 and 2, the KOP receptor affinity, selectivity, and agonist activity were modulated by introducing bulkier N-substituents, a 2-fluoro substitution, and additional hydroxyl groups at positions 3' and 4'. Several analogues showed subnanomolar affinity and excellent KOP receptor selectivity acting as full or partial agonists, and one as an antagonist. The new diphenethylamines displayed antinociceptive efficacies with increased potencies than U50,488, 1 and 2 in the writhing assay and without inducing motor dysfunction after sc administration in mice.
Collapse
Affiliation(s)
| | | | - Tanila Ben Haddou
- Department of Pharmaceutical
Chemistry, Institute of Pharmacy and Center for Molecular Biosciences
Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Aquilino Lantero
- Department of Pharmaceutical
Chemistry, Institute of Pharmacy and Center for Molecular Biosciences
Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Michael Mairegger
- Department of Pharmaceutical
Chemistry, Institute of Pharmacy and Center for Molecular Biosciences
Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Helmut Schmidhammer
- Department of Pharmaceutical
Chemistry, Institute of Pharmacy and Center for Molecular Biosciences
Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Mariana Spetea
- Department of Pharmaceutical
Chemistry, Institute of Pharmacy and Center for Molecular Biosciences
Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| |
Collapse
|
36
|
Spetea M, Eans SO, Ganno ML, Lantero A, Mairegger M, Toll L, Schmidhammer H, McLaughlin JP. Selective κ receptor partial agonist HS666 produces potent antinociception without inducing aversion after i.c.v. administration in mice. Br J Pharmacol 2017; 174:2444-2456. [PMID: 28494108 PMCID: PMC5513865 DOI: 10.1111/bph.13854] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/09/2017] [Accepted: 05/03/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE The κ receptor has a central role in modulating neurotransmission in central and peripheral neuronal circuits that subserve pain and other behavioural responses. Although κ receptor agonists do not produce euphoria or lead to respiratory suppression, they induce dysphoria and sedation. We hypothesized that brain-penetrant κ receptor ligands possessing biased agonism towards G protein signalling over β-arrestin2 recruitment would produce robust antinociception with fewer associated liabilities. EXPERIMENTAL APPROACH Two new diphenethylamines with high κ receptor selectivity, HS665 and HS666, were assessed following i.c.v. administration in mouse assays of antinociception with the 55°C warm-water tail withdrawal test, locomotor activity in the rotorod and conditioned place preference. The [35 S]-GTPγS binding and β-arrestin2 recruitment in vitro assays were used to characterize biased agonism. KEY RESULTS HS665 (κ receptor agonist) and HS666 (κ receptor partial agonist) demonstrated dose-dependent antinociception after i.c.v. administration mediated by the κ receptor. These highly selective κ receptor ligands displayed varying biased signalling towards G protein coupling in vitro, consistent with a reduced liability profile, reflected by reduced sedation and absence of conditioned place aversion for HS666. CONCLUSIONS AND IMPLICATIONS HS665 and HS666 activate central κ receptors to produce potent antinociception, with HS666 displaying pharmacological characteristics of a κ receptor analgesic with reduced liability for aversive effects correlating with its low efficacy in the β-arrestin2 signalling pathway. Our data provide further understanding of the contribution of central κ receptors in pain suppression, and the prospect of dissociating the antinociceptive effects of HS665 and HS666 from κ receptor-mediated adverse effects.
Collapse
Affiliation(s)
- Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
| | - Shainnel O Eans
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
- Department of PharmacodynamicsUniversity of FloridaGainesvilleFLUSA
| | | | - Aquilino Lantero
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Michael Mairegger
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Lawrence Toll
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
| | - Helmut Schmidhammer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI)University of InnsbruckInnsbruckAustria
| | - Jay P McLaughlin
- Torrey Pines Institute for Molecular StudiesPort St. LucieFLUSA
- Department of PharmacodynamicsUniversity of FloridaGainesvilleFLUSA
| |
Collapse
|
37
|
Xie JY, De Felice M, Kopruszinski CM, Eyde N, LaVigne J, Remeniuk B, Hernandez P, Yue X, Goshima N, Ossipov M, King T, Streicher JM, Navratilova E, Dodick D, Rosen H, Roberts E, Porreca F. Kappa opioid receptor antagonists: A possible new class of therapeutics for migraine prevention. Cephalalgia 2017; 37:780-794. [PMID: 28376659 DOI: 10.1177/0333102417702120] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Stress is the most commonly reported migraine trigger. Dynorphin, an endogenous opioid peptide acting preferentially at kappa opioid receptors (KORs), is a key mediator of stress responses. The aim of this study was to use an injury-free rat model of functional cephalic pain with features of migraine and medication overuse headache (MOH) to test the possible preventive benefit of KOR blockade on stress-induced cephalic pain. Methods Following sumatriptan priming to model MOH, rats were hyper-responsive to environmental stress, demonstrating delayed cephalic and extracephalic allodynia and increased levels of CGRP in the jugular blood, consistent with commonly observed clinical outcomes during migraine. Nor-binaltorphimine (nor-BNI), a long-acting KOR antagonist or CYM51317, a novel short-acting KOR antagonist, were given systemically either during sumatriptan priming or immediately before environmental stress challenge. The effects of KOR blockade in the amygdala on stress-induced allodynia was determined by administration of nor-BNI into the right or left central nucleus of the amygdala (CeA). Results KOR blockade prevented both stress-induced allodynia and increased plasma CGRP. Stress increased dynorphin content and phosphorylated KOR in both the left and right CeA in sumatriptan-primed rats. However, KOR blockade only in the right CeA prevented stress-induced cephalic allodynia as well as extracephalic allodynia, measured in either the right or left hindpaws. U69,593, a KOR agonist, given into the right, but not the left, CeA, produced allodynia selectively in sumatriptan-primed rats. Both stress and U69,593-induced allodynia were prevented by right CeA U0126, a mitogen-activated protein kinase inhibitor, presumably acting downstream of KOR. Conclusions Our data reveal a novel lateralized KOR circuit that mediated stress-induced cutaneous allodynia and increased plasma CGRP in an injury-free model of functional cephalic pain with features of migraine and medication overuse headache. Selective, small molecule, orally available, and reversible KOR antagonists are currently in development and may represent a novel class of preventive therapeutics for migraine.
Collapse
Affiliation(s)
- Jennifer Y Xie
- 1 Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | - Milena De Felice
- 2 School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Caroline M Kopruszinski
- 3 Department of Pharmacology, Biological Sciences Section, Federal University of Parana, Curitiba, Brazil
| | - Nathan Eyde
- 1 Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | - Justin LaVigne
- 1 Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | - Bethany Remeniuk
- 1 Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | - Pablo Hernandez
- 1 Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | - Xu Yue
- 1 Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | - Naomi Goshima
- 1 Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | - Michael Ossipov
- 1 Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | - Tamara King
- 4 Department of Biomedical Sciences, College of Osteopathic Medicine, Center for Excellence in the Neurosciences, University of New England, Biddeford, ME, USA
| | - John M Streicher
- 1 Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | - Edita Navratilova
- 1 Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA
| | | | - Hugh Rosen
- 6 Scripps Research Institute, La Jolla, CA, USA
| | - Ed Roberts
- 6 Scripps Research Institute, La Jolla, CA, USA
| | - Frank Porreca
- 1 Department of Pharmacology, Arizona Health Sciences Center, University of Arizona, Tucson, AZ, USA.,5 Mayo Clinic, Phoenix, AZ USA
| |
Collapse
|
38
|
Grechko OY, Litvinov RA, Spasov AA, Rashchenko AI, Shtareva DM, Anisimova VA, Minkin VI. Study of μ- and δ-Opioid Activities in Agents with Various κ-Receptor Selectivity. Bull Exp Biol Med 2017; 162:632-635. [PMID: 28361412 DOI: 10.1007/s10517-017-3674-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Indexed: 12/18/2022]
Abstract
A putative opioid agonist RU-1205 was ineffective within in vitro model of electrically induced contractions of rat ileum assessing the μ- and δ-opioid receptor pathways, while morphine inhibited these contractions in a dose-dependent and naloxone-reversible manners with EC50=2.6×10-7 M. In vivo experiments revealed no significant effects of RU-1205 on respiration and gastrointestinal tract contractile activity. In contrast, butorphanol decreased respiration rate by 25% (25-100 mg/kg) and slowed down the transit of labeled particles along the small intestine by 77.1% (1 mg/kg) and by 45.5% (10 mg/kg). Morphine-induced inhibition of peristalsis was dose-dependent with maximum effect (by 68.6%) observed in the dose of 10 mg/kg. It was concluded that the effects of RU-1205 are not related to activation μ- and δ-opioid receptors known to mediate the effects of non-selective opioid agonist morphine and agonist-antagonist butorphanol.
Collapse
Affiliation(s)
- O Yu Grechko
- Volgograd State Medical University, Volgograd, Russia
| | - R A Litvinov
- Volgograd State Medical University, Volgograd, Russia.
| | - A A Spasov
- Volgograd State Medical University, Volgograd, Russia.,Volgograd Medical Research Center, Volgograd, Russia
| | | | - D M Shtareva
- Volgograd State Medical University, Volgograd, Russia
| | - V A Anisimova
- Research Institute of Physical and Organic Chemistry, Southern Federal University, Rostov-on-Don, Russia
| | - V I Minkin
- Research Institute of Physical and Organic Chemistry, Southern Federal University, Rostov-on-Don, Russia
| |
Collapse
|
39
|
Taylor GT, Manzella F. Kappa Opioids, Salvinorin A and Major Depressive Disorder. Curr Neuropharmacol 2016; 14:165-76. [PMID: 26903446 PMCID: PMC4825947 DOI: 10.2174/1570159x13666150727220944] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/11/2015] [Accepted: 07/24/2015] [Indexed: 12/13/2022] Open
Abstract
Opioids are traditionally associated with pain, analgesia and drug abuse. It is now clear,
however, that the opioids are central players in mood. The implications for mood disorders, particularly
clinical depression, suggest a paradigm shift from the monoamine neurotransmitters to the opioids either
alone or in interaction with monoamine neurons. We have a special interest in dynorphin, the last of
the major endogenous opioids to be isolated and identified. Dynorphin is derived from the Greek word
for power, dynamis, which hints at the expectation that the neuropeptide held for its discoverers. Yet,
dynorphin and its opioid receptor subtype, kappa, has always taken a backseat to the endogenous b-endorphin and the
exogenous morphine that both bind the mu opioid receptor subtype. That may be changing as the dynorphin/ kappa system
has been shown to have different, often opposite, neurophysiological and behavioral influences. This includes major
depressive disorder (MDD). Here, we have undertaken a review of dynorphin/ kappa neurobiology as related to behaviors,
especially MDD. Highlights include the unique features of dynorphin and kappa receptors and the special relation of a
plant-based agonist of the kappa receptor salvinorin A. In addition to acting as a kappa opioid agonist, we conclude that
salvinorin A has a complex pharmacologic profile, with potential additional mechanisms of action. Its unique neurophysiological
effects make Salvinorina A an ideal candidate for MDD treatment research.
Collapse
Affiliation(s)
| | - Francesca Manzella
- Behavioral Neuroscience/ Psychology Univ. Missouri - St. Louis, One University Blvd, St. Louis, MO 63121 USA.
| |
Collapse
|
40
|
Ramos-Colon CN, Lee YS, Remesic M, Hall SM, LaVigne J, Davis P, Sandweiss AJ, McIntosh MI, Hanson J, Largent-Milnes TM, Vanderah TW, Streicher J, Porreca F, Hruby VJ. Structure-Activity Relationships of [des-Arg 7]Dynorphin A Analogues at the κ Opioid Receptor. J Med Chem 2016; 59:10291-10298. [PMID: 27797517 DOI: 10.1021/acs.jmedchem.6b01411] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Dynorphin A (Dyn A) is an endogenous ligand for the opioid receptors with preference for the κ opioid receptor (KOR), and its structure-activity relationship (SAR) has been extensively studied at the KOR to develop selective potent agonists and antagonists. Numerous SAR studies have revealed that the Arg7 residue is essential for KOR activity. In contrast, our systematic SAR studies on [des-Arg7]Dyn A analogues found that Arg7 is not a key residue and even deletion of the residue does not affect biological activities at the KOR. In addition, it was also found that [des-Arg7]Dyn A(1-9)-NH2 is a minimum pharmacophore and its modification at the N-terminus leads to selective KOR antagonists. A lead ligand, 14, with high affinity and antagonist activity showed improved metabolic stability and could block antinociceptive effects of a KOR selective agonist, FE200665, in vivo, indicating high potential to treat KOR mediated disorders such as stress-induced relapse.
Collapse
Affiliation(s)
- Cyf N Ramos-Colon
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Yeon Sun Lee
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Michael Remesic
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Sara M Hall
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Justin LaVigne
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Peg Davis
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Alexander J Sandweiss
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Mary I McIntosh
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Jessica Hanson
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Tally M Largent-Milnes
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Todd W Vanderah
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - John Streicher
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Frank Porreca
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| | - Victor J Hruby
- Department of Pharmacology and Toxicology, ‡Department of Chemistry and Biochemistry, and §Department of Pharmacology, University of Arizona , Tucson, Arizona 85721, United States
| |
Collapse
|
41
|
Negrete R, García Gutiérrez MS, Manzanares J, Maldonado R. Involvement of the dynorphin/KOR system on the nociceptive, emotional and cognitive manifestations of joint pain in mice. Neuropharmacology 2016; 116:315-327. [PMID: 27567942 DOI: 10.1016/j.neuropharm.2016.08.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/29/2016] [Accepted: 08/23/2016] [Indexed: 12/18/2022]
Abstract
Joint pain is a major clinical problem mainly associated to osteoarthritis, and characterized by articular cartilage degradation resulting in a complex chronic pain state that includes nociceptive, emotional and cognitive manifestations. Memory impairment, depressive- and anxiety-like symptoms have been reported to be associated with chronic pain, leading to a decrease of life quality. In this study, we evaluated the involvement of the endogenous dynorphin/kappa opioid receptor (KOR) system on the nociceptive, emotional, cognitive, neurochemical and epigenetic manifestations of joint pain. The murine model of monosodium iodoacetate (MIA) was used to induce joint pain in knockout mice for KOR (KOR-KO), prodynorphin (PDYN-KO) and their wild-type (WT) littermates. KOR-KO and PDYN-KO mice developed mechanical allodynia after intra-articular injection of MIA. This allodynia was significantly increased in both KOR-KO and PDYN-KO when compared to WT mice. Accordingly, both mutants showed increased microglial activation on the lumbar section of the spinal cord after MIA. The emotional responses were evaluated by measuring anxiety-like behaviour in the elevated plus maze and anhedonia as depressive-like behaviour, and cognitive alterations in the object recognition paradigm. Emotional and cognitive impairments after joint pain were differently modified in KOR-KO and PDYN-KO mice. Alterations of corticotropin-releasing factor (CRF) on the amygdala and hippocampus and down regulation of histone 3 acetylation on the amygdala suggest a possible mechanism to explain these emotional and cognitive manifestations. Our results reveal a specific involvement of the dynorphin/KOR system on joint pain manifestations that are usually associated to osteoarthritis.
Collapse
Affiliation(s)
- Roger Negrete
- Laboratory of Neuropharmacology, Department of Experimental and Health Science, Pompeu Fabra University (CEXS-UPF), Barcelona, Spain
| | - María Salud García Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Alicante, Spain; Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Science, Pompeu Fabra University (CEXS-UPF), Barcelona, Spain.
| |
Collapse
|
42
|
Guerrieri E, Bermudez M, Wolber G, Berzetei-Gurske IP, Schmidhammer H, Spetea M. Structural determinants of diphenethylamines for interaction with the κ opioid receptor: Synthesis, pharmacology and molecular modeling studies. Bioorg Med Chem Lett 2016; 26:4769-4774. [PMID: 27567368 DOI: 10.1016/j.bmcl.2016.08.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 10/21/2022]
Abstract
The κ opioid (KOP) receptor crystal structure in an inactive state offers nowadays a valuable platform for inquiry into receptor function. We describe the synthesis, pharmacological evaluation and docking calculations of KOP receptor ligands from the class of diphenethylamines using an active-like structure of the KOP receptor attained by molecular dynamics simulations. The structure-activity relationships derived from computational studies was in accordance with pharmacological activities of targeted diphenethylamines at the KOP receptor established by competition binding and G protein activation in vitro assays. Our analysis identified that agonist binding results in breaking of the Arg156-Thr273 hydrogen bond, which stabilizes the inactive receptor conformation, and a crucial hydrogen bond with His291 is formed. Compounds with a phenolic 4-hydroxy group do not form the hydrogen bond with His291, an important residue for KOP affinity and agonist activity. The size of the N-substituent hosted by the hydrophobic pocket formed by Val108, Ile316 and Tyr320 considerably influences binding and selectivity, with the n-alkyl size limit being five carbon atoms, while bulky substituents turn KOP agonists in antagonists. Thus, combination of experimental and molecular modeling strategies provides an initial framework for understanding the structural features of diphenethylamines that are essential to promote binding affinity and selectivity for the KOP receptor, and may be involved in transduction of the ligand binding event into molecular changes, ultimately leading to receptor activation.
Collapse
Affiliation(s)
- Elena Guerrieri
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Marcel Bermudez
- Institute of Pharmacy, Freie Universität Berlin, D-14195 Berlin, Germany
| | - Gerhard Wolber
- Institute of Pharmacy, Freie Universität Berlin, D-14195 Berlin, Germany
| | - Ilona P Berzetei-Gurske
- Biosciences Division, SRI International, 333 Ravenswood Avenue, Menlo Park, CA 94025, United States
| | - Helmut Schmidhammer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria.
| |
Collapse
|
43
|
Khaliq T, Williams TD, Senadheera SN, Aldrich JV. Development of a robust, sensitive and selective liquid chromatography-tandem mass spectrometry assay for the quantification of the novel macrocyclic peptide kappa opioid receptor antagonist [D-Trp]CJ-15,208 in plasma and application to an initial pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1028:11-15. [PMID: 27318293 DOI: 10.1016/j.jchromb.2016.05.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 10/21/2022]
Abstract
Selective kappa opioid receptor (KOR) antagonists may have therapeutic potential as treatments for substance abuse and mood disorders. Since [D-Trp]CJ-15,208 (cyclo[Phe-d-Pro-Phe-d-Trp]) is a novel potent KOR antagonist in vivo, it is imperative to evaluate its pharmacokinetic properties to assist the development of analogs as potential therapeutic agents, necessitating the development and validation of a quantitative method for determining its plasma levels. A method for quantifying [D-Trp]CJ-15,208 was developed employing high performance liquid chromatography-tandem mass spectrometry in mouse plasma. Sample preparation was accomplished through a simple one-step protein precipitation method with acetonitrile, and [D-Trp]CJ-15,208 analyzed following HPLC separation on a Hypersil BDS C8 column. Multiple reaction monitoring (MRM), based on the transitions m/z 578.1→217.1 and 245.0, was specific for [D-Trp]CJ-15,208, and MRM based on the transition m/z 566.2→232.9 was specific for the internal standard without interference from endogenous substances in blank mouse plasma. The assay was linear over the concentration range 0.5-500ng/mL with a mean r(2)=0.9987. The mean inter-day accuracy and precision for all calibration standards were 93-118% and 8.9%, respectively. The absolute recoveries were 85±6% and 81±9% for [D-Trp]CJ-15,208 and the internal standard, respectively. The analytical method had excellent sensitivity with a lower limit of quantification of 0.5ng/mL using a sample volume of 20μL. The method was successfully applied to an initial pharmacokinetic study of [D-Trp]CJ-15,208 following intravenous administration to mice.
Collapse
Affiliation(s)
- Tanvir Khaliq
- Department of Medicinal Chemistry, the University of Kansas, Lawrence, KS 66045, USA; Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32610, USA
| | - Todd D Williams
- Mass Spectrometry and Analytical Proteomics Laboratory, the University of Kansas, Lawrence, KS 66045, USA
| | - Sanjeewa N Senadheera
- Department of Medicinal Chemistry, the University of Kansas, Lawrence, KS 66045, USA
| | - Jane V Aldrich
- Department of Medicinal Chemistry, the University of Kansas, Lawrence, KS 66045, USA; Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
44
|
Albert-Vartanian A, Boyd MR, Hall AL, Morgado SJ, Nguyen E, Nguyen VPH, Patel SP, Russo LJ, Shao AJ, Raffa RB. Will peripherally restricted kappa-opioid receptor agonists (pKORAs) relieve pain with less opioid adverse effects and abuse potential? J Clin Pharm Ther 2016; 41:371-82. [DOI: 10.1111/jcpt.12404] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 04/29/2016] [Indexed: 01/27/2023]
Affiliation(s)
| | - M. R. Boyd
- School of Pharmacy; Temple University; Philadelphia PA USA
| | - A. L. Hall
- School of Pharmacy; Temple University; Philadelphia PA USA
| | - S. J. Morgado
- School of Pharmacy; Temple University; Philadelphia PA USA
| | - E. Nguyen
- School of Pharmacy; Temple University; Philadelphia PA USA
| | | | - S. P. Patel
- School of Pharmacy; Temple University; Philadelphia PA USA
| | - L. J. Russo
- School of Pharmacy; Temple University; Philadelphia PA USA
| | - A. J. Shao
- School of Pharmacy; Temple University; Philadelphia PA USA
| | - R. B. Raffa
- School of Pharmacy; Temple University; Philadelphia PA USA
| |
Collapse
|
45
|
Opioid κ Receptors as a Molecular Target for the Creation of a New Generation of Analgesic Drugs. Pharm Chem J 2016. [DOI: 10.1007/s11094-016-1388-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Chartoff EH, Ebner SR, Sparrow A, Potter D, Baker PM, Ragozzino ME, Roitman MF. Relative Timing Between Kappa Opioid Receptor Activation and Cocaine Determines the Impact on Reward and Dopamine Release. Neuropsychopharmacology 2016; 41:989-1002. [PMID: 26239494 PMCID: PMC4748424 DOI: 10.1038/npp.2015.226] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 07/12/2015] [Accepted: 07/14/2015] [Indexed: 12/31/2022]
Abstract
Negative affective states can increase the rewarding value of drugs of abuse and promote drug taking. Chronic cocaine exposure increases levels of the neuropeptide dynorphin, an endogenous ligand at kappa opioid receptors (KOR) that suppresses dopamine release in the nucleus accumbens (NAc) and elicits negative affective states upon drug withdrawal. However, there is evidence that the effects of KOR activation on affective state are biphasic: immediate aversive effects are followed by delayed increases in reward. The impact of KOR-induced affective states on reward-related effects of cocaine over time is not known. We hypothesize that the initial aversive effects of KOR activation increase, whereas the delayed rewarding effects decrease, the net effects of cocaine on reward and dopamine release. We treated rats with cocaine at various times (15 min to 48 h) after administration of the selective KOR agonist salvinorin A (salvA). Using intracranial self-stimulation and fast scan cyclic voltammetry, we found that cocaine-induced increases in brain stimulation reward and evoked dopamine release in the NAc core were potentiated when cocaine was administered within 1 h of salvA, but attenuated when administered 24 h after salvA. Quantitative real-time PCR was used to show that KOR and prodynorphin mRNA levels were decreased in the NAc, whereas tyrosine hydroxylase and dopamine transporter mRNA levels and tissue dopamine content were increased in the ventral tegmental area 24 h post-salvA. These findings raise the possibility that KOR activation-as occurs upon withdrawal from chronic cocaine-modulates vulnerability to cocaine in a time-dependent manner.
Collapse
Affiliation(s)
- Elena H Chartoff
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA,Department of Psychiatry, Harvard Medical School, McLean Hospital, MRC 218, 115 Mill Street, Belmont, MA 02478, USA, Tel: +1 617 855 2022, Fax: +1 617 855 2023, E-mail:
| | - Shayla R Ebner
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, USA
| | - Angela Sparrow
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - David Potter
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, USA
| | - Phillip M Baker
- Graduate Program in Neuroscience, University of Illinois at Chicago, Chicago, IL, USA
| | - Michael E Ragozzino
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, USA
| | - Mitchell F Roitman
- Department of Psychology, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
47
|
Sirohi S, Aldrich JV, Walker BM. Species differences in the effects of the κ-opioid receptor antagonist zyklophin. Alcohol 2016; 51:43-9. [PMID: 26992699 DOI: 10.1016/j.alcohol.2015.11.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/24/2015] [Accepted: 11/24/2015] [Indexed: 01/02/2023]
Abstract
We have shown that dysregulation of the dynorphin/kappa-opioid receptor (DYN/KOR) system contributes to escalated alcohol self-administration in alcohol dependence and that KOR antagonists with extended durations of action selectively reduce escalated alcohol consumption in alcohol-dependent animals. As KOR antagonism has gained widespread attention as a potential therapeutic target to treat alcoholism and multiple neuropsychiatric disorders, we tested the effect of zyklophin (a short-acting KOR antagonist) on escalated alcohol self-administration in rats made alcohol-dependent using intermittent alcohol vapor exposure. Following dependence induction, zyklophin was infused centrally prior to alcohol self-administration sessions and locomotor activity tests during acute withdrawal. Zyklophin did not impact alcohol self-administration or locomotor activity in either exposure condition. To investigate the neurobiological basis of this atypical effect for a KOR antagonist, we utilized a κ-, μ-, and δ-opioid receptor agonist-stimulated GTPyS coupling assay to examine the opioid receptor specificity of zyklophin in the rat brain and mouse brain. In rats, zyklophin did not affect U50488-, DAMGO-, or DADLE-stimulated GTPyS coupling, whereas the prototypical KOR antagonist nor-binaltorphimine (norBNI) attenuated U50488-induced stimulation in the rat brain tissue at concentrations that did not impact μ- and δ-receptor function. To reconcile the discrepancy between the present rat data and published mouse data, comparable GTPyS assays were conducted using mouse brain tissue; zyklophin effects were consistent with KOR antagonism in mice. Moreover, at higher concentrations, zyklophin exhibited agonist properties in rat and mouse brains. These results identify species differences in zyklophin efficacy that, given the rising interest in the development of short-duration KOR antagonists, should provide valuable information for therapeutic development efforts.
Collapse
Affiliation(s)
- Sunil Sirohi
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA, USA; Department of Integrative Physiology & Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Jane V Aldrich
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL, USA
| | - Brendan M Walker
- Laboratory of Alcoholism and Addictions Neuroscience, Department of Psychology, Washington State University, Pullman, WA, USA; Department of Integrative Physiology & Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, USA; Graduate Program in Neuroscience, Washington State University, Pullman, WA, USA; Translational Addiction Research Center, Washington State University, Pullman, WA, USA.
| |
Collapse
|
48
|
Brust A, Croker DE, Colless B, Ragnarsson L, Andersson Å, Jain K, Garcia-Caraballo S, Castro J, Brierley SM, Alewood PF, Lewis RJ. Conopeptide-Derived κ-Opioid Agonists (Conorphins): Potent, Selective, and Metabolic Stable Dynorphin A Mimetics with Antinociceptive Properties. J Med Chem 2016; 59:2381-95. [PMID: 26859603 DOI: 10.1021/acs.jmedchem.5b00911] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Opioid receptor screening of a conopeptide library led to a novel selective κ-opioid agonist peptide (conorphin T). Intensive medicinal chemistry, guided by potency, selectivity, and stability assays generated a pharmacophore model supporting rational design of highly potent and selective κ-opioid receptor (KOR) agonists (conorphins) with exceptional plasma stability. Conorphins are defined by a hydrophobic benzoprolyl moiety, a double arginine sequence, a spacer amino acid followed by a hydrophobic residue and a C-terminal vicinal disulfide moiety. The pharmacophore model was supported by computational docking studies, revealing receptor-ligand interactions similar to KOR agonist dynorphin A (1-8). A conorphin agonist inhibited colonic nociceptors in a mouse tissue model of chronic visceral hypersensitivity, suggesting the potential of KOR agonists for the treatment of chronic abdominal pain. This new conorphine KOR agonist class and pharmacophore model provide opportunities for future rational drug development and probes for exploring the role of the κ-opioid receptor.
Collapse
Affiliation(s)
- Andreas Brust
- Xenome Limited , Brisbane, Queensland 4068, Australia.,Institute for Molecular Biosciences, The University of Queensland , Brisbane, Queensland, 4072, Australia
| | - Daniel E Croker
- Xenome Limited , Brisbane, Queensland 4068, Australia.,Institute for Molecular Biosciences, The University of Queensland , Brisbane, Queensland, 4072, Australia
| | - Barbara Colless
- Xenome Limited , Brisbane, Queensland 4068, Australia.,Institute for Molecular Biosciences, The University of Queensland , Brisbane, Queensland, 4072, Australia
| | - Lotten Ragnarsson
- Xenome Limited , Brisbane, Queensland 4068, Australia.,Institute for Molecular Biosciences, The University of Queensland , Brisbane, Queensland, 4072, Australia
| | - Åsa Andersson
- Xenome Limited , Brisbane, Queensland 4068, Australia.,Institute for Molecular Biosciences, The University of Queensland , Brisbane, Queensland, 4072, Australia
| | - Kapil Jain
- Institute for Molecular Biosciences, The University of Queensland , Brisbane, Queensland, 4072, Australia
| | - Sonia Garcia-Caraballo
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Disease, Discipline of Medicine, The University of Adelaide, South Australian Health and Medical Research Institute, SAHMRI , Adelaide, SA 5000, Australia
| | - Joel Castro
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Disease, Discipline of Medicine, The University of Adelaide, South Australian Health and Medical Research Institute, SAHMRI , Adelaide, SA 5000, Australia
| | - Stuart M Brierley
- Visceral Pain Group, Centre for Nutrition and Gastrointestinal Disease, Discipline of Medicine, The University of Adelaide, South Australian Health and Medical Research Institute, SAHMRI , Adelaide, SA 5000, Australia
| | - Paul F Alewood
- Institute for Molecular Biosciences, The University of Queensland , Brisbane, Queensland, 4072, Australia
| | - Richard J Lewis
- Xenome Limited , Brisbane, Queensland 4068, Australia.,Institute for Molecular Biosciences, The University of Queensland , Brisbane, Queensland, 4072, Australia
| |
Collapse
|
49
|
New approaches to treating pain. Bioorg Med Chem Lett 2016; 26:1103-19. [DOI: 10.1016/j.bmcl.2015.12.103] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/28/2015] [Accepted: 12/29/2015] [Indexed: 12/11/2022]
|
50
|
Abstract
Chronic pain is one of the most ubiquitous diseases in the world, but treatment is difficult with conventional methods, due to undesirable side effects of treatments and unknown mechanisms of pathological pain states. The endogenous peptide, dynorphin A has long been established as a target for the treatment of pain. Interestingly, this unique peptide has both inhibitory (opioid in nature) and excitatory activities (nonopioid) in the CNS. Both of these effects have been found to play a role in pain and much work has been done to develop therapeutics to enhance the inhibitory effects. Here we will review the dynorphin A compounds that have been designed for the modulation of pain and will discuss where the field stands today.
Collapse
|