1
|
Rehrauer KJ, Cunningham CW. IUPHAR Review - Bivalent and bifunctional opioid receptor ligands as novel analgesics. Pharmacol Res 2023; 197:106966. [PMID: 37865129 DOI: 10.1016/j.phrs.2023.106966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Though efficacious in managing chronic, severe pain, opioid analgesics are accompanied by significant adverse effects including constipation, tolerance, dependence, and respiratory depression. The life-threatening risks associated with µ opioid receptor agonist-based analgesics challenges their use in clinic. A rational approach to combatting these adverse effects is to develop agents that incorporate activity at a second pharmacologic target in addition to µ opioid receptor activation. The promise of such bivalent or bifunctional ligands is the development of an analgesic with an improved side effect profile. In this review, we highlight ongoing efforts in the development of bivalent and bifunctional analgesics that combine µ agonism with efficacy at κ and δ opioid receptors, the nociceptin opioid peptide (NOP) receptor, σ receptors, and cannabinoid receptors. Several examples of bifunctional analgesics in preclinical and clinical development are highlighted, as are strategies being employed toward the rational design of novel agents.
Collapse
Affiliation(s)
- Kyle J Rehrauer
- Department of Pharmaceutical and Administrative Sciences, Concordia University Wisconsin School of Pharmacy, 12800 N. Lake Shore Drive, Mequon, WI 53092, USA
| | - Christopher W Cunningham
- Department of Pharmaceutical and Administrative Sciences, Concordia University Wisconsin School of Pharmacy, 12800 N. Lake Shore Drive, Mequon, WI 53092, USA; CUW Center for Structure-Based Drug Discovery and Development, Concordia University Wisconsin School of Pharmacy, 12800 N. Lake Shore Drive, Mequon, WI 53092, USA.
| |
Collapse
|
2
|
Soyer A, Leterrier S, Breuil L, Goislard M, Leroy C, Saba W, Thibault K, Bo GD, Bottlaender M, Caillé F, Goutal S, Tournier N. Validation of a pharmacological imaging challenge using 11C-buprenorphine and 18F-2-fluoro-2-deoxy-D-glucose positron emission tomography to study the effects of buprenorphine to the rat brain. Front Neurosci 2023; 17:1181786. [PMID: 37234261 PMCID: PMC10205997 DOI: 10.3389/fnins.2023.1181786] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/17/2023] [Indexed: 05/27/2023] Open
Abstract
Aim Buprenorphine mainly acts as an agonist of mu-opioid receptors (mu-OR). High dose buprenorphine does not cause respiratory depression and can be safely administered to elicit typical opioid effects and explore pharmacodynamics. Acute buprenorphine, associated with functional and quantitative neuroimaging, may therefore provide a fully translational pharmacological challenge to explore the variability of response to opioids in vivo. We hypothesized that the CNS effects of acute buprenorphine could be monitored through changes in regional brain glucose metabolism, assessed using 18F-FDG microPET in rats. Materials and methods First, level of receptor occupancy associated with a single dose of buprenorphine (0.1 mg/kg, s.c) was investigated through blocking experiments using 11C-buprenorphine PET imaging. Behavioral study using the elevated plus-maze test (EPM) was performed to assess the impact of the selected dose on anxiety and also locomotor activity. Then, brain PET imaging using 18F-FDG was performed 30 min after injection of unlabeled buprenorphine (0.1 mg/kg, s.c) vs. saline. Two different 18F-FDG PET acquisition paradigms were compared: (i) 18F-FDG injected i.v. under anesthesia and (ii) 18F-FDG injected i.p. in awake animals to limit the impact of anesthesia. Results The selected dose of buprenorphine fully blocked the binding of 11C-buprenorphine in brain regions, suggesting complete receptor occupancy. This dose had no significant impact on behavioral tests used, regardless of the anesthetized/awake handling paradigm. In anesthetized rats, injection of unlabeled buprenorphine decreased the brain uptake of 18F-FDG in most brain regions except in the cerebellum which could be used as a normalization region. Buprenorphine treatment significantly decreased the normalized brain uptake of 18F-FDG in the thalamus, striatum and midbrain (p < 0.05), where binding of 11C-buprenorphine was the highest. The awake paradigm did not improve sensitivity and impact of buprenorphine on brain glucose metabolism could not be reliably estimated. Conclusion Buprenorphine (0.1 mg/kg, s.c) combined with 18F-FDG brain PET in isoflurane anesthetized rats provides a simple pharmacological imaging challenge to investigate the CNS effects of full receptor occupancy by this partial mu-OR agonist. Sensitivity of the method was not improved in awake animals. This strategy may be useful to investigate de desensitization of mu-OR associated with opioid tolerance in vivo.
Collapse
Affiliation(s)
- Amélie Soyer
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Sarah Leterrier
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Louise Breuil
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Maud Goislard
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Claire Leroy
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Wadad Saba
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Karine Thibault
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
- Department of Toxicology and Chemical Risks, Armed Forces Biomedical Research Institute, Bretigny sur Orge, France
| | - Gregory Dal Bo
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
- Department of Toxicology and Chemical Risks, Armed Forces Biomedical Research Institute, Bretigny sur Orge, France
| | - Michel Bottlaender
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Fabien Caillé
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Sébastien Goutal
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Nicolas Tournier
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| |
Collapse
|
3
|
Kong L, Shu X, Tang S, Ye R, Sun H, Jiang S, Li Z, Chai J, Fang Y, Lan Y, Yu L, Xie Q, Fu W, Wang Y, Li W, Qiu Z, Liu J, Shao L. SLL-627 Is a Highly Selective and Potent κ Opioid Receptor (KOR) Agonist with an Unexpected Nonreduction in Locomotor Activity. J Med Chem 2022; 65:10377-10392. [PMID: 35900351 DOI: 10.1021/acs.jmedchem.2c00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Undue central nervous system (CNS) side effects including dysphoria and sedation remain to be a challenge for the development of κ opioid receptor (KOR) agonists as effective and safe analgesics. On the basis of our previous work on morphinan-based KOR agonists, a series of 7α-methyl-7β-substituted northebaine derivatives were designed, synthesized, and biologically assayed. Among others, compound 4a (SLL-627) has been identified as a highly selective and potent KOR agonist both in vitro and in vivo, and its molecular basis was also examined and discussed. Besides low liability to conditioned place aversion (CPA) test, treatment of SLL-627 was associated with a nonreduction in locomotor activity, compared to most of the other arylacetamide- or morphinan-based KOR agonists which generally exhibited apparently sedative effects. This unexpected finding provides new insights to dissociate analgesia from sedation for future discovery of innovative KOR agonists.
Collapse
Affiliation(s)
- Linghui Kong
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Xuelian Shu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, No. 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Siyuan Tang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Rongrong Ye
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, No. 100 Haiquan Road, Shanghai 201418, China
| | - Huijiao Sun
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Shuang Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, No. 138, Xianlin Road, Nanjing 210023, China
| | - Zixiang Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Jingrui Chai
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yun Fang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Yinjie Lan
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Linqian Yu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Qiong Xie
- 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
| | - Yujun Wang
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Zhuibai Qiu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Jinggen Liu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China.,State Key Laboratory of Medical Neurobiology, Fudan University, No. 138 Yixueyuan Road, Shanghai 200032, China
| |
Collapse
|
4
|
Coluzzi F, Rullo L, Scerpa MS, Losapio LM, Rocco M, Billeci D, Candeletti S, Romualdi P. Current and Future Therapeutic Options in Pain Management: Multi-mechanistic Opioids Involving Both MOR and NOP Receptor Activation. CNS Drugs 2022; 36:617-632. [PMID: 35616826 PMCID: PMC9166888 DOI: 10.1007/s40263-022-00924-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/18/2022] [Indexed: 12/24/2022]
Abstract
Opioids are widely used in chronic pain management, despite major concerns about their risk of adverse events, particularly abuse, misuse, and respiratory depression from overdose. Multi-mechanistic opioids, such as tapentadol and buprenorphine, have been widely studied as a valid alternative to traditional opioids for their safer profile. Special interest was focused on the role of the nociceptin opioid peptide (NOP) receptor in terms of analgesia and improved tolerability. Nociceptin opioid peptide receptor agonists were shown to reinforce the antinociceptive effect of mu opioid receptor (MOR) agonists and modulate some of their adverse effects. Therefore, multi-mechanistic opioids involving both MOR and NOP receptor activation became a major field of pharmaceutical and clinical investigations. Buprenorphine was re-discovered in a new perspective, as an atypical analgesic and as a substitution therapy for opioid use disorders; and buprenorphine derivatives have been tested in animal models of nociceptive and neuropathic pain. Similarly, cebranopadol, a full MOR/NOP receptor agonist, has been clinically evaluated for its potent analgesic efficacy and better tolerability profile, compared with traditional opioids. This review overviews pharmacological mechanisms of the NOP receptor system, including its role in pain management and in the development of opioid tolerance. Clinical data on buprenorphine suggest its role as a safer alternative to traditional opioids, particularly in patients with non-cancer pain; while data on cebranopadol still require phase III study results to approve its introduction on the market. Other bifunctional MOR/NOP receptor ligands, such as BU08028, BU10038, and AT-121, are currently under pharmacological investigations and could represent promising analgesic agents for the future.
Collapse
Affiliation(s)
- Flaminia Coluzzi
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, Latina, Italy
- Unit Anesthesia, Intensive Care and Pain Medicine, Sant'Andrea University Hospital, Rome, Italy
| | - Laura Rullo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy
| | - Maria Sole Scerpa
- Unit Anesthesia, Intensive Care and Pain Medicine, Sant'Andrea University Hospital, Rome, Italy
| | - Loredana Maria Losapio
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy
| | - Monica Rocco
- Department of Surgical and Medical Science and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy.
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy
| |
Collapse
|
5
|
Marton J, Fekete A, Cumming P, Hosztafi S, Mikecz P, Henriksen G. Diels-Alder Adducts of Morphinan-6,8-Dienes and Their Transformations. Molecules 2022; 27:2863. [PMID: 35566212 PMCID: PMC9102320 DOI: 10.3390/molecules27092863] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023] Open
Abstract
6,14-ethenomorphinans are semisynthetic opiate derivatives containing an ethylene bridge between positions 6 and 14 in ring-C of the morphine skeleton that imparts a rigid molecular structure. These compounds represent an important family of opioid receptor ligands in which the 6,14-etheno bridged structural motif originates from a [4 + 2] cycloaddition of morphinan-6,8-dienes with dienophiles. Certain 6,14-ethenomorphinans having extremely high affinity for opioid receptors are often non-selective for opioid receptor subtypes, but this view is now undergoing some revision. The agonist 20R-etorphine and 20R-dihydroetorphine are several thousand times more potent analgesics than morphine, whereas diprenorphine is a high-affinity non-selective antagonist. The partial agonist buprenorphine is used as an analgesic in the management of post-operative pain or in substitution therapy for opiate addiction, sometimes in combination with the non-selective antagonist naloxone. In the context of the current opioid crisis, we communicated a summary of several decades of work toward generating opioid analgesics with lesser side effects or abuse potential. Our summary placed a focus on Diels-Alder reactions of morphinan-6,8-dienes and subsequent transformations of the cycloadducts. We also summarized the pharmacological aspects of radiolabeled 6,14-ethenomorphinans used in molecular imaging of opioid receptors.
Collapse
Affiliation(s)
- János Marton
- ABX Advanced Biochemical Compounds Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Strasse 10-14, D-01454 Radeberg, Germany
| | - Anikó Fekete
- Department of Medical Imaging, Division of Nuclear Medicine and Translational Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (A.F.); (P.M.)
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Freiburgstraße 18, 3010 Bern, Switzerland;
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Sándor Hosztafi
- Institute of Pharmaceutical Chemistry, Semmelweis Medical University, Högyes Endre utca 9, H-1092 Budapest, Hungary;
| | - Pál Mikecz
- Department of Medical Imaging, Division of Nuclear Medicine and Translational Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (A.F.); (P.M.)
| | - Gjermund Henriksen
- Norwegian Medical Cyclotron Centre Ltd., Sognsvannsveien 20, N-0372 Oslo, Norway
- Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
- Institute of Physics, University of Oslo, Sem Sælands vei 24, N-0371 Oslo, Norway
| |
Collapse
|
6
|
Brailey-Partridge J, Carey JS, Lovell CJ, Taylor GM. Synthesis of 1-Chloronaloxone and 2-Chloronaloxone. Org Process Res Dev 2021. [DOI: 10.1021/acs.oprd.1c00288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - John S. Carey
- Indivior UK Ltd., Henry Boot Way, Priory Park, Hull HU4 7DY, U.K
| | | | - George M. Taylor
- Indivior UK Ltd., Henry Boot Way, Priory Park, Hull HU4 7DY, U.K
| |
Collapse
|
7
|
Bossert JM, Townsend EA, Altidor LKP, Fredriksson I, Shekara A, Husbands S, Sulima A, Rice KC, Banks ML, Shaham Y. Sex differences in the effect of chronic delivery of the buprenorphine analogue BU08028 on heroin relapse and choice in a rat model of opioid maintenance. Br J Pharmacol 2021; 179:227-241. [PMID: 34505281 DOI: 10.1111/bph.15679] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Maintenance treatment with opioid agonists (buprenorphine, methadone) decreases opioid use and relapse. We recently modelled maintenance treatment in rats and found that chronic delivery of buprenorphine or the μ opioid receptor partial agonist TRV130 decreased relapse to oxycodone seeking and taking. Here, we tested the buprenorphine analogue BU08028 on different heroin relapse-related measures and heroin versus food choice. EXPERIMENTAL APPROACH For relapse assessment, we trained male and female rats to self-administer heroin (6 h·day-1 , 14 days) in Context A and then implanted osmotic minipumps containing BU08028 (0, 0.03 or 0.1 mg·kg-1 ·d-1 ). Effects of chronic BU08028 delivery were tested on (1) incubation of heroin-seeking in a non-drug Context B, (2) extinction responding reinforced by heroin-associated discrete cues in Context B, (3) reinstatement of heroin-seeking induced by re-exposure to Context A and (4) re-acquisition of heroin self-administration in Context A. For choice assessment, we tested the effect of chronic BU08028 delivery on heroin versus food choice. KEY RESULTS Chronic BU08028 delivery decreased incubation of heroin seeking. Unexpectedly, BU08028 increased re-acquisition of heroin self-administration selectively in females. Chronic BU08028 had minimal effects on context-induced reinstatement and heroin versus food choice in both sexes. Finally, exploratory post hoc analyses suggest that BU08028 decreased extinction responding selectively in males. CONCLUSIONS AND IMPLICATIONS Chronic BU08028 delivery had both beneficial and detrimental, sex-dependent, effects on different triggers of heroin relapse and minimal effects on heroin choice in both sexes. Results suggest that BU08028 would not be an effective opioid maintenance treatment in humans.
Collapse
Affiliation(s)
| | - E Andrew Townsend
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | | | - Ida Fredriksson
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, Maryland, USA
| | - Aniruddha Shekara
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, Maryland, USA
| | - Stephen Husbands
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Agnieszka Sulima
- Molecular Targets and Medications Discovery Branch, IRP/NIDA/NIH, Baltimore, Maryland, USA.,Chemical Biology Research Branch, IRP/NIAAA/NIH, Rockville, Maryland, USA
| | - Kenner C Rice
- Molecular Targets and Medications Discovery Branch, IRP/NIDA/NIH, Baltimore, Maryland, USA.,Chemical Biology Research Branch, IRP/NIAAA/NIH, Rockville, Maryland, USA
| | - Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Yavin Shaham
- Behavioral Neuroscience Branch, IRP/NIDA/NIH, Baltimore, Maryland, USA
| |
Collapse
|
8
|
He Q, Wei Y, Liu X, Ye R, Kong L, Li Z, Jiang S, Yu L, Chai J, Xie Q, Fu W, Wang Y, Li W, Qiu Z, Liu J, Shao L. Discovery of an M-Substituted N-Cyclopropylmethyl-7α-phenyl-6,14-endoethanotetrahydronorthebaine as a Selective, Potent, and Orally Active κ-Opioid Receptor Agonist with an Improved Central Nervous System Safety Profile. J Med Chem 2021; 64:12414-12433. [PMID: 34387468 DOI: 10.1021/acs.jmedchem.1c01082] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The search for selective kappa opioid receptor (κOR) agonists with an improved safety profile is an area of interest in opioid research. In this work, a series of m-substituted analogs were designed, synthesized, and assayed, resulting in the identification of compound 6c (SLL-1206) as a κOR agonist with single-digit nanomolar activities. The subtype selectivity of compound 6c appeared to be a consequence of an enormous decrease in the affinity for μOR and δOR, rather than a significant increase in the affinity for κOR, which was not the case for SLL-039, another selective and potent κOR agonist identified in our previous work. Besides reduced central nervous system effects, SLL-1206 exhibited substantially improved physicochemical and pharmacokinetic properties compared with SLL-039, with increases of over 20-fold in aqueous solubility and approximately 40-fold in oral bioavailability in rats.
Collapse
Affiliation(s)
- Qian He
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Yuanyuan Wei
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, 555 Zuchongzhi Road, Shanghai 201203, China
- School of Basic Medicine Sciences and Clinical Pharmacy, China Pharmaceutical University, No.639 Longmian Road, Nanjing 210009, China
| | - Xiao Liu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Rongrong Ye
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, China
| | - Linghui Kong
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Zixiang Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Shuang Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, No.138, Xianlin Road, Nanjing 210023, China
| | - Linqian Yu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Jingrui Chai
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Qiong Xie
- 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
| | - Yujun Wang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Zhuibai Qiu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Jinggen Liu
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
- State Key Laboratory of Medical Neurobiology, Fudan University, No. 138 Yixueyuan Road, Shanghai 200032, China
| |
Collapse
|
9
|
Finke AO, Ravaeva MY, Krasnov VI, Cheretaev IV, Chuyan EN, Baev DS, Shults EE. Cross‐Coupling‐Cyclocondensation Reaction Sequence to Access a Library of Ring‐C Bridged Pyrimidino‐tetrahydrothebaines and Pyrimidinotetrahydrooripavines. ChemistrySelect 2021. [DOI: 10.1002/slct.202101790] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Anastasija O. Finke
- Laboratory of Medicinal Chemistry Laboratory of Pharmaceutical reseach Laboratory of magnetic radiospectroscopy Novosibirsk institute of Organic Chemistry Lavrentyev Ave 9 630090 Novosibirsk Russian Federation
| | - Marina Y. Ravaeva
- Biology and chemical department V.I. Vernadsky Crimean Federal University Republic of Crimea Vernadskogo Avenue, 4 Simferopol 295007 Russian Federation
| | - Vyacheslav I. Krasnov
- Laboratory of Medicinal Chemistry Laboratory of Pharmaceutical reseach Laboratory of magnetic radiospectroscopy Novosibirsk institute of Organic Chemistry Lavrentyev Ave 9 630090 Novosibirsk Russian Federation
| | - Igor V. Cheretaev
- Biology and chemical department V.I. Vernadsky Crimean Federal University Republic of Crimea Vernadskogo Avenue, 4 Simferopol 295007 Russian Federation
| | - Elena N. Chuyan
- Biology and chemical department V.I. Vernadsky Crimean Federal University Republic of Crimea Vernadskogo Avenue, 4 Simferopol 295007 Russian Federation
| | - Dmitry S. Baev
- Laboratory of Medicinal Chemistry Laboratory of Pharmaceutical reseach Laboratory of magnetic radiospectroscopy Novosibirsk institute of Organic Chemistry Lavrentyev Ave 9 630090 Novosibirsk Russian Federation
| | - Elvira E. Shults
- Laboratory of Medicinal Chemistry Laboratory of Pharmaceutical reseach Laboratory of magnetic radiospectroscopy Novosibirsk institute of Organic Chemistry Lavrentyev Ave 9 630090 Novosibirsk Russian Federation
| |
Collapse
|
10
|
Kiguchi N, Ding H, Kishioka S, Ko MC. Nociceptin/Orphanin FQ Peptide Receptor-Related Ligands as Novel Analgesics. Curr Top Med Chem 2021; 20:2878-2888. [PMID: 32384033 DOI: 10.2174/1568026620666200508082615] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 12/21/2022]
Abstract
Despite similar distribution patterns and intracellular events observed in the nociceptin/ orphanin FQ peptide (NOP) receptor and other opioid receptors, NOP receptor activation displays unique pharmacological profiles. Several researchers have identified a variety of peptide and nonpeptide ligands to determine the functional roles of NOP receptor activation and observed that NOP receptor- related ligands exhibit pain modality-dependent pain processing. Importantly, NOP receptor activation results in anti-nociception and anti-hypersensitivity at the spinal and supraspinal levels regardless of the experimental settings in non-human primates (NHPs). Given that the NOP receptor agonists synergistically enhance mu-opioid peptide (MOP) receptor agonist-induced anti-nociception, it has been hypothesized that dual NOP and MOP receptor agonists may display promising functional properties as analgesics. Accumulating evidence indicates that the mixed NOP/opioid receptor agonists demonstrate favorable functional profiles. In NHP studies, bifunctional NOP/MOP partial agonists (e.g., AT-121, BU08028, and BU10038) exerted potent anti-nociception via NOP and MOP receptor activation; however, dose-limiting adverse effects associated with the MOP receptor activation, including respiratory depression, itch sensation, physical dependence, and abuse liability, were not observed. Moreover, a mixed NOP/opioid receptor agonist, cebranopadol, presented promising outcomes in clinical trials as a novel analgesic. Collectively, the dual agonistic actions on NOP and MOP receptors, with appropriate binding affinities and efficacies, may be a viable strategy to develop innovative and safe analgesics.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Huiping Ding
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| | - Shiroh Kishioka
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Mei-Chuan Ko
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States
| |
Collapse
|
11
|
Meyer ME, Doshi A, Yasuda D, Zaveri NT. Structure-Based SAR in the Design of Selective or Bifunctional Nociceptin (NOP) Receptor Agonists. AAPS JOURNAL 2021; 23:68. [PMID: 33974173 DOI: 10.1208/s12248-021-00589-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/28/2021] [Indexed: 11/30/2022]
Abstract
The nociceptin opioid receptor (NOP), the fourth member of the opioid receptor family, and its endogenous peptide ligand, nociceptin or orphanin FQ (N/OFQ), play a vital role in several central nervous system pathways regulating pain, reward, feeding, anxiety, motor control and learning/memory. Both selective NOP agonists as well as bifunctional agonists at the NOP and mu opioid receptor (MOP) have potential therapeutic applications in CNS disorders related to these processes. Using Surflex-Dock protocols, we conducted a computational structure-activity study of four scaffold classes of NOP ligands with varying NOP-MOP selectivity. By docking these compounds into the orthosteric binding sites within an active-state NOP homology model, and an active-state MOP crystal structure, the goal of this study was to use a structure-based drug design approach to modulate NOP affinity and NOP vs. MOP selectivity. We first docked four parent compounds (no side chain) to determine their binding interactions within the NOP and MOP binding pockets. Various polar sidechains were added to the heterocyclic A-pharmacophore to modulate NOP ligand affinity. The substitutions mainly contained a 1-2 carbon chain with a polar substituent such as an amine, alcohol, sulfamide, or guanidine. The SAR analysis is focused on the impact of structural changes in the sidechain, such as chain length, hydrogen bonding capability, and basic vs neutral functional groups on binding affinity and selectivity at both NOP and MOP receptors. This study highlights structural modifications that can be leveraged to rationally design both selective NOP and bifunctional NOP-MOP agonists with different ratios of functional efficacy.
Collapse
Affiliation(s)
- Michael E Meyer
- Astraea Therapeutics, LLC, 320 Logue Avenue, Mountain View, California, 94043, USA
| | - Arpit Doshi
- Astraea Therapeutics, LLC, 320 Logue Avenue, Mountain View, California, 94043, USA
| | - Dennis Yasuda
- Astraea Therapeutics, LLC, 320 Logue Avenue, Mountain View, California, 94043, USA
| | - Nurulain T Zaveri
- Astraea Therapeutics, LLC, 320 Logue Avenue, Mountain View, California, 94043, USA.
| |
Collapse
|
12
|
Pharmacokinetic neuroimaging to study the dose-related brain kinetics and target engagement of buprenorphine in vivo. Neuropsychopharmacology 2021; 46:1220-1228. [PMID: 33603137 PMCID: PMC8115308 DOI: 10.1038/s41386-021-00976-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/18/2020] [Accepted: 01/24/2021] [Indexed: 12/31/2022]
Abstract
A wide range of buprenorphine doses are used for either pain management or maintenance therapy in opioid addiction. The complex in vitro profile of buprenorphine, with affinity for µ-, δ-, and κ-opioid receptors (OR), makes it difficult to predict its dose-related neuropharmacology in vivo. In rats, microPET imaging and pretreatment by OR antagonists were performed to assess the binding of radiolabeled buprenorphine (microdose 11C-buprenorphine) to OR subtypes in vivo (n = 4 per condition). The µ-selective antagonist naloxonazine (10 mg/kg) and the non-selective OR antagonist naloxone (1 mg/kg) blocked the binding of 11C-buprenorphine, while pretreatment by the δ-selective (naltrindole, 3 mg/kg) or the κ-selective antagonist (norbinaltorphimine, 10 mg/kg) did not. In four macaques, PET imaging and kinetic modeling enabled description of the regional brain kinetics of 11C-buprenorphine, co-injected with increasing doses of unlabeled buprenorphine. No saturation of the brain penetration of buprenorphine was observed for doses up to 0.11 mg/kg. Regional differences in buprenorphine-associated receptor occupancy were observed. Analgesic doses of buprenorphine (0.003 and 0.006 mg/kg), respectively, occupied 20% and 49% of receptors in the thalamus while saturating the low but significant binding observed in cerebellum and occipital cortex. Occupancy >90% was achieved in most brain regions with plasma concentrations >7 µg/L. PET data obtained after co-injection of an analgesic dose of buprenorphine (0.003 mg/kg) predicted the binding potential of microdose 11C-buprenorphine. This strategy could be further combined with pharmacodynamic exploration or pharmacological MRI to investigate the neuropharmacokinetics and neuroreceptor correlate, at least at µ-OR, of the acute effects of buprenorphine in humans.
Collapse
|
13
|
Translational value of non-human primates in opioid research. Exp Neurol 2021; 338:113602. [PMID: 33453211 DOI: 10.1016/j.expneurol.2021.113602] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/06/2021] [Accepted: 01/10/2021] [Indexed: 01/02/2023]
Abstract
Preclinical opioid research using animal models not only provides mechanistic insights into the modulation of opioid analgesia and its associated side effects, but also validates drug candidates for improved treatment options for opioid use disorder. Non-human primates (NHPs) have served as a surrogate species for humans in opioid research for more than five decades. The translational value of NHP models is supported by the documented species differences between rodents and primates regarding their behavioral and physiological responses to opioid-related ligands and that NHP studies have provided more concordant results with human studies. This review highlights the utilization of NHP models in five aspects of opioid research, i.e., analgesia, abuse liability, respiratory depression, physical dependence, and pruritus. Recent NHP studies have found that (1) mixed mu opioid and nociceptin/orphanin FQ peptide receptor partial agonists appear to be safe, non-addictive analgesics and (2) mu opioid receptor- and mixed opioid receptor subtype-based medications remain the only two classes of drugs that are effective in alleviating opioid-induced adverse effects. Given the recent advances in pharmaceutical sciences and discoveries of novel targets, NHP studies are posed to identify the translational gap and validate therapeutic targets for the treatment of opioid use disorder. Pharmacological studies using NHPs along with multiple outcome measures (e.g., behavior, physiologic function, and neuroimaging) will continue to facilitate the research and development of improved medications to curb the opioid epidemic.
Collapse
|
14
|
Islam A, Rahman MA, Brenner MB, Moore A, Kellmyer A, Buechler HM, DiGiorgio F, Verchio VR, McCracken L, Sumi M, Hartley R, Lizza JR, Moura-Letts G, Fischer BD, Keck TM. Abuse Liability, Anti-Nociceptive, and Discriminative Stimulus Properties of IBNtxA. ACS Pharmacol Transl Sci 2020; 3:907-920. [DOI: 10.1021/acsptsci.0c00066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Bradford D. Fischer
- Cooper Medical School of Rowan University, 401 Broadway, Camden, New Jersey 08103, United States
| | - Thomas M. Keck
- Cooper Medical School of Rowan University, 401 Broadway, Camden, New Jersey 08103, United States
| |
Collapse
|
15
|
Krüll J, Fehler SK, Hofmann L, Nebel N, Maschauer S, Prante O, Gmeiner P, Lanig H, Hübner H, Heinrich MR. Synthesis, Radiosynthesis and Biological Evaluation of Buprenorphine-Derived Phenylazocarboxamides as Novel μ-Opioid Receptor Ligands. ChemMedChem 2020; 15:1175-1186. [PMID: 32378310 PMCID: PMC7383964 DOI: 10.1002/cmdc.202000180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Indexed: 12/12/2022]
Abstract
Targeted structural modifications have led to a novel type of buprenorphine-derived opioid receptor ligand displaying an improved selectivity profile for the μ-OR subtype. On this basis, it is shown that phenylazocarboxamides may serve as useful bioisosteric replacements for the widely occurring cinnamide units, without loss of OR binding affinity or subtype selectivity. This study further includes functional experiments pointing to weak partial agonist properties of the novel μ-OR ligands, as well as docking and metabolism experiments. Finally, the unique bifunctional character of phenylazocarboxylates, herein serving as precursors for the azocarboxamide subunit, was exploited to demonstrate the accessibility of an 18 F-fluorinated analogue.
Collapse
Affiliation(s)
- Jasmin Krüll
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Stefanie K. Fehler
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Laura Hofmann
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Natascha Nebel
- Department of Nuclear MedicineMolecular Imaging and RadiochemistryFriedrich-Alexander-Universität Erlangen-NürnbergSchwabachanlage 1291054ErlangenGermany
| | - Simone Maschauer
- Department of Nuclear MedicineMolecular Imaging and RadiochemistryFriedrich-Alexander-Universität Erlangen-NürnbergSchwabachanlage 1291054ErlangenGermany
| | - Olaf Prante
- Department of Nuclear MedicineMolecular Imaging and RadiochemistryFriedrich-Alexander-Universität Erlangen-NürnbergSchwabachanlage 1291054ErlangenGermany
| | - Peter Gmeiner
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Harald Lanig
- Central Institute for Scientific Computing (ZISC)Friedrich-Alexander-Universität Erlangen-NürnbergMartensstr. 5a91058ErlangenGermany
| | - Harald Hübner
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| | - Markus R. Heinrich
- Department of Chemistry and PharmacyPharmaceutical ChemistryFriedrich-Alexander-Universität Erlangen-NürnbergNikolaus-Fiebiger-Str. 1091058ErlangenGermany
| |
Collapse
|
16
|
Xiao L, Wang Y, Zhang M, Wu W, Kong L, Ma Y, Xu X, Liu X, He Q, Qian Y, Sun H, Wu H, Lin C, Huang H, Ye R, Jiang S, Ye RF, Yuan C, Fang S, Xue D, Yang X, Chen H, Zheng Y, Yu L, Xie Q, Zheng L, Fu W, Li W, Qiu Z, Liu J, Shao L. Discovery of a Highly Selective and Potent κ Opioid Receptor Agonist from N-Cyclopropylmethyl-7α-phenyl-6,14-endoethanotetrahydronorthebaines with Reduced Central Nervous System (CNS) Side Effects Navigated by the Message-Address Concept. J Med Chem 2019; 62:11054-11070. [PMID: 31738550 DOI: 10.1021/acs.jmedchem.9b00857] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Effective and safe analgesics represent an unmet medical need for the treatment of acute and chronic pain. A series of N-cyclopropylmethyl-7α-phenyl-6,14-endoethanotetrahydronorthebaines were designed, synthesized, and assayed, leading to the discovery of a benzylamine derivative (compound 4, SLL-039) as a highly selective and potent κ opioid agonist (κ, Ki = 0.47 nM, κ/μ = 682, κ/δ = 283), which was confirmed by functional assays in vitro and antinociceptive assays in vivo. The in vivo effect could be blocked by pretreatment with the selective κ antagonist nor-BNI. Moreover, this compound did not induce sedation, a common dose limiting effect of κ opioid receptor agonists, at its analgesic dose compared to U50,488H. The dissociation of sedation/antinociception found in SLL-039 was assumed to be correlated with the occupation of its benzamide motif in a unique subsite involving V1182.63, W124EL1, and E209EL2.
Collapse
Affiliation(s)
- Li Xiao
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Yujun Wang
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Mumei Zhang
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Weiwei Wu
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China.,University of Chinese Academy of Sciences , No. 19A Yuquan Road , Beijing 100049 , China
| | - Linghui Kong
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Yan Ma
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China.,Shanghai University School of Life Sciences , No. 99 Shangda Road , Shanghai 200444 , China
| | - Xuejun Xu
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Xiao Liu
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Qian He
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Yuanyuan Qian
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Huijiao Sun
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Haihao Wu
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Cheng Lin
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Huoming Huang
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Rongrong Ye
- Shanghai Institute of Technology , No. 100 Haiquan Road , Shanghai 201418 , China
| | - Shuang Jiang
- Nanjing University of Chinese Medicine , No. 138 Xianlin Avenue , Nanjing 210023 , China
| | - Ru-Feng Ye
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China
| | - Congmin Yuan
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Shengyang Fang
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Dengqi Xue
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Xicheng Yang
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Hao Chen
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Yilin Zheng
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Linqian Yu
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Lan Zheng
- Minhang Hospital , Fudan University , No. 170 Xinsong Road , Shanghai 201199 , China
| | - Wei Fu
- 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
| | - Zhuibai Qiu
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China
| | - Jinggen Liu
- CAS Key Laboratory of Receptor Research , Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science , 555 Zuchongzhi Road , Shanghai 201203 , China.,University of Chinese Academy of Sciences , No. 19A Yuquan Road , Beijing 100049 , China
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy , Fudan University , No. 826 Zhangheng Road , Shanghai 201203 , China.,State Key Laboratory of Medical Neurobiology , Fudan University , No. 138 Yixueyuan Road , Shanghai 200032 , China
| |
Collapse
|
17
|
A Survey of Molecular Imaging of Opioid Receptors. Molecules 2019; 24:molecules24224190. [PMID: 31752279 PMCID: PMC6891617 DOI: 10.3390/molecules24224190] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 01/09/2023] Open
Abstract
The discovery of endogenous peptide ligands for morphine binding sites occurred in parallel with the identification of three subclasses of opioid receptor (OR), traditionally designated as μ, δ, and κ, along with the more recently defined opioid-receptor-like (ORL1) receptor. Early efforts in opioid receptor radiochemistry focused on the structure of the prototype agonist ligand, morphine, although N-[methyl-11C]morphine, -codeine and -heroin did not show significant binding in vivo. [11C]Diprenorphine ([11C]DPN), an orvinol type, non-selective OR antagonist ligand, was among the first successful PET tracers for molecular brain imaging, but has been largely supplanted in research studies by the μ-preferring agonist [11C]carfentanil ([11C]Caf). These two tracers have the property of being displaceable by endogenous opioid peptides in living brain, thus potentially serving in a competition-binding model. Indeed, many clinical PET studies with [11C]DPN or [11C]Caf affirm the release of endogenous opioids in response to painful stimuli. Numerous other PET studies implicate μ-OR signaling in aspects of human personality and vulnerability to drug dependence, but there have been very few clinical PET studies of μORs in neurological disorders. Tracers based on naltrindole, a non-peptide antagonist of the δ-preferring endogenous opioid enkephalin, have been used in PET studies of δORs, and [11C]GR103545 is validated for studies of κORs. Structures such as [11C]NOP-1A show selective binding at ORL-1 receptors in living brain. However, there is scant documentation of δ-, κ-, or ORL1 receptors in healthy human brain or in neurological and psychiatric disorders; here, clinical PET research must catch up with recent progress in radiopharmaceutical chemistry.
Collapse
|
18
|
Effects of stimulation of mu opioid and nociceptin/orphanin FQ peptide (NOP) receptors on alcohol drinking in rhesus monkeys. Neuropsychopharmacology 2019; 44:1476-1484. [PMID: 30970376 PMCID: PMC6784996 DOI: 10.1038/s41386-019-0390-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022]
Abstract
Alcohol use disorder (AUD) persists as a devastating public health problem; widely effective pharmacological treatments are needed. Evidence from rodent models suggests that stimulating brain receptors for the neuropeptide nociceptin/orphanin FQ (NOP) can decrease ethanol drinking. We characterized the effects of the mu opioid peptide (MOP) receptor agonist buprenorphine and the buprenorphine analog (2S)-2-[(5R,6R,7R,14S)-N-cyclopropylmethyl-4,5-epoxy-6,14-ethano-3-hydroxy-6 methoxymorphinan-7-yl]-3,3-dimethylpentan-2-ol (BU08028), which stimulates MOP and NOP receptors, in a translational nonhuman primate model of AUD. Rhesus monkeys drank a 4% ethanol solution 6 h per day, 5 days per week via an operant behavioral panel in their home cages. To assess behavioral selectivity, monkeys responded via a photo-optic switch to earn food pellets. After characterizing the acute effects of BU08028 (0.001-0.01 mg/kg, i.m.) and buprenorphine (0.003-0.056 mg/kg, i.m.), the drugs were administered chronically using a model of pharmacotherapy assessment that incorporates clinical aspects of AUD and treatment. Acutely, both drugs decreased ethanol drinking at doses that did not affect food-maintained responding. During chronic treatment, effects of BU08028 and buprenorphine were maintained for several weeks without development of tolerance or emergence of adverse effects. BU08028 was ~0.5 and 1.0 log units more potent in acute and chronic studies, respectively. The selective NOP receptor agonist SCH 221510 also selectively decreased ethanol intakes when given acutely (0.03-1.0 mg/kg, i.m.), whereas the MOP antagonist naltrexone (1.7-5.6 mg/kg, i.m.) decreased both ethanol intake and food pellets delivered. These data demonstrate that bifunctional MOP/NOP agonists, which may have therapeutic advantages to MOP-selective drugs, can decrease alcohol drinking in nonhuman primates.
Collapse
|
19
|
Cunningham CW, Elballa WM, Vold SU. Bifunctional opioid receptor ligands as novel analgesics. Neuropharmacology 2019; 151:195-207. [PMID: 30858102 DOI: 10.1016/j.neuropharm.2019.03.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/30/2019] [Accepted: 03/02/2019] [Indexed: 12/12/2022]
Abstract
Prolonged treatment of chronic severe pain with opioid analgesics is frought with problematic adverse effects including tolerance, dependence, and life-threatening respiratory depression. Though these effects are mediated predominately through preferential activation of μ opioid peptide (μOP) receptors, there is an emerging appreciation that actions at κOP and δOP receptors contribute to the observed pharmacologic and behavioral profile of μOP receptor agonists and may be targeted simultaneously to afford improved analgesic effects. Recent developments have also identified the related nociceptin opioid peptide (NOP) receptor as a key modulator of the effects of μOP receptor signaling. We review here the available literature describing OP neurotransmitter systems and highlight recent drug and probe design strategies.
Collapse
Affiliation(s)
| | - Waleed M Elballa
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, Mequon, WI, USA.
| | - Stephanie U Vold
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, Mequon, WI, USA.
| |
Collapse
|
20
|
BU10038 as a safe opioid analgesic with fewer side-effects after systemic and intrathecal administration in primates. Br J Anaesth 2019; 122:e146-e156. [PMID: 30916003 DOI: 10.1016/j.bja.2018.10.065] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 08/29/2018] [Accepted: 10/23/2018] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The marked increase in mis-use of prescription opioids has greatly affected our society. One potential solution is to develop improved analgesics which have agonist action at both mu opioid peptide (MOP) and nociceptin/orphanin FQ peptide (NOP) receptors. BU10038 is a recently identified bifunctional MOP/NOP partial agonist. The aim of this study was to determine the functional profile of systemic or spinal delivery of BU10038 in primates after acute and chronic administration. METHODS A series of behavioural and physiological assays have been established specifically to reflect the therapeutic (analgesia) and side-effects (abuse potential, respiratory depression, itch, physical dependence, and tolerance) of opioid analgesics in rhesus monkeys. RESULTS After systemic administration, BU10038 (0.001-0.01 mg kg-1) dose-dependently produced long-lasting antinociceptive and antihypersensitive effects. Unlike the MOP agonist oxycodone, BU10038 lacked reinforcing effects (i.e. little or no abuse liability), and BU10038 did not compromise the physiological functions of primates including respiration, cardiovascular activities, and body temperature at antinociceptive doses and a 10-30-fold higher dose (0.01-0.1 mg kg-1). After intrathecal administration, BU10038 (3 μg) exerted morphine-comparable antinociception and antihypersensitivity without itch scratching responses. Unlike morphine, BU10038 did not cause the development of physical dependence and tolerance after repeated and chronic administration. CONCLUSIONS These in vivo findings demonstrate the translational potential of bifunctional MOP/NOP receptor agonists such as BU10038 as a safe, non-addictive analgesic with fewer side-effects in primates. This study strongly supports that bifunctional MOP/NOP agonists may provide improved analgesics and an alternative solution for the ongoing prescription opioid crisis.
Collapse
|
21
|
Abstract
The development of nonpeptide systemically active small-molecule NOP-targeted ligands has contributed tremendously to validating the NOP receptor as a promising target for therapeutics. Although a NOP-targeted compound is not yet approved for clinical use, a few NOP ligands are in clinical trials for various indications. Both successful and failed human clinical trials with NOP ligands provide opportunities for rational development of new and improved NOP-targeted compounds. A few years after the discovery of the NOP receptor in 1994, and its de-orphanization upon discovery of the endogenous peptide nociceptin/orphanin FQ (N/OFQ) in 1995, there was a significant effort in the pharmaceutical industry to discover nonpeptide NOP ligands from hits obtained from high-throughput screening campaigns of compound libraries. Depending on the therapeutic indication to be pursued, NOP agonists and antagonists were discovered, and some were optimized as clinical candidates. Advances such as G protein-coupled receptor (GPCR) structure elucidation, functional selectivity in ligand-driven GPCR activation, and multi-targeted ligands provide new scope for the rational design of novel NOP ligands fine-tuned for successful clinical translation. This article reviews the field of nonpeptide NOP ligand drug design in the context of these exciting developments and highlights new optimized nonpeptide NOP ligands possessing interesting functional profiles, which are particularly attractive for several unmet clinical applications involving NOP receptor pharmacomodulation.
Collapse
|
22
|
Kumar V, Polgar WE, Cami-Kobeci G, Thomas MP, Khroyan TV, Toll L, Husbands SM. Synthesis, Biological Evaluation, and SAR Studies of 14β-phenylacetyl Substituted 17-cyclopropylmethyl-7, 8-dihydronoroxymorphinones Derivatives: Ligands With Mixed NOP and Opioid Receptor Profile. Front Psychiatry 2018; 9:430. [PMID: 30283364 PMCID: PMC6156383 DOI: 10.3389/fpsyt.2018.00430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/21/2018] [Indexed: 12/22/2022] Open
Abstract
A series of 14β-acyl substituted 17-cyclopropylmethyl-7,8-dihydronoroxymorphinone compounds has been synthesized and evaluated for affinity and efficacy for mu (MOP), kappa (KOP), and delta (DOP) opioid receptors and nociceptin/orphanin FQ peptide (NOP) receptors. The majority of the new ligands displayed high binding affinities for the three opioid receptors, and moderate affinity for NOP receptors. The affinities for NOP receptors are of particular interest as most classical opioid ligands do not bind to NOP receptors. The predominant activity in the [35S]GTPγS assay was partial agonism at each receptor. The results are consistent with our prediction that an appropriate 14β side chain would access a binding site within the NOP receptor and result in substantially higher affinity than displayed by the parent compound naltrexone. Molecular modeling studies, utilizing the recently reported structure of the NOP receptor, are also consistent with this interpretation.
Collapse
Affiliation(s)
- Vinod Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda, India
| | | | - Gerta Cami-Kobeci
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Mark P. Thomas
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | | | - Lawrence Toll
- Department of Biomedical Sciences, Florida Atlantic University, Boca Raton, FL, United States
| | - Stephen M. Husbands
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| |
Collapse
|
23
|
Bidlack JM, Knapp BI, Deaver DR, Plotnikava M, Arnelle D, Wonsey AM, Fern Toh M, Pin SS, Namchuk MN. In Vitro Pharmacological Characterization of Buprenorphine, Samidorphan, and Combinations Being Developed as an Adjunctive Treatment of Major Depressive Disorder. J Pharmacol Exp Ther 2018; 367:267-281. [PMID: 30108159 DOI: 10.1124/jpet.118.249839] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022] Open
Abstract
A combination of buprenorphine (BUP) and samidorphan (SAM) at a 1:1 (mg/mg) fixed-ratio dose is being investigated as an adjunctive treatment of major depressive disorder (BUP/SAM, ALKS 5461). Both [3H]BUP and [3H]SAM bound to the μ-, κ-, and δ-opioid receptors (MOR, KOR, and DOR, respectively) with Kd values of 3 nM or less. [3H]BUP dissociated from the MOR more slowly than [3H]SAM did. In the [35S]GTPγS assay, BUP was a partial agonist at the MOR, KOR, and DOR. SAM was an antagonist at the MOR and a partial agonist at the KOR and DOR. The pharmacology of the combination of SAM and BUP was characterized at ratios like the molar ratios of both compounds at steady state in humans. In all assessments, SAM reduced the efficacy of BUP at the MOR without altering its potency. At the KOR, SAM had no significant effect on the activity of BUP. In bioluminescent resonance energy transfer assays, SAM, naltrexone, and naloxone were partial agonists when the MOR was coupled to the Gα oB and Gα z, and were antagonists when coupled to Gα i At the KOR, SAM was a partial agonist activating Gα oA and Gα oB and a full agonist in stimulating Gα z SAM inhibited BUP's recruitment of β-arrestin to the MOR, suggesting an attenuation of BUP's efficacy in activating G proteins correlated with an inhibition of β-arrestin recruitment. The collective data suggest that SAM attenuates the efficacy of BUP under all conditions tested at the MOR and DOR but had little effect on BUP activity at the KOR.
Collapse
Affiliation(s)
- Jean M Bidlack
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Brian I Knapp
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Daniel R Deaver
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Margarita Plotnikava
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Derrick Arnelle
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Angela M Wonsey
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - May Fern Toh
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Sokhom S Pin
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Mark N Namchuk
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| |
Collapse
|
24
|
Abstract
The opioid receptor system plays a major role in the regulation of mood, reward, and pain. The opioid receptors therefore make attractive targets for the treatment of many different conditions, including pain, depression, and addiction. However, stimulation or blockade of any one opioid receptor type often leads to on-target adverse effects that limit the clinical utility of a selective opioid agonist or antagonist. Literature precedent suggests that the opioid receptors do not act in isolation and that interactions among the opioid receptors and between the opioid receptors and other proteins may produce clinically useful targets. Multifunctional ligands have the potential to elicit desired outcomes with reduced adverse effects by allowing for the activation of specific receptor conformations and/or signaling pathways promoted as a result of receptor oligomerization or crosstalk. In this chapter, we describe several classes of multifunctional ligands that interact with at least one opioid receptor. These ligands have been designed for biochemical exploration and the treatment of a wide variety of conditions, including multiple kinds of pain, depression, anxiety, addiction, and gastrointestinal disorders. The structures, pharmacological utility, and therapeutic drawbacks of these classes of ligands are discussed.
Collapse
Affiliation(s)
- Jessica P Anand
- Department of Pharmacology, Medical School and the Edward F. Domino Research Center, University of Michigan, Ann Arbor, MI, USA.
| | - Deanna Montgomery
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
25
|
Butelman ER, Kreek MJ. Discriminative Stimulus Properties of Opioid Ligands: Progress and Future Directions. Curr Top Behav Neurosci 2018; 39:175-192. [PMID: 27225498 DOI: 10.1007/7854_2016_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Opioid receptors (MOP-r, KOP-r, DOP-r, as well as NOP-r) and their endogenous neuropeptide agonist systems are involved in diverse neurobiological and behavioral functions, in health and disease. These functions include pain and analgesia, addictions, and psychiatric diseases (e.g., depression-, anxiety-like, and stress-related disorders). Drug discrimination assays have been used to characterize the behavioral pharmacology of ligands with affinity at MOP-r, KOP-r, or DOP-r (and to a lesser extent NOP-r). Therefore, drug discrimination studies with opioid ligands have an important continuing role in translational investigations of diseases that are affected by these neurobiological targets and their pharmacotherapy.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, Box 171, New York, NY, 10065, USA.
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, Box 171, New York, NY, 10065, USA
| |
Collapse
|
26
|
A novel orvinol analog, BU08028, as a safe opioid analgesic without abuse liability in primates. Proc Natl Acad Sci U S A 2016; 113:E5511-8. [PMID: 27573832 DOI: 10.1073/pnas.1605295113] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Despite the critical need, no previous research has substantiated safe opioid analgesics without abuse liability in primates. Recent advances in medicinal chemistry have led to the development of ligands with mixed mu opioid peptide (MOP)/nociceptin-orphanin FQ peptide (NOP) receptor agonist activity to achieve this objective. BU08028 is a novel orvinol analog that displays a similar binding profile to buprenorphine with improved affinity and efficacy at NOP receptors. The aim of this preclinical study was to establish the functional profile of BU08028 in monkeys using clinically used MOP receptor agonists for side-by-side comparisons in various well-honed behavioral and physiological assays. Systemic BU08028 (0.001-0.01 mg/kg) produced potent long-lasting (i.e., >24 h) antinociceptive and antiallodynic effects, which were blocked by MOP or NOP receptor antagonists. More importantly, the reinforcing strength of BU08028 was significantly lower than that of cocaine, remifentanil, or buprenorphine in monkeys responding under a progressive-ratio schedule of drug self-administration. Unlike MOP receptor agonists, BU08028 at antinociceptive doses and ∼10- to 30-fold higher doses did not cause respiratory depression or cardiovascular adverse events as measured by telemetry devices. After repeated administration, the monkeys developed acute physical dependence on morphine, as manifested by precipitated withdrawal signs, such as increased respiratory rate, heart rate, and blood pressure. In contrast, monkeys did not show physical dependence on BU08028. These in vivo findings in primates not only document the efficacy and tolerability profile of bifunctional MOP/NOP receptor agonists, but also provide a means of translating such ligands into therapies as safe and potentially abuse-free opioid analgesics.
Collapse
|
27
|
Guillemyn K, Starnowska J, Lagard C, Dyniewicz J, Rojewska E, Mika J, Chung NN, Utard V, Kosson P, Lipkowski AW, Chevillard L, Arranz-Gibert P, Teixidó M, Megarbane B, Tourwé D, Simonin F, Przewlocka B, Schiller PW, Ballet S. Bifunctional Peptide-Based Opioid Agonist-Nociceptin Antagonist Ligands for Dual Treatment of Acute and Neuropathic Pain. J Med Chem 2016; 59:3777-92. [PMID: 27035422 DOI: 10.1021/acs.jmedchem.5b01976] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Herein, the opioid pharmacophore H-Dmt-d-Arg-Aba-β-Ala-NH2 (7) was linked to peptide ligands for the nociceptin receptor. Combination of 7 and NOP ligands (e.g., H-Arg-Tyr-Tyr-Arg-Ile-Lys-NH2) led to binding affinities in the low nanomolar domain. In vitro, the hybrids behaved as agonists at the opioid receptors and antagonists at the nociceptin receptor. Intravenous administration of hybrid 13a (H-Dmt-d-Arg-Aba-β-Ala-Arg-Tyr-Tyr-Arg-Ile-Lys-NH2) to mice resulted in potent and long lasting antinociception in the tail-flick test, indicating that 13a was able to permeate the BBB. This was further supported by a cell-based BBB model. All hybrids alleviated allodynia and hyperalgesia in neuropathic pain models. Especially with respect to hyperalgesia, they showed to be more effective than the parent compounds. Hybrid 13a did not result in significant respiratory depression, in contrast to an equipotent analgesic dose of morphine. These hybrids hence represent a promising avenue toward analgesics for the dual treatment of acute and neuropathic pain.
Collapse
Affiliation(s)
- Karel Guillemyn
- Research Group of Organic Chemistry, Departments of Chemistry and Bio-engineering Sciences, Vrije Universiteit Brussel , Pleinlaan 2, 1050 Brussels, Belgium
| | - Joanna Starnowska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences , Smetna 12, PL 31-343 Kraków, Poland
| | - Camille Lagard
- Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Réanimation Médicale et Toxicologique, Inserm U1144, Université Paris Descartes UMR-S 1144, Université Paris Didero, UMR-S 1144 , Paris, France
| | - Jolanta Dyniewicz
- Neuropeptide Laboratory, Medical Research Centre, Polish Academy of Sciences , 5 Pawinskiego Street, PL 02-106 Warsaw, Poland
| | - Ewelina Rojewska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences , Smetna 12, PL 31-343 Kraków, Poland
| | - Joanna Mika
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences , Smetna 12, PL 31-343 Kraków, Poland
| | - Nga N Chung
- Department of Chemical Biology and Peptide Research, Clinical Research Institute , 110 Avenue Des Pins Ouest, Montreal, Quebec H2W 1R7, Canada
| | - Valérie Utard
- University of Strasbourg, CNRS, UMR7242, ESBS , 67412 Illkirch-Graffenstaden, France
| | - Piotr Kosson
- Neuropeptide Laboratory, Medical Research Centre, Polish Academy of Sciences , 5 Pawinskiego Street, PL 02-106 Warsaw, Poland
| | - Andrzej W Lipkowski
- Neuropeptide Laboratory, Medical Research Centre, Polish Academy of Sciences , 5 Pawinskiego Street, PL 02-106 Warsaw, Poland
| | - Lucie Chevillard
- Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Réanimation Médicale et Toxicologique, Inserm U1144, Université Paris Descartes UMR-S 1144, Université Paris Didero, UMR-S 1144 , Paris, France
| | - Pol Arranz-Gibert
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST) , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Meritxell Teixidó
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST) , Baldiri Reixac 10, 08028 Barcelona, Spain
| | - Bruno Megarbane
- Assistance Publique-Hôpitaux de Paris, Hôpital Lariboisière, Réanimation Médicale et Toxicologique, Inserm U1144, Université Paris Descartes UMR-S 1144, Université Paris Didero, UMR-S 1144 , Paris, France
| | - Dirk Tourwé
- Research Group of Organic Chemistry, Departments of Chemistry and Bio-engineering Sciences, Vrije Universiteit Brussel , Pleinlaan 2, 1050 Brussels, Belgium
| | - Frédéric Simonin
- University of Strasbourg, CNRS, UMR7242, ESBS , 67412 Illkirch-Graffenstaden, France
| | - Barbara Przewlocka
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences , Smetna 12, PL 31-343 Kraków, Poland
| | - Peter W Schiller
- Department of Chemical Biology and Peptide Research, Clinical Research Institute , 110 Avenue Des Pins Ouest, Montreal, Quebec H2W 1R7, Canada
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bio-engineering Sciences, Vrije Universiteit Brussel , Pleinlaan 2, 1050 Brussels, Belgium
| |
Collapse
|
28
|
Zielińska M, Ben Haddou T, Cami-Kobeci G, Sałaga M, Jarmuż A, Padysz M, Kordek R, Spetea M, Husbands SM, Fichna J. Anti-inflammatory effect of dual nociceptin and opioid receptor agonist, BU08070, in experimental colitis in mice. Eur J Pharmacol 2015; 765:582-90. [PMID: 26404500 DOI: 10.1016/j.ejphar.2015.09.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 09/12/2015] [Accepted: 09/15/2015] [Indexed: 02/06/2023]
Abstract
Endogenous opioid and nociceptin systems are widely distributed in the gastrointestinal tract where they seem to play a crucial role in maintaining the intestinal homeostasis. The aim of our study was to assess whether activation of nociceptin (NOP) and µ-opioid (MOP) receptors by a mixed NOP/MOP receptor agonist, BU08070, induces anti-inflammatory response in experimental colitis. The anti-inflammatory effect of BU08070 (1 mg/kg i.p.) was characterized in the mouse model of 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis, based on the assessment of the macroscopic and microscopic total damage scores and determination of myeloperoxidase (MPO) activity and TNF-α level in the colon. The effect of BU08070 on cell viability and NF-κB was characterized in THP-1 Blue cell line. The antinociceptive activity of BU08070 was examined in mustard oil-induced mouse model of abdominal pain. A potent anti-inflammatory effect of BU08070 (1 mg/kg i.p.) was observed as indicated by decrease in macroscopic damage score (1.88±0.39 vs. 5.19±0.43 units in TNBS alone treated mice), MPO activity (2.29±0.37 vs. 9.64±2.55 units) and TNF-α level in the colon (35.85±2.45 vs. 49.79±3.81 pg/ml). The anti-inflammatory effect of BU08070 was reversed by selective NOP and MOP receptor antagonists. BU08070 produced concentration-dependent inhibition of TNF-α and LPS-induced NF-κB activation. BU08070 exerted antinociceptive action in mice with experimental colitis. In conclusion, BU08070 significantly reduced the severity of colitis in TNBS-treated mice compared with controls. These results suggest that BU08070 is a potential therapeutic agent for inflammatory bowel diseases therapy.
Collapse
Affiliation(s)
- Marta Zielińska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Tanila Ben Haddou
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Gerta Cami-Kobeci
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Maciej Sałaga
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Agata Jarmuż
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Milena Padysz
- Department of Gastroenterology, Faculty of Military Medicine, Medical University of Lodz, Lodz, Poland
| | - Radzisław Kordek
- Department of Pathology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | | | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
29
|
Cueva JP, Roche C, Ostovar M, Kumar V, Clark MJ, Hillhouse TM, Lewis JW, Traynor JR, Husbands SM. C7β-methyl analogues of the orvinols: the discovery of kappa opioid antagonists with nociceptin/orphanin FQ peptide (NOP) receptor partial agonism and low, or zero, efficacy at mu opioid receptors. J Med Chem 2015; 58:4242-9. [PMID: 25898137 PMCID: PMC4450370 DOI: 10.1021/acs.jmedchem.5b00130] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Buprenorphine is a successful analgesic and treatment for opioid abuse, with both activities relying on its partial agonist activity at mu opioid receptors. However, there is substantial interest in its activities at the kappa opioid and nociceptin/orphanin FQ peptide receptors. This has led to an interest in developing compounds with a buprenorphine-like pharmacological profile but with lower efficacy at mu opioid receptors. The present article describes aryl ring analogues of buprenorphine in which the standard C20-methyl group has been moved to the C7β position, resulting in ligands with the desired profile. In particular, moving the methyl group has resulted in far more robust kappa opioid antagonist activity than seen in the standard orvinol series. Of the compounds synthesized, a number, including 15a, have a profile of interest for the development of drug abuse relapse prevention therapies or antidepressants and others (e.g., 8c), as analgesics with a reduced side-effect profile.
Collapse
Affiliation(s)
- Juan Pablo Cueva
- †Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, United Kingdom
| | - Christopher Roche
- †Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, United Kingdom
| | - Mehrnoosh Ostovar
- †Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, United Kingdom
| | - Vinod Kumar
- †Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, United Kingdom
| | - Mary J Clark
- ‡Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Todd M Hillhouse
- ‡Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - John W Lewis
- †Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, United Kingdom
| | - John R Traynor
- ‡Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Stephen M Husbands
- †Department of Pharmacy and Pharmacology, University of Bath, Bath, BA2 7AY, United Kingdom
| |
Collapse
|
30
|
Khroyan TV, Wu J, Polgar WE, Cami-Kobeci G, Fotaki N, Husbands SM, Toll L. BU08073 a buprenorphine analogue with partial agonist activity at μ-receptors in vitro but long-lasting opioid antagonist activity in vivo in mice. Br J Pharmacol 2014; 172:668-80. [PMID: 24903063 DOI: 10.1111/bph.12796] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 05/22/2014] [Accepted: 05/25/2014] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Buprenorphine is a potent analgesic with high affinity at μ, δ and κ and moderate affinity at nociceptin opioid (NOP) receptors. Nevertheless, NOP receptor activation modulates the in vivo activity of buprenorphine. Structure activity studies were conducted to design buprenorphine analogues with high affinity at each of these receptors and to characterize them in in vitro and in vivo assays. EXPERIMENTAL APPROACH Compounds were tested for binding affinity and functional activity using [(35) S]GTPγS binding at each receptor and a whole-cell fluorescent assay at μ receptors. BU08073 was evaluated for antinociceptive agonist and antagonist activity and for its effects on anxiety in mice. KEY RESULTS BU08073 bound with high affinity to all opioid receptors. It had virtually no efficacy at δ, κ and NOP receptors, whereas at μ receptors, BU08073 has similar efficacy as buprenorphine in both functional assays. Alone, BU08073 has anxiogenic activity and produces very little antinociception. However, BU08073 blocks morphine and U50,488-mediated antinociception. This blockade was not evident at 1 h post-treatment, but is present at 6 h and remains for up to 3-6 days. CONCLUSIONS AND IMPLICATIONS These studies provide structural requirements for synthesis of 'universal' opioid ligands. BU08073 had high affinity for all the opioid receptors, with moderate efficacy at μ receptors and reduced efficacy at NOP receptors, a profile suggesting potential analgesic activity. However, in vivo, BU08073 had long-lasting antagonist activity, indicating that its pharmacokinetics determined both the time course of its effects and what receptor-mediated effects were observed. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2.
Collapse
|
31
|
Kumar V, Ridzwan IE, Grivas K, Lewis JW, Clark MJ, Meurice C, Jimenez-Gomez C, Pogozheva I, Mosberg H, Traynor JR, Husbands SM. Selectively promiscuous opioid ligands: discovery of high affinity/low efficacy opioid ligands with substantial nociceptin opioid peptide receptor affinity. J Med Chem 2014; 57:4049-57. [PMID: 24761755 PMCID: PMC4033651 DOI: 10.1021/jm401964y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
![]()
Emerging clinical and preclinical
evidence suggests that a compound
displaying high affinity for μ, κ, and δ opioid
(MOP, KOP, and DOP) receptors and antagonist activity at each, coupled
with moderate affinity and efficacy at nociceptin opioid peptide (NOP)
receptors will have utility as a relapse prevention agent for multiple
types of drug abuse. Members of the orvinol family of opioid ligands
have the desired affinity profile but have typically displayed substantial
efficacy at MOP and or KOP receptors. In this study it is shown that
a phenyl ring analogue (1d) of buprenorphine displays
the desired profile in vitro with high, nonselective affinity for
the MOP, KOP, and DOP receptors coupled with moderate affinity for
NOP receptors. In vivo, 1d lacked any opioid agonist
activity and was an antagonist of both the MOP receptor agonist morphine
and the KOP receptor agonist ethylketocyclazocine, confirming the
desired opioid receptor profile in vivo.
Collapse
Affiliation(s)
- Vinod Kumar
- Department of Pharmacy and Pharmacology, University of Bath , Bath, BA2 7AY, U.K
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Sobczak M, Cami-Kobeci G, Sałaga M, Husbands SM, Fichna J. Novel mixed NOP/MOP agonist BU08070 alleviates pain and inhibits gastrointestinal motility in mouse models mimicking diarrhea-predominant irritable bowel syndrome symptoms. Eur J Pharmacol 2014; 736:63-9. [PMID: 24815321 DOI: 10.1016/j.ejphar.2014.04.038] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/23/2014] [Accepted: 04/24/2014] [Indexed: 12/13/2022]
Abstract
The opioid and nociceptin systems play a crucial role in the maintenance of homeostasis in the gastrointestinal (GI) tract. The aim of this study was to characterize the effect of BU08070, a novel mixed MOP/NOP agonist, on mouse intestinal contractility in vitro and GI motility in vivo in physiological conditions and in animal models mimicking symptoms of irritable bowel syndrome (IBS), including diarrhea and abdominal pain. The effect of BU08070 on muscle contractility in vitro was characterized in the ileum and colon. To assess the effect of BU08070 in vivo, the following parameters were assessed: whole GI transit, gastric emptying, geometric center, colonic bead expulsion, fecal pellet output and time to castor oil-induced diarrhea. The antinociceptive activity of BU08070 was characterized in the mustard oil (MO)-induced abdominal pain model and the writhing test, alone and in the presence of MOP and NOP antagonists. in vitro, BU08070 (10(-10)-10(-6) M) inhibited colonic and ileal smooth muscle contractions in a concentration-dependent manner. in vivo, BU08070 prolonged the whole GI transit and inhibited colonic bead expulsion. The antitransit and antidiarrheal effects of BU08070 were observed already at the dose of 0.1 mg/kg (i.p.). BU08070 reversed hypermotility and reduced pain in mouse models mimicking IBS-D symptoms. Our results suggest that BU08070 has a potential of becoming an efficient drug in IBS-D therapy. Here we also validate mixed NOP/MOP receptor targeting as possible future treatment of functional GI diseases.
Collapse
Affiliation(s)
- Marta Sobczak
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Gerta Cami-Kobeci
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Maciej Sałaga
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | | | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland.
| |
Collapse
|
33
|
Lin AP, Ko MC. The therapeutic potential of nociceptin/orphanin FQ receptor agonists as analgesics without abuse liability. ACS Chem Neurosci 2013; 4:214-24. [PMID: 23421672 DOI: 10.1021/cn300124f] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Although mu opioid (MOP) receptor agonists are the most commonly used analgesics for the treatment of moderate to severe pain in the clinic, the side effects of MOP agonists such as abuse liability limit their value as a medication. Research to identify novel analgesics without adverse effects is pivotal to advance the health care of humans. The nociceptin/orphanin FQ peptide (NOP) receptor, the fourth opioid receptor subtype, mediates distinctive actions in nonhuman primates which suggests the possibility that activity at this receptor may result in strong analgesia in the absence of virtually all of the side effects associated with MOP agonists. The present review highlights the recent progress of pharmacological studies of NOP-related ligands in primates. Selective NOP agonists, either peptidic or nonpeptidic, produce full analgesia in various assays in primates, when delivered systemically or intrathecally. Yet small molecule NOP agonists do not serve as reinforcers, indicating a lack of abuse liability. Given that NOP agonists have low abuse liability and that coactivation of NOP and MOP receptors produces synergistic antinociception, it is worth developing bifunctional NOP/MOP ligands. The outcomes of these studies and recent developments provide new perspectives to establish a translational bridge for understanding the biobehavioral functions of NOP receptors in primates and for facilitating the development of NOP-related ligands as a new generation of analgesics without abuse liability in humans.
Collapse
Affiliation(s)
- Ann P. Lin
- Department of Psychology, School
of Arts and Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Mei-Chuan Ko
- Department of Pharmacology,
Medical School, University of Michigan,
Ann Arbor, Michigan 48109, United States
| |
Collapse
|
34
|
Abstract
This paper is the thirty-fourth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2011 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
35
|
Cremeans CM, Gruley E, Kyle DJ, Ko MC. Roles of μ-opioid receptors and nociceptin/orphanin FQ peptide receptors in buprenorphine-induced physiological responses in primates. J Pharmacol Exp Ther 2012; 343:72-81. [PMID: 22743574 PMCID: PMC3464037 DOI: 10.1124/jpet.112.194308] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 06/20/2012] [Indexed: 11/22/2022] Open
Abstract
Buprenorphine is known as a μ-opioid peptide (MOP) receptor agonist, but its antinociception is compromised by the activation of nociceptin/orphanin FQ peptide (NOP) receptors in rodents. The aim of this study was to investigate the roles of MOP and NOP receptors in regulating buprenorphine-induced physiological responses in primates (rhesus monkeys). The effects of MOP antagonist (naltrexone), NOP antagonist [(±)-1-[(3R*,4R*)-1-(cyclooctylmethyl)-3-(hydroxymethyl)-4-piperidinyl]-3-ethyl-1,3-dihydro-2H-benzimidazol-2-one (J-113397)], and NOP agonists [(1S,3aS)-8-(2,3,3a,4,5,6-hexahydro-1H-phenalen-1-yl)-1-phenyl-1,3,8-triaza-spiro[4.5] decan-4-one (Ro 64-6198) and 3-endo-8-[bis(2-methylphenyl)methyl]-3-phenyl-8-azabicyclo[3.2.1]octan-3-ol (SCH 221510)] on buprenorphine were studied in three functional assays for measuring analgesia, respiratory depression, and itch in primates. Over the dose range of 0.01 to 0.1 mg/kg, buprenorphine dose-dependently produced antinociception, respiratory depression, and itch/scratching responses, and there was a ceiling effect at higher doses (0.1-1 mg/kg). Naltrexone (0.03 mg/kg) produced similar degrees of rightward shifts of buprenorphine's dose-response curves for all three endpoints. Mean pK(B) values of naltrexone (8.1-8.3) confirmed that MOP receptors mediated mainly buprenorphine-induced antinociception, respiratory depression, and itch/scratching. In contrast, J-113397 (0.1 mg/kg) did not change buprenorphine-induced physiological responses, indicating that there were no functional NOP receptors in buprenorphine-induced effects. More importantly, both NOP agonists, Ro 64-6198 and SCH 221510, enhanced buprenorphine-induced antinociception without respiratory depression and itch/ scratching. The dose-addition analysis revealed that buprenorphine in combination with the NOP agonist synergistically produced antinociceptive effects. These findings provided functional evidence that the activation of NOP receptors did not attenuate buprenorphine-induced antinociception in primates; instead, the coactivation of MOP and NOP receptors produced synergistic antinociception without other side effects. This study strongly supports the therapeutic potential of mixed MOP/NOP agonists as innovative analgesics.
Collapse
Affiliation(s)
- Colette M Cremeans
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-5632, USA
| | | | | | | |
Collapse
|