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
|
Thouaye M, Yalcin I. Neuropathic pain: From actual pharmacological treatments to new therapeutic horizons. Pharmacol Ther 2023; 251:108546. [PMID: 37832728 DOI: 10.1016/j.pharmthera.2023.108546] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 09/07/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023]
Abstract
Neuropathic pain, caused by a lesion or disease affecting the somatosensory system, affects between 3 and 17% of the general population. The treatment of neuropathic pain is challenging due to its heterogeneous etiologies, lack of objective diagnostic tools and resistance to classical analgesic drugs. First-line treatments recommended by the Special Interest Group on Neuropathic Pain (NeuPSIG) and European Federation of Neurological Societies (EFNS) include gabapentinoids, tricyclic antidepressants (TCAs) and selective serotonin noradrenaline reuptake inhibitors (SNRIs). Nevertheless these treatments have modest efficacy or dose limiting side effects. There is therefore a growing number of preclinical and clinical studies aim at developing new treatment strategies to treat neuropathic pain with better efficacy, selectivity, and less side effects. In this review, after a brief description of the mechanisms of action, efficacy, and limitations of current therapeutic drugs, we reviewed new preclinical and clinical targets currently under investigation, as well as promising non-pharmacological alternatives and their potential co-use with pharmacological treatments.
Collapse
Affiliation(s)
- Maxime Thouaye
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Ipek Yalcin
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France; Department of Psychiatry and Neuroscience, Université Laval, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
3
|
Ding H, Kiguchi N, Dobbins M, Romero-Sandoval EA, Kishioka S, Ko MC. Nociceptin Receptor-Related Agonists as Safe and Non-addictive Analgesics. Drugs 2023; 83:771-793. [PMID: 37209211 PMCID: PMC10948013 DOI: 10.1007/s40265-023-01878-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2023] [Indexed: 05/22/2023]
Abstract
As clinical use of currently available opioid analgesics is often impeded by dose-limiting adverse effects, such as abuse liability and respiratory depression, new approaches have been pursued to develop safe, effective, and non-addictive pain medications. After the identification of the nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor more than 25 years ago, NOP receptor-related agonists have emerged as a promising target for developing novel and effective opioids that modulate the analgesic and addictive properties of mu-opioid peptide (MOP) receptor agonists. In this review, we highlight the effects of the NOP receptor-related agonists compared with those of MOP receptor agonists in experimental rodent and more translational non-human primate (NHP) models and the development status of key NOP receptor-related agonists as potential safe and non-addictive analgesics. Several lines of evidence demonstrated that peptidic and non-peptidic NOP receptor agonists produce potent analgesic effects by intrathecal delivery in NHPs. Moreover, mixed NOP/MOP receptor partial agonists (e.g., BU08028, BU10038, and AT-121) display potent analgesic effects when administered intrathecally or systemically, without eliciting adverse effects, such as respiratory depression, itch behavior, and signs of abuse liability. More importantly, cebranopadol, a mixed NOP/opioid receptor agonist with full efficacy at NOP and MOP receptors, produces robust analgesic efficacy with reduced adverse effects, conferring promising outcomes in clinical studies. A balanced coactivation of NOP and MOP receptors is a strategy that warrants further exploration and refinement for the development of novel analgesics with a safer and effective profile.
Collapse
Affiliation(s)
- Huiping Ding
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Norikazu Kiguchi
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, 640-8156, Japan
| | - MaryBeth Dobbins
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - E Alfonso Romero-Sandoval
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Shiroh Kishioka
- Faculty of Wakayama Health Care Sciences, Takarazuka University of Medical and Health Care, Wakayama, 640-8392, Japan
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
4
|
Caminski ES, Antunes FTT, Souza IA, Dallegrave E, Zamponi GW. Regulation of N-type calcium channels by nociceptin receptors and its possible role in neurological disorders. Mol Brain 2022; 15:95. [PMID: 36434658 PMCID: PMC9700961 DOI: 10.1186/s13041-022-00982-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/15/2022] [Indexed: 11/27/2022] Open
Abstract
Activation of nociceptin opioid peptide receptors (NOP, a.k.a. opioid-like receptor-1, ORL-1) by the ligand nociceptin/orphanin FQ, leads to G protein-dependent regulation of Cav2.2 (N-type) voltage-gated calcium channels (VGCCs). This typically causes a reduction in calcium currents, triggering changes in presynaptic calcium levels and thus neurotransmission. Because of the widespread expression patterns of NOP and VGCCs across multiple brain regions, the dorsal horn of the spinal cord, and the dorsal root ganglia, this results in the alteration of numerous neurophysiological features. Here we review the regulation of N-type calcium channels by the NOP-nociceptin system in the context of neurological conditions such as anxiety, addiction, and pain.
Collapse
Affiliation(s)
- Emanuelle Sistherenn Caminski
- grid.412344.40000 0004 0444 6202Graduate Program in Health Sciences, Laboratory of Research in Toxicology (LAPETOX), Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS Brazil
| | - Flavia Tasmin Techera Antunes
- grid.22072.350000 0004 1936 7697Department of Clinical Neurosciences, University of Calgary, Calgary, AB Canada ,grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| | - Ivana Assis Souza
- grid.22072.350000 0004 1936 7697Department of Clinical Neurosciences, University of Calgary, Calgary, AB Canada ,grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| | - Eliane Dallegrave
- grid.412344.40000 0004 0444 6202Graduate Program in Health Sciences, Laboratory of Research in Toxicology (LAPETOX), Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS Brazil
| | - Gerald W. Zamponi
- grid.22072.350000 0004 1936 7697Department of Clinical Neurosciences, University of Calgary, Calgary, AB Canada ,grid.22072.350000 0004 1936 7697Hotchkiss Brain Institute, University of Calgary, Calgary, AB Canada
| |
Collapse
|
5
|
Wtorek K, Ghidini A, Gentilucci L, Adamska-Bartłomiejczyk A, Piekielna-Ciesielska J, Ruzza C, Sturaro C, Calò G, Pieretti S, Kluczyk A, McDonald J, Lambert DG, Janecka A. Synthesis, Biological Activity and Molecular Docking of Chimeric Peptides Targeting Opioid and NOP Receptors. Int J Mol Sci 2022; 23:12700. [PMID: 36293553 PMCID: PMC9604311 DOI: 10.3390/ijms232012700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
Recently, mixed opioid/NOP agonists came to the spotlight for their favorable functional profiles and promising outcomes in clinical trials as novel analgesics. This study reports on two novel chimeric peptides incorporating the fragment Tyr-c[D-Lys-Phe-Phe]Asp-NH2 (RP-170), a cyclic peptide with high affinity for µ and κ opioid receptors (or MOP and KOP, respectively), conjugated with the peptide Ac-RYYRIK-NH2, a known ligand of the nociceptin/orphanin FQ receptor (NOP), yielding RP-170-RYYRIK-NH2 (KW-495) and RP-170-Gly3-RYYRIK-NH2 (KW-496). In vitro, the chimeric KW-496 gained affinity for KOP, hence becoming a dual KOP/MOP agonist, while KW-495 behaved as a mixed MOP/NOP agonist with low nM affinity. Hence, KW-495 was selected for further in vivo experiments. Intrathecal administration of this peptide in mice elicited antinociceptive effects in the hot-plate test; this action was sensitive to both the universal opioid receptor antagonist naloxone and the selective NOP antagonist SB-612111. The rotarod test revealed that KW-495 administration did not alter the mice motor coordination performance. Computational studies have been conducted on the two chimeras to investigate the structural determinants at the basis of the experimental activities, including any role of the Gly3 spacer.
Collapse
Affiliation(s)
- Karol Wtorek
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Alessia Ghidini
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Luca Gentilucci
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | | | | | - Chiara Ruzza
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Chiara Sturaro
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Girolamo Calò
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo Meneghetti 2, 35131 Padova, Italy
| | - Stefano Pieretti
- Istituto Superiore di Sanità, National Center for Drug Research and Evaluation, 00161 Rome, Italy
| | - Alicja Kluczyk
- Faculty of Chemistry, University of Wroclaw, 50-383 Wroclaw, Poland
| | - John McDonald
- Department of Cardiovascular Sciences, University of Leicester, Anaesthesia, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - David G. Lambert
- Department of Cardiovascular Sciences, University of Leicester, Anaesthesia, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| |
Collapse
|
6
|
Zhang J, Song C, Dai J, Li L, Yang X, Chen Z. Mechanism of opioid addiction and its intervention therapy: Focusing on the reward circuitry and mu-opioid receptor. MedComm (Beijing) 2022; 3:e148. [PMID: 35774845 PMCID: PMC9218544 DOI: 10.1002/mco2.148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 05/03/2022] [Accepted: 05/07/2022] [Indexed: 11/09/2022] Open
Abstract
Opioid abuse and addiction have become a global pandemic, posing tremendous health and social burdens. The rewarding effects and the occurrence of withdrawal symptoms are the two mainstays of opioid addiction. Mu-opioid receptors (MORs), a member of opioid receptors, play important roles in opioid addiction, mediating both the rewarding effects of opioids and opioid withdrawal syndrome (OWS). The underlying mechanism of MOR-mediated opioid rewarding effects and withdrawal syndrome is of vital importance to understand the nature of opioid addiction and also provides theoretical basis for targeting MORs to treat drug addiction. In this review, we first briefly introduce the basic concepts of MORs, including their structure, distribution in the nervous system, endogenous ligands, and functional characteristics. We focused on the brain circuitry and molecular mechanism of MORs-mediated opioid reward and withdrawal. The neuroanatomical and functional elements of the neural circuitry of the reward system underlying opioid addiction were thoroughly discussed, and the roles of MOR within the reward circuitry were also elaborated. Furthermore, we interrogated the roles of MORs in OWS, along with the structural basis and molecular adaptions of MORs-mediated withdrawal syndrome. Finally, current treatment strategies for opioid addiction targeting MORs were also presented.
Collapse
Affiliation(s)
- Jia‐Jia Zhang
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Chang‐Geng Song
- Department of NeurologyXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Ji‐Min Dai
- Department of Hepatobiliary SurgeryXijing HospitalThe Fourth Military Medical UniversityXi'anChina
| | - Ling Li
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Xiang‐Min Yang
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| | - Zhi‐Nan Chen
- National Translational Science Center for Molecular Medicine & Department of Cell BiologyThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
7
|
Attenuated G protein signaling and minimal receptor phosphorylation as a biochemical signature of low side-effect opioid analgesics. Sci Rep 2022; 12:7154. [PMID: 35504962 PMCID: PMC9065038 DOI: 10.1038/s41598-022-11189-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 04/01/2022] [Indexed: 12/20/2022] Open
Abstract
Multi-receptor targeting has been proposed as a promising strategy for the development of opioid analgesics with fewer side effects. Cebranopadol and AT-121 are prototypical bifunctional ligands targeting the nociceptin/orphanin FQ peptide receptor (NOP) and µ-opioid receptor (MOP) that elicit potent analgesia in humans and nonhuman primates, respectively. Cebranopadol was reported to produce typical MOP-related side effects such as respiratory depression and reward, whereas AT-121 appeared to be devoid of these liabilities. However, the molecular basis underlying different side effect profiles in opioid analgesics remains unknown. Here, we examine agonist-induced receptor phosphorylation and G protein signaling profiles of a series of chemically diverse mixed MOP/NOP agonists, including cebranopadol and AT-121. We found that these compounds produce strikingly different MOP phosphorylation profiles. Cebranopadol, AT-034 and AT-324 stimulated extensive MOP phosphorylation, whereas AT-201 induced selective phosphorylation at S375 only. AT-121, on the other hand, did not promote any detectable MOP phosphorylation. Conversely, none of these compounds was able to elicit strong NOP phosphorylation and low NOP receptor phosphorylation correlated with partial agonism in a GIRK-channel assay. Our results suggest a close correlation between MOP receptor phosphorylation and side effect profile. Thus, bifunctional MOP/NOP opioid ligands combining low efficacy G protein signaling at both NOP and MOP with no detectable receptor phosphorylation appear to be devoid of side-effects such as respiratory depression, abuse liability or tolerance development, as with AT-121.
Collapse
|
8
|
Spotlight on Nociceptin/Orphanin FQ Receptor in the Treatment of Pain. Molecules 2022; 27:molecules27030595. [PMID: 35163856 PMCID: PMC8838650 DOI: 10.3390/molecules27030595] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 01/24/2023] Open
Abstract
In our society today, pain has become a main source of strain on most individuals. It is crucial to develop novel treatments against pain while focusing on decreasing their adverse effects. Throughout the extent of development for new pain therapies, the nociceptin/orphanin FQ receptor (NOP receptor) has appeared to be an encouraging focal point. Concentrating on NOP receptor to treat chronic pain with limited range of unwanted effects serves as a suitable alternative to prototypical opioid morphine that could potentially lead to life-threatening effects caused by respiratory depression in overdose, as well as generate abuse and addiction. In addition to these harmful effects, the uprising opioid epidemic is responsible for becoming one of the most disastrous public health issues in the US. In this article, the contributing molecular and cellular structure in controlling the cellular trafficking of NOP receptor and studies that support the role of NOP receptor and its ligands in pain management are reviewed.
Collapse
|
9
|
Wang Y, Qin D, Guo Z, Shi F, Cannella N, Ciccocioppo R, Li H. Research progress on the potential novel analgesic BU08028. Eur J Pharmacol 2022; 914:174678. [PMID: 34875275 DOI: 10.1016/j.ejphar.2021.174678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 11/03/2022]
Abstract
Pain is a common symptom accompanying several clinical conditions and causes serious distress to patients. Addressing pain management is an important aspect of disease treatment, including cancer therapy. Opioid analgesics used to manage pain in human and veterinary medicine have been associated with substance dependence and other adverse effects, thereby limiting their application. Thus, the development of opioid analgesics with good safety profiles with minimal adverse effects and no addictive effects, is presently the focus of pain research. As a new potential analgesic, (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) has fewer adverse effects than other analgesics and is expected to be a safer alternative. In this review, we discuss the development of the opioid analog BU08028 and summarize its analgesic effects and biological characteristics, including efficiency, safety, and tolerance. Furthermore, we elaborate on studies showing the bifunctional effect of BU08028, which targets both mu opioid peptide and nociceptin-orphanin FQ peptide receptors, as well as the unique advantages of using BU08028 over single-target opioid agonists. Previous studies have suggested that BU08028 can not only weaken the reward and abuse effects of opioids and other drugs, but also enhance the anti-nociceptive effect of the mu opioid peptide receptors, making it a potent analgesic. Besides, we describe studies suggesting that BU08028 inhibits the effects of alcohol, making it a candidate drug for the management of alcohol addiction. Our review suggests that BU08028 is a potential novel medicine for managing pain and addiction.
Collapse
Affiliation(s)
- Ya Wang
- School of Chemical Engineering, Changchun University of Technology, Changchun, 130012, China
| | - Di Qin
- China-Japan Union Hospital of Jilin University, Changchun, 130033, China.
| | - Zhihua Guo
- School of Chemical Engineering, Changchun University of Technology, Changchun, 130012, China
| | - Fuqiang Shi
- School of Chemical Engineering, Changchun University of Technology, Changchun, 130012, China
| | - Nazzareno Cannella
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, Camerino, 62032, Italy
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna delle Carceri 9, Camerino, 62032, Italy
| | - Hongwu Li
- School of Chemical Engineering, Changchun University of Technology, Changchun, 130012, China.
| |
Collapse
|
10
|
Kiguchi N, Ko MC. Potential therapeutic targets for the treatment of opioid abuse and pain. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2021; 93:335-371. [PMID: 35341570 PMCID: PMC10948018 DOI: 10.1016/bs.apha.2021.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Although μ-opioid peptide (MOP) receptor agonists are effective analgesics available in clinical settings, their serious adverse effects put limits on their use. The marked increase in abuse and misuse of prescription opioids for pain relief and opioid overdose mortality in the past decade has seriously impacted society. Therefore, safe analgesics that produce potent analgesic effects without causing MOP receptor-related adverse effects are needed. This review highlights the potential therapeutic targets for the treatment of opioid abuse and pain based on available evidence generated through preclinical studies and clinical trials. To ameliorate the abuse-related effects of opioids, orexin-1 receptor antagonists and mixed nociceptin/MOP partial agonists have shown promising results in translational aspects of animal models. There are several promising non-opioid targets for selectively inhibiting pain-related responses, including nerve growth factor inhibitors, voltage-gated sodium channel inhibitors, and cannabinoid- and nociceptin-related ligands. We have also discussed several emerging and novel targets. The current medications for opioid abuse are opioid receptor-based ligands. Although neurobiological studies in rodents have discovered several non-opioid targets, there is a translational gap between rodents and primates. Given that the neuroanatomical aspects underlying opioid abuse and pain are different between rodents and primates, it is pivotal to investigate the functional profiles of these non-opioid compounds compared to those of clinically used drugs in non-human primate models before initiating clinical trials. More pharmacological studies of the functional efficacy, selectivity, and tolerability of these newly discovered compounds in non-human primates will accelerate the development of effective medications for opioid abuse and pain.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan.
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
11
|
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: 12] [Impact Index Per Article: 3.0] [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
|
12
|
Dumitrascuta M, Bermudez M, Trovato O, De Neve J, Ballet S, Wolber G, Spetea M. Antinociceptive Efficacy of the µ-Opioid/Nociceptin Peptide-Based Hybrid KGNOP1 in Inflammatory Pain without Rewarding Effects in Mice: An Experimental Assessment and Molecular Docking. Molecules 2021; 26:3267. [PMID: 34071603 PMCID: PMC8198056 DOI: 10.3390/molecules26113267] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/18/2021] [Accepted: 05/26/2021] [Indexed: 01/09/2023] Open
Abstract
Opioids are the most effective analgesics, with most clinically available opioids being agonists to the µ-opioid receptor (MOR). The MOR is also responsible for their unwanted effects, including reward and opioid misuse leading to the current public health crisis. The imperative need for safer, non-addictive pain therapies drives the search for novel leads and new treatment strategies. In this study, the recently discovered MOR/nociceptin (NOP) receptor peptide hybrid KGNOP1 (H-Dmt-D-Arg-Aba-β-Ala-Arg-Tyr-Tyr-Arg-Ile-Lys-NH2) was evaluated following subcutaneous administration in mouse models of acute (formalin test) and chronic inflammatory pain (Complete Freund's adjuvant-induced paw hyperalgesia), liabilities of spontaneous locomotion, conditioned place preference, and the withdrawal syndrome. KGNOP1 demonstrated dose-dependent antinociceptive effects in the formalin test, and efficacy in attenuating thermal hyperalgesia with prolonged duration of action. Antinociceptive effects of KGNOP1 were reversed by naltrexone and SB-612111, indicating the involvement of both MOR and NOP receptor agonism. In comparison with morphine, KGNOP1 was more potent and effective in mouse models of inflammatory pain. Unlike morphine, KGNOP1 displayed reduced detrimental liabilities, as no locomotor impairment nor rewarding and withdrawal effects were observed. Docking of KGNOP1 to the MOR and NOP receptors and subsequent 3D interaction pattern analyses provided valuable insights into its binding mode. The mixed MOR/NOP receptor peptide KGNOP1 holds promise in the effort to develop new analgesics for the treatment of various pain states with fewer MOR-mediated side effects, particularly abuse and dependence liabilities.
Collapse
Affiliation(s)
- Maria Dumitrascuta
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (M.D.); (O.T.)
| | - Marcel Bermudez
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, D-14195 Berlin, Germany; (M.B.); (G.W.)
| | - Olga Trovato
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (M.D.); (O.T.)
| | - Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium; (J.D.N.); (S.B.)
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium; (J.D.N.); (S.B.)
| | - Gerhard Wolber
- Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2+4, D-14195 Berlin, Germany; (M.B.); (G.W.)
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria; (M.D.); (O.T.)
| |
Collapse
|
13
|
Lu JJ, Polgar WE, Mann A, Dasgupta P, Schulz S, Zaveri NT. Differential In Vitro Pharmacological Profiles of Structurally Diverse Nociceptin Receptor Agonists in Activating G Protein and Beta-Arrestin Signaling at the Human Nociceptin Opioid Receptor. Mol Pharmacol 2021; 100:7-18. [PMID: 33958480 DOI: 10.1124/molpharm.120.000076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 03/26/2021] [Indexed: 11/22/2022] Open
Abstract
Agonists at the nociceptin opioid peptide receptor (NOP) are under investigation as therapeutics for nonaddicting analgesia, opioid use disorder, Parkinson's disease, and other indications. NOP full and partial agonists have both been of interest, particularly since NOP partial agonists show a reduced propensity for behavioral disruption than NOP full agonists. Here, we investigated the in vitro pharmacological properties of chemically diverse NOP receptor agonists in assays measuring functional activation of the NOP receptor such as guanosine 5'-O-[gamma-thio]triphosphate (GTPγS) binding, cAMP inhibition, G protein-coupled inwardly rectifying potassium (GIRK) channel activation, phosphorylation, β-arrestin recruitment and receptor internalization. When normalized to the efficacy of the natural agonist nociceptin/orphanin FQ (N/OFQ), we found that different functional assays that measure intrinsic activity produce inconsistent levels of agonist efficacy, particularly for ligands that were partial agonists. Agonist efficacy obtained in the GTPγS assay tended to be lower than that in the cAMP and GIRK assays. These structurally diverse NOP agonists also showed differential receptor phosphorylation profiles at the phosphosites we examined and induced varying levels of receptor internalization. Interestingly, although the rank order for β-arrestin recruitment by these NOP agonists was consistent with their ability to induce receptor internalization, their phosphorylation signatures at the time point we investigated were not indicative of the levels of β-arrestin recruitment or internalization induced by these agonists. It is possible that other phosphorylation sites, yet to be identified, drive the recruitment of NOP receptor ensembles and subsequent receptor trafficking by some nonpeptide NOP agonists. These findings potentially help understand NOP agonist pharmacology in the context of ligand-activated receptor trafficking. SIGNIFICANCE STATEMENT: Chemically diverse agonist ligands at the nociceptin opioid receptor G protein-coupled receptor showed differential efficacy for activating downstream events after receptor binding, in a suite of functional assays measuring guanosine 5'-O-[gamma-thio]triphosphate binding, cAMP inhibition, G protein-coupled inwardly rectifying protein channel activation, β-arrestin recruitment, receptor internalization and receptor phosphorylation. These analyses provide a context for understanding nociceptin opioid peptide receptor (NOP) agonist pharmacology driven by ligand-induced differential NOP receptor signaling.
Collapse
Affiliation(s)
- James J Lu
- Astraea Therapeutics, Mountain View, California (J.J.L., W.E.P., N.T.Z.); and Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany (A.M., P.D., S.S.)
| | - Willma E Polgar
- Astraea Therapeutics, Mountain View, California (J.J.L., W.E.P., N.T.Z.); and Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany (A.M., P.D., S.S.)
| | - Anika Mann
- Astraea Therapeutics, Mountain View, California (J.J.L., W.E.P., N.T.Z.); and Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany (A.M., P.D., S.S.)
| | - Pooja Dasgupta
- Astraea Therapeutics, Mountain View, California (J.J.L., W.E.P., N.T.Z.); and Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany (A.M., P.D., S.S.)
| | - Stefan Schulz
- Astraea Therapeutics, Mountain View, California (J.J.L., W.E.P., N.T.Z.); and Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany (A.M., P.D., S.S.)
| | - Nurulain T Zaveri
- Astraea Therapeutics, Mountain View, California (J.J.L., W.E.P., N.T.Z.); and Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany (A.M., P.D., S.S.)
| |
Collapse
|
14
|
Kiguchi N, Kishioka S, Ko MC. [Therapeutic potentials of safe opioid analgesics targeting nociceptin/orphanin FQ peptide receptor]. Nihon Yakurigaku Zasshi 2021; 156:139-144. [PMID: 33952840 DOI: 10.1254/fpj.20106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
After the identification of nociceptin/orphanin FQ (N/OFQ) peptide (NOP) and its cognate receptor, the unique functional profiles of the N/OFQ-NOP receptor system have been uncovered. NOP receptors are distributed in the key regions that regulate pain and reward processing in the central nervous system. In non-human primates (NHPs), activation of the NOP receptor causes antinociception and anti-hypersensitivity via spinal and supraspinal effects. Moreover, activation of the NOP receptor attenuates dopaminergic transmission and potentiates mu-opioid peptide (MOP) receptor-mediated analgesia. Here, we highlight the functional profiles of bifunctional NOP and MOP receptor agonists based on their promising effects for the treatment of pain and drug abuse. Bifunctional NOP/MOP receptor "partial" agonists, such as AT-121, BU08028, and BU10038, exert potent analgesic effects without MOP receptor-related side effects such as abuse liability, respiratory depression, physical dependence, and itching in NHPs. These novel NOP/MOP receptor agonists reduce rewarding and the reinforcing effects of abused drugs. Furthermore, a mixed NOP/opioid receptor "full" agonist, cebranopadol, is undergoing several clinical trials, and the therapeutic advantage of the coactivation of NOP and MOP receptors has also been confirmed in humans. Therefore, this class of drugs that coactivate NOP and MOP receptors proposes a wide therapeutic range with fewer side effects, indicating a greater potential for the development of novel safer opioid analgesics.
Collapse
Affiliation(s)
| | - Shiroh Kishioka
- Faculty of Wakayama Health Care Sciences, Takarazuka University of Medical and Health Care
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest School of Medicine
| |
Collapse
|
15
|
Multifunctional Opioid-Derived Hybrids in Neuropathic Pain: Preclinical Evidence, Ideas and Challenges. Molecules 2020; 25:molecules25235520. [PMID: 33255641 PMCID: PMC7728063 DOI: 10.3390/molecules25235520] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022] Open
Abstract
When the first- and second-line therapeutics used to treat neuropathic pain (NP) fail to induce efficient analgesia—which is estimated to relate to more than half of the patients—opioid drugs are prescribed. Still, the pathological changes following the nerve tissue injury, i.a. pronociceptive neuropeptide systems activation, oppose the analgesic effects of opiates, enforcing the use of relatively high therapeutic doses in order to obtain satisfying pain relief. In parallel, the repeated use of opioid agonists is associated with burdensome adverse effects due to compensatory mechanisms that arise thereafter. Rational design of hybrid drugs, in which opioid ligands are combined with other pharmacophores that block the antiopioid action of pronociceptive systems, delivers the opportunity to ameliorate the NP-oriented opioid treatment via addressing neuropathological mechanisms shared both by NP and repeated exposition to opioids. Therewith, the new dually acting drugs, tailored for the specificity of NP, can gain in efficacy under nerve injury conditions and have an improved safety profile as compared to selective opioid agonists. The current review presents the latest ideas on opioid-comprising hybrid drugs designed to treat painful neuropathy, with focus on their biological action, as well as limitations and challenges related to this therapeutic approach.
Collapse
|
16
|
Bogacka J, Ciapała K, Pawlik K, Kwiatkowski K, Dobrogowski J, Przeklasa-Muszynska A, Mika J. CCR4 Antagonist (C021) Administration Diminishes Hypersensitivity and Enhances the Analgesic Potency of Morphine and Buprenorphine in a Mouse Model of Neuropathic Pain. Front Immunol 2020; 11:1241. [PMID: 32760393 PMCID: PMC7372009 DOI: 10.3389/fimmu.2020.01241] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022] Open
Abstract
Neuropathic pain is a chronic condition that remains a major clinical problem owing to high resistance to available therapy. Recent studies have indicated that chemokine signaling pathways are crucial in the development of painful neuropathy; however, the involvement of CC chemokine receptor 4 (CCR4) has not been fully elucidated thus far. Therefore, the aim of our research was to investigate the role of CCR4 in the development of tactile and thermal hypersensitivity, the effectiveness of morphine/buprenorphine, and opioid-induced tolerance in mice exposed to chronic constriction injury (CCI) of the sciatic nerve. The results of our research demonstrated that a single intrathecal or intraperitoneal administration of C021, a CCR4 antagonist, dose dependently diminished neuropathic pain-related behaviors in CCI-exposed mice. After sciatic nerve injury, the spinal expression of CCL17 and CCL22 remained unchanged in contrast to that of CCL2, which was significantly upregulated until day 14 after CCI. Importantly, our results provide evidence that in naive mice, CCL2 may evoke pain-related behaviors through CCR4 because its pronociceptive effects are diminished by C021. In CCI-exposed mice, the pharmacological blockade of CCR4 enhanced the analgesic properties of morphine/buprenorphine and delayed the development of morphine-induced tolerance, which was associated with the silencing of IBA-1 activation in cells and decrease in CCL2 production. The obtained data suggest that the pharmacological blockade of CCR4 may be a new potential therapeutic target for neuropathic pain polytherapy.
Collapse
Affiliation(s)
- Joanna Bogacka
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Katarzyna Ciapała
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Katarzyna Pawlik
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Klaudia Kwiatkowski
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Jan Dobrogowski
- Department of Pain Research and Treatment, Chair of Anesthesiology and Intensive Therapy, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Przeklasa-Muszynska
- Department of Pain Research and Treatment, Chair of Anesthesiology and Intensive Therapy, Jagiellonian University Medical College, Krakow, Poland
| | - Joanna Mika
- Department of Pain Pharmacology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
17
|
Kiguchi N, Ding H, Ko MC. Therapeutic potentials of NOP and MOP receptor coactivation for the treatment of pain and opioid abuse. J Neurosci Res 2020; 100:191-202. [PMID: 32255240 DOI: 10.1002/jnr.24624] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/26/2020] [Accepted: 03/17/2020] [Indexed: 12/21/2022]
Abstract
Following the identification of the nociceptin/orphanin FQ (N/OFQ) peptide (NOP) as an endogenous ligand for the NOP receptor, ample evidence has revealed unique functional profiles of the N/OFQ-NOP receptor system. NOP receptors are expressed in key neural substrates involved in pain and reward modulation. In nonhuman primates (NHPs), NOP receptor activation effectively exerts antinociception and anti-hypersensitivity at the spinal and supraspinal levels. Moreover, NOP receptor activation inhibits dopaminergic transmission and synergistically enhances mu-opioid peptide (MOP) receptor-mediated analgesia. In this article, we have discussed the functional profiles of ligands with dual NOP and MOP receptor agonist activities and highlight their optimal functional efficacy for pain relief and drug abuse treatment. Through coactivation of NOP and MOP receptors, bifunctional NOP/MOP receptor "partial" agonists (e.g., AT-121, BU08028, and BU10038) reveal a wider therapeutic window with fewer side effects. These newly developed ligands potently induce antinociception without MOP receptor agonist-associated side effects such as abuse potential, respiratory depression, itching sensation, and physical dependence. In addition, in both rodent and NHP models, bifunctional NOP/MOP receptor agonists can attenuate reward processing and/or the reinforcing effects of opioids and other abused drugs. While a mixed NOP/opioid receptor "full" agonist cebranopadol is undergoing clinical trials, bifunctional NOP/MOP "partial" agonists exhibit promising therapeutic profiles in translational NHP models for the treatment of pain and opioid abuse. This class of drugs demonstrates the therapeutic advantage of NOP and MOP receptor coactivation, indicating a greater potential for future development.
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, USA
| | - Mei-Chuan Ko
- Department of Physiology & Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,W.G. Hefner Veterans Affairs Medical Center, Salisbury, NC, USA
| |
Collapse
|
18
|
Pacifico S, Albanese V, Illuminati D, Fantinati A, Marzola E, Ferrari F, Neto JA, Sturaro C, Ruzza C, Calò G, Preti D, Guerrini R. Tetrabranched Hetero-Conjugated Peptides as Bifunctional Agonists of the NOP and Mu Opioid Receptors. Bioconjug Chem 2019; 30:2444-2451. [DOI: 10.1021/acs.bioconjchem.9b00519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Salvatore Pacifico
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, Ferrara 44121, Italy
| | - Valentina Albanese
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, Ferrara 44121, Italy
| | - Davide Illuminati
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, Ferrara 44121, Italy
| | - Anna Fantinati
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, Ferrara 44121, Italy
| | - Erika Marzola
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, Ferrara 44121, Italy
| | - Federica Ferrari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara Via Fossato di Mortara 17/19, Ferrara 44121, Italy
| | - Joaquim Azevedo Neto
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara Via Fossato di Mortara 17/19, Ferrara 44121, Italy
| | - Chiara Sturaro
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara Via Fossato di Mortara 17/19, Ferrara 44121, Italy
| | - Chiara Ruzza
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara Via Fossato di Mortara 17/19, Ferrara 44121, Italy
| | - Girolamo Calò
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara Via Fossato di Mortara 17/19, Ferrara 44121, Italy
| | - Delia Preti
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, Ferrara 44121, Italy
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Luigi Borsari 46, Ferrara 44121, Italy
| |
Collapse
|
19
|
Lambert DG. Mixed mu-nociceptin/orphanin FQ opioid receptor agonists and the search for the analgesic holy grail. Br J Anaesth 2019; 122:e95-e97. [DOI: 10.1016/j.bja.2019.02.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/12/2019] [Accepted: 02/20/2019] [Indexed: 11/30/2022] Open
|
20
|
Mann A, Moulédous L, Froment C, O'Neill PR, Dasgupta P, Günther T, Brunori G, Kieffer BL, Toll L, Bruchas MR, Zaveri NT, Schulz S. Agonist-selective NOP receptor phosphorylation correlates in vitro and in vivo and reveals differential post-activation signaling by chemically diverse agonists. Sci Signal 2019; 12:12/574/eaau8072. [PMID: 30914485 DOI: 10.1126/scisignal.aau8072] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Agonists of the nociceptin/orphanin FQ opioid peptide (NOP) receptor, a member of the opioid receptor family, are under active investigation as novel analgesics, but their modes of signaling are less well characterized than those of other members of the opioid receptor family. Therefore, we investigated whether different NOP receptor ligands showed differential signaling or functional selectivity at the NOP receptor. Using newly developed phosphosite-specific antibodies to the NOP receptor, we found that agonist-induced NOP receptor phosphorylation occurred primarily at four carboxyl-terminal serine (Ser) and threonine (Thr) residues, namely, Ser346, Ser351, Thr362, and Ser363, and proceeded with a temporal hierarchy, with Ser346 as the first site of phosphorylation. G protein-coupled receptor kinases 2 and 3 (GRK2/3) cooperated during agonist-induced phosphorylation, which, in turn, facilitated NOP receptor desensitization and internalization. A comparison of structurally distinct NOP receptor agonists revealed dissociation in functional efficacies between G protein-dependent signaling and receptor phosphorylation. Furthermore, in NOP-eGFP and NOP-eYFP mice, NOP receptor agonists induced multisite phosphorylation and internalization in a dose-dependent and agonist-selective manner that could be blocked by specific antagonists. Our study provides new tools to study ligand-activated NOP receptor signaling in vitro and in vivo. Differential agonist-selective NOP receptor phosphorylation by chemically diverse NOP receptor agonists suggests that differential signaling by NOP receptor agonists may play a role in NOP receptor ligand pharmacology.
Collapse
Affiliation(s)
- Anika Mann
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany.
| | - Lionel Moulédous
- Research Center on Animal Cognition, Center for Integrative Biology, Toulouse University, CNRS, UPS, 31062 Toulouse Cedex 09, France
| | - Carine Froment
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, 31077 Toulouse Cedex 04, France
| | - Patrick R O'Neill
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Pooja Dasgupta
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany
| | - Thomas Günther
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany
| | - Gloria Brunori
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Brigitte L Kieffer
- Douglas Research Center, Department of Psychiatry, Faculty of Medicine, McGill University, Montreal, QC H3A 1A1, Canada
| | - Lawrence Toll
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Michael R Bruchas
- Center for the Neurobiology of Addiction, Pain, and Emotion, Departments of Anesthesiology and Pharmacology, University of Washington, Seattle, WA 98195, USA
| | | | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Drackendorfer Str. 1, Jena 07747, Germany.
| |
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
|
Abstract
The nociceptin/orphanin FQ peptide (NOP) receptor-related ligands have been demonstrated in preclinical studies for several therapeutic applications. This article highlights (1) how nonhuman primates (NHP) were used to facilitate the development and application of positron emission tomography tracers in humans; (2) effects of an endogenous NOP ligand, nociceptin/orphanin FQ, and its interaction with mu opioid peptide (MOP) receptor agonists; and (3) promising functional profiles of NOP-related agonists in NHP as analgesics and treatment for substance use disorders. NHP models offer the most phylogenetically appropriate evaluation of opioid and non-opioid receptor functions and drug effects. Based on preclinical and clinical data of ligands with mixed NOP/MOP receptor agonist activity, several factors including their intrinsic efficacies for activating NOP versus MOP receptors and different study endpoints in NHP could contribute to different pharmacological profiles. Ample evidence from NHP studies indicates that bifunctional NOP/MOP receptor agonists have opened an exciting avenue for developing safe, effective medications with fewer side effects for treating pain and drug addiction. In particular, bifunctional NOP/MOP partial agonists hold a great potential as (1) effective spinal analgesics without itch side effects; (2) safe, nonaddictive analgesics without opioid side effects such as respiratory depression; and (3) effective medications for substance use disorders.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
23
|
Calo G, Lambert DG. Nociceptin/orphanin FQ receptor ligands and translational challenges: focus on cebranopadol as an innovative analgesic. Br J Anaesth 2018; 121:1105-1114. [PMID: 30336855 PMCID: PMC6208290 DOI: 10.1016/j.bja.2018.06.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/18/2018] [Accepted: 07/09/2018] [Indexed: 12/17/2022] Open
Abstract
Opioids are characterised as classical (mu, delta, and kappa) along with the non-classical nociceptin/orphanin FQ (N/OFQ) receptor or NOP. Targeting NOP has therapeutic indications in control of the cardiovascular and respiratory systems and micturition, and a profile as an antidepressant. For all of these indications, there are translational human data. Opioids such as morphine and fentanyl (activating the mu receptor) are the mainstay of pain treatment in the perioperative period, despite a challenging side-effect profile. Opioids in general have poor efficacy in neuropathic pain. Moreover, longer term use is associated with tolerance. There is good evidence interactions between opioid receptors, and receptor co-activation can reduce side-effects without compromising analgesia; this is particularly true for mu and NOP co-activation. Recent pharmaceutical development has produced a mixed opioid/NOP agonist, cebranopadol. This new chemical entity is effective in animal models of nociceptive and neuropathic pain with greater efficacy in the latter. In animal models, there is little evidence for respiratory depression, and tolerance (compared with morphine) only develops after long treatment periods. There is now early phase clinical development in diabetic neuropathy, cancer pain, and low back pain where cebranopadol displays significant efficacy. In 1996, N/OFQ was formally identified with an innovative analgesic profile. Approximately 20 yr later, cebranopadol as a clinical ligand is advancing through the human trials process.
Collapse
Affiliation(s)
- G Calo
- Section of Pharmacology, Department of Medical Sciences, National Institute of Neurosciences, University of Ferrara, Ferrara, Italy.
| | - D G Lambert
- Department of Cardiovascular Sciences, University of Leicester, Anaesthesia, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester, UK
| |
Collapse
|
24
|
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: 0.4] [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
|
25
|
Ali A, Arif AW, Bhan C, Kumar D, Malik MB, Sayyed Z, Akhtar KH, Ahmad MQ. Managing Chronic Pain in the Elderly: An Overview of the Recent Therapeutic Advancements. Cureus 2018; 10:e3293. [PMID: 30443463 PMCID: PMC6235641 DOI: 10.7759/cureus.3293] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
A majority of the elderly suffer from chronic pain that significantly alters their daily activities and imposes an enormous burden on health care. Multiple comorbidities and the risk of polypharmacy in the elderly make it a challenge to determine the appropriate drug, dosage, and maintenance of therapy. Opioids are the most commonly used agents for this purpose in the elderly. The aim of this article is to discuss both the current well-established therapies used for managing chronic pain in the elderly and also the emerging newer therapies.
Collapse
Affiliation(s)
- Asad Ali
- Medicine, CMH Lahore Medical College and Institute of Dentistry, Lahore, PAK
| | | | - Chandur Bhan
- Internal Medicine, Chandka Medical College Hospital, Larkana, PAK
| | - Deepak Kumar
- Internal Medicine, Jinnah Postgraduate Medical Centre, Karachi, PAK
| | | | - Zohaib Sayyed
- Pediatrics, Shaikh Khalifa Bin Zayed Al-Nahyan Medical and Dental College, Bahawalpur, PAK
| | | | | |
Collapse
|
26
|
Ding H, Kiguchi N, Yasuda D, Daga PR, Polgar WE, Lu JJ, Czoty PW, Kishioka S, Zaveri NT, Ko MC. A bifunctional nociceptin and mu opioid receptor agonist is analgesic without opioid side effects in nonhuman primates. Sci Transl Med 2018; 10:eaar3483. [PMID: 30158150 PMCID: PMC6295194 DOI: 10.1126/scitranslmed.aar3483] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/07/2018] [Accepted: 08/09/2018] [Indexed: 11/02/2022]
Abstract
Misuse of prescription opioids, opioid addiction, and overdose underscore the urgent need for developing addiction-free effective medications for treating severe pain. Mu opioid peptide (MOP) receptor agonists provide very effective pain relief. However, severe side effects limit their use in the clinical setting. Agonists of the nociceptin/orphanin FQ peptide (NOP) receptor have been shown to modulate the antinociceptive and reinforcing effects of MOP agonists. We report the discovery and development of a bifunctional NOP/MOP receptor agonist, AT-121, which has partial agonist activity at both NOP and MOP receptors. AT-121 suppressed oxycodone's reinforcing effects and exerted morphine-like analgesic effects in nonhuman primates. AT-121 treatment did not induce side effects commonly associated with opioids, such as respiratory depression, abuse potential, opioid-induced hyperalgesia, and physical dependence. Our results in nonhuman primates suggest that bifunctional NOP/MOP agonists with the appropriate balance of NOP and MOP agonist activity may provide a dual therapeutic action for safe and effective pain relief and treating prescription opioid abuse.
Collapse
Affiliation(s)
- Huiping Ding
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Norikazu Kiguchi
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | | | | | | | - James J Lu
- Astraea Therapeutics, Mountain View, CA 94043, USA
| | - Paul W Czoty
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Shiroh Kishioka
- Department of Pharmacology, Wakayama Medical University, Wakayama, Japan
| | | | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
- W.G. Hefner Veterans Affairs Medical Center, Salisbury, NC 28144, USA
| |
Collapse
|
27
|
Wu Q, Liu L. ORL 1 Activation Mediates a Novel ORL 1 Receptor Agonist SCH221510 Analgesia in Neuropathic Pain in Rats. J Mol Neurosci 2018; 66:10-16. [PMID: 30074175 DOI: 10.1007/s12031-018-1140-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/25/2018] [Indexed: 01/24/2023]
Abstract
Opioid receptor like 1 (ORL1) receptor activation displayed an anti-nociceptive effect at spinal level for acute and neuropathic pain. SCH221510, an orally active non-peptide ORL1 agonist, was reported to be effective in treating neuropathic pain. The present study used ORL1 antagonist and siRNA to investigate that ORL1 activation mediates intrathecal SCH221510 analgesia in neuropathic pain induced by chronic constrictive injury (CCI) to rat sciatic nerve. Paw withdrawal latency and 50% mechanical threshold were measured for thermal and mechanical hypersensitivity in rats. CCI significantly decreased paw withdrawal latency and mechanical threshold. SCH221510 (3, 10, 30 μg) or ORL1 antagonist ([Nphe1]nociceptin(1-13)NH2, 10 μg) was intrathecally injected to test the behavioral effects on neuropathic pain. Intrathecal siRNA was started on 1 day before CCI surgery and maintained for 7 days. L4-L5 spinal cord ORL1 mRNA and protein were measured by real-time PCR and Western blot. The effect of intrathecal siRNA on SCH2210510 was tested in CCI rats on day 7. Intrathecal SCH221510 dose-dependently reduced thermal and mechanical hypersensitivity induced by CCI. [Nphe1]nociceptin(1-13)NH2 blocked SCH221510 analgesia in CCI rats. Intrathecal siRNA blocked ORL1 mRNA and protein increase induced by CCI. Intrathecal ORL1 siRNA did not change thermal and mechanical hypersensitivity induced by nerve injury. Intrathecal siRNA blocked SCH221510 analgesia in neuropathic pain at spinal level. Conclusively, ORL1 activation mediates SCH221510 analgesia in neuropathic pain at spinal level. The results warrant a potential clinically applicable drug in treating neuropathic pain.
Collapse
Affiliation(s)
- Qiang Wu
- Department of Neurology, Xinxiang Central Hospital, 56 Jin Hui Da Street, Xinxiang, 453000, Henan, China.
| | - Li Liu
- Department of Neurology, Luoyang First People's Hospital, Luoyang, 471000, Henan, China
| |
Collapse
|
28
|
Günther T, Dasgupta P, Mann A, Miess E, Kliewer A, Fritzwanker S, Steinborn R, Schulz S. Targeting multiple opioid receptors - improved analgesics with reduced side effects? Br J Pharmacol 2018; 175:2857-2868. [PMID: 28378462 PMCID: PMC6016677 DOI: 10.1111/bph.13809] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 02/24/2017] [Accepted: 03/15/2017] [Indexed: 01/12/2023] Open
Abstract
Classical opioid analgesics, including morphine, mediate all of their desired and undesired effects by specific activation of the μ-opioid receptor (μ receptor). The use of morphine for treating chronic pain, however, is limited by the development of constipation, respiratory depression, tolerance and dependence. Analgesic effects can also be mediated through other members of the opioid receptor family such as the κ-opioid receptor (κ receptor), δ-opioid receptor (δ receptor) and the nociceptin/orphanin FQ peptide receptor (NOP receptor). Currently, a new generation of opioid analgesics is being developed that can simultaneously bind with high affinity to multiple opioid receptors. With this new action profile, it is hoped that additional analgesic effects and fewer side effects can be achieved. Recent research is mainly focused on the development of bifunctional μ/NOP receptor agonists, which has already led to novel lead structures such as the spiroindole-based cebranopadol and a compound class with a piperidin-4-yl-1,3-dihydroindol-2-one backbone (SR16835/AT-202 and SR14150/AT-200). In addition, the ornivol BU08028 is an analogue of the clinically well-established buprenorphine. Moreover, the morphinan-based nalfurafine exerts its effect with a dominant κ receptor-component and is therefore utilized in the treatment of pruritus. The very potent dihydroetorphine is a true multi-receptor opioid ligand in that it binds to μ, κ and δ receptors. The main focus of this review is to assess the paradigm of opioid ligands targeting multiple receptors with a single chemical entity. We reflect on this rationale by discussing the biological actions of particular multi-opioid receptor ligands, but not on their medicinal chemistry and design. LINKED ARTICLES This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.
Collapse
Affiliation(s)
- Thomas Günther
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich‐Schiller‐UniversityJenaGermany
| | - Pooja Dasgupta
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich‐Schiller‐UniversityJenaGermany
| | - Anika Mann
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich‐Schiller‐UniversityJenaGermany
| | - Elke Miess
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich‐Schiller‐UniversityJenaGermany
| | - Andrea Kliewer
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich‐Schiller‐UniversityJenaGermany
| | - Sebastian Fritzwanker
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich‐Schiller‐UniversityJenaGermany
| | - Ralph Steinborn
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich‐Schiller‐UniversityJenaGermany
| | - Stefan Schulz
- Institute of Pharmacology and ToxicologyJena University Hospital, Friedrich‐Schiller‐UniversityJenaGermany
| |
Collapse
|
29
|
Sałat K, Furgała A, Sałat R. Evaluation of cebranopadol, a dually acting nociceptin/orphanin FQ and opioid receptor agonist in mouse models of acute, tonic, and chemotherapy-induced neuropathic pain. Inflammopharmacology 2018; 26:361-374. [PMID: 29071457 PMCID: PMC5859690 DOI: 10.1007/s10787-017-0405-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/06/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Cebranopadol (a.k.a. GRT-6005) is a dually acting nociceptin/orphanin FQ and opioid receptor agonist that has been recently developed in Phase 2 clinical trials for painful diabetic neuropathy or cancer pain. It also showed analgesic properties in various rat models of pain and had a better safety profile as compared to equi-analgesic doses of morphine. Since antinociceptive properties of cebranopadol have been studied mainly in rat models, in the present study, we assessed analgesic activity of subcutaneous cebranopadol (10 mg/kg) in various mouse pain models. METHODS We used models of acute, tonic, and chronic pain induced by thermal and chemical stimuli, with a particular emphasis on pharmacoresistant chronic neuropathic pain evoked by oxaliplatin in which cebranopadol was used alone or in combination with simvastatin. KEY RESULTS As shown in the hot plate test, the analgesic activity of cebranopadol developed more slowly as compared to morphine (90-120 min vs. 60 min). Cebranopadol displayed a significant antinociceptive activity in acute pain models, i.e., the hot plate, writhing, and capsaicin tests. It attenuated nocifensive responses in both phases of the formalin test and reduced cold allodynia in oxaliplatin-induced neuropathic pain model. Its efficacy was similar to that of morphine. Used in combination and administered simultaneously, 4 or 6 h after simvastatin, cebranopadol did not potentiate antiallodynic activity of this cholesterol-lowering drug. Cebranopadol did not induce any motor deficits in the rotarod test. CONCLUSION Cebranopadol may have significant potential for the treatment of various pain types, including inflammatory and chemotherapy-induced neuropathic pain.
Collapse
Affiliation(s)
- Kinga Sałat
- Chair of Pharmacodynamics, Department of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St, 30-688, Krakow, Poland.
| | - Anna Furgała
- Chair of Pharmacodynamics, Department of Pharmacodynamics, Jagiellonian University Medical College, 9 Medyczna St, 30-688, Krakow, Poland
| | - Robert Sałat
- Faculty of Production Engineering, Warsaw University of Life Sciences, 164 Nowoursynowska St, 02-787, Warsaw, Poland
| |
Collapse
|
30
|
Kallupi M, Shen Q, de Guglielmo G, Yasuda D, Journigan VB, Zaveri NT, Ciccocioppo R. Buprenorphine requires concomitant activation of NOP and MOP receptors to reduce cocaine consumption. Addict Biol 2018. [PMID: 28635181 DOI: 10.1111/adb.12513] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Buprenorphine's clinical use is approved for the treatment of heroin addiction; however, evidence supporting its efficacy in cocaine abuse also exists. While for heroin it has been demonstrated that the effect of buprenorphine is mediated by its ability to activate μ-opioid peptide receptor (MOP) receptors, the mechanism through which it attenuates cocaine intake remains elusive. We explored this mechanism using operant models where rodents were trained to chronically self-administer cocaine for 2 hours daily. Buprenorphine (0.3, 1.0 and 3.0 mg/kg) given intraperitoneally 90 minutes before access to cocaine significantly and dose dependently reduced its intake. Pre-treatment with naltrexone or with the selective nociceptin/orphanin FQ peptide (NOP) antagonist SB-612111 did not prevent buprenorphine-induced reduction of cocaine intake. However, when naltrexone and SB-612111 were combined, the effect of buprenorphine on cocaine was completely prevented. To confirm that co-activation of MOP and NOP receptors is the underlying mechanism through which buprenorphine reduces cocaine intake, three compounds, namely, AT-034, AT-201 and AT-202, with a range of affinity and intrinsic activity profiles for MOP and NOP receptors, but weak ability for kappa-opioid peptide receptor (KOP) transmission, were tested. Consistent with our hypothesis based on buprenorphine's effects, results demonstrated that AT-034 and AT-201, which co-activate MOP and NOP receptors, reduced cocaine self-administration like buprenorphine. AT-202, which selectively stimulates NOP receptors, was not effective. Together, these data demonstrate that for buprenorphine, co-activation of MOP and NOP receptors is essential to reduce cocaine consumption. These results open new vistas on the treatment of cocaine addiction by developing compounds with mixed MOP/NOP agonist properties.
Collapse
Affiliation(s)
- Marsida Kallupi
- School of Pharmacy, Pharmacology Unit; University of Camerino; Italy
- Committee on the Neurobiology of Addictive Disorders; The Scripps Research Institute; La Jolla CA USA
| | - Qianwei Shen
- School of Pharmacy, Pharmacology Unit; University of Camerino; Italy
| | - Giordano de Guglielmo
- School of Pharmacy, Pharmacology Unit; University of Camerino; Italy
- Committee on the Neurobiology of Addictive Disorders; The Scripps Research Institute; La Jolla CA USA
| | | | | | | | | |
Collapse
|
31
|
Maldonado R, Baños JE, Cabañero D. Usefulness of knockout mice to clarify the role of the opioid system in chronic pain. Br J Pharmacol 2018; 175:2791-2808. [PMID: 29124744 DOI: 10.1111/bph.14088] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 12/29/2022] Open
Abstract
Several lines of knockout mice deficient in the genes encoding each component of the endogenous opioid system have been used for decades to clarify the specific role of the different opioid receptors and peptide precursors in many physiopathological conditions. The use of these genetically modified mice has improved our knowledge of the specific involvement of each endogenous opioid component in nociceptive transmission during acute and chronic pain conditions. The present review summarizes the recent advances obtained using these genetic tools in understanding the role of the opioid system in the pathophysiological mechanisms underlying chronic pain. Behavioural data obtained in these chronic pain models are discussed considering the peculiarities of the behavioural phenotype of each line of knockout mice. These studies have identified the crucial role of specific components of the opioid system in different manifestations of chronic pain and have also opened new possible therapeutic approaches, such as the development of opioid compounds simultaneously targeting several opioid receptors. However, several questions still remain open and require further experimental effort to be clarified. The novel genetic tools now available to manipulate specific neuronal populations and precise genome editing in mice will facilitate in a near future the elucidation of the role of each component of the endogenous opioid system in chronic pain. LINKED ARTICLES This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.
Collapse
Affiliation(s)
- Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Josep Eladi Baños
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
32
|
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
|
33
|
|
34
|
Imam MZ, Kuo A, Ghassabian S, Smith MT. Progress in understanding mechanisms of opioid-induced gastrointestinal adverse effects and respiratory depression. Neuropharmacology 2017; 131:238-255. [PMID: 29273520 DOI: 10.1016/j.neuropharm.2017.12.032] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 12/18/2017] [Accepted: 12/19/2017] [Indexed: 02/06/2023]
Abstract
Opioids evoke analgesia through activation of opioid receptors (predominantly the μ opioid receptor) in the central nervous system. Opioid receptors are abundant in multiple regions of the central nervous system and the peripheral nervous system including enteric neurons. Opioid-related adverse effects such as constipation, nausea, and vomiting pose challenges for compliance and continuation of the therapy for chronic pain management. In the post-operative setting opioid-induced depression of respiration can be fatal. These critical limitations warrant a better understanding of their underpinning cellular and molecular mechanisms to inform the design of novel opioid analgesic molecules that are devoid of these unwanted side-effects. Research efforts on opioid receptor signalling in the past decade suggest that differential signalling pathways and downstream molecules preferentially mediate distinct pharmacological effects. Additionally, interaction among opioid receptors and, between opioid receptor and non-opioid receptors to form signalling complexes shows that opioid-induced receptor signalling is potentially more complicated than previously thought. This complexity provides an opportunity to identify and probe relationships between selective signalling pathway specificity and in vivo production of opioid-related adverse effects. In this review, we focus on current knowledge of the mechanisms thought to transduce opioid-induced gastrointestinal adverse effects (constipation, nausea, vomiting) and respiratory depression.
Collapse
Affiliation(s)
- Mohammad Zafar Imam
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia; UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Andy Kuo
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Sussan Ghassabian
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Maree T Smith
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia; School of Pharmacy, Faculty of Health and Behavioural Sciences, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
35
|
Zhang R, Xu B, Zhang MN, Zhang T, Wang ZL, Zhao G, Zhao GH, Li N, Fang Q, Wang R. Peripheral and central sites of action for anti-allodynic activity induced by the bifunctional opioid/NPFF receptors agonist BN-9 in inflammatory pain model. Eur J Pharmacol 2017; 813:122-129. [DOI: 10.1016/j.ejphar.2017.07.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 07/04/2017] [Accepted: 07/26/2017] [Indexed: 10/19/2022]
|
36
|
Palmisano M, Mercatelli D, Caputi FF, Carretta D, Romualdi P, Candeletti S. N/OFQ system in brain areas of nerve-injured mice: its role in different aspects of neuropathic pain. GENES, BRAIN, AND BEHAVIOR 2017; 16:537-545. [PMID: 28000999 DOI: 10.1111/gbb.12365] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/15/2016] [Accepted: 12/18/2016] [Indexed: 12/28/2022]
Abstract
Several studies showed that chronic pain causes reorganization and functional alterations of supraspinal brain regions. The nociceptin-NOP receptor system is one of the major systems involved in pain control and much evidence also suggested its implication in stress, anxiety and depression. Therefore, we investigated the nociceptin-NOP system alterations in selected brain regions in a neuropathic pain murine model. Fourteen days after the common sciatic nerve ligature, polymerase chain reaction (PCR) analysis indicated a significant decrease of pronociceptin and NOP receptor mRNA levels in the thalamus; these alterations could contribute to the decrease of the thalamic inhibitory function reported in neuropathic pain condition. Nociceptin peptide and NOP mRNA increased in the anterior cingulate cortex (ACC) and not in the somatosensory cortex, suggesting a peculiar involvement of this system in pain regulating circuitry. Similarly to the ACC, an increase of nociceptin peptide levels was observed in the amygdala. Finally, the pronociceptin and NOP mRNAs decrease observed in the hypothalamus reflects the lack of hypothalamus-pituitary-adrenal axis activation, already reported in neuropathic pain models. Our data indicate that neuropathic pain conditions affect the supraspinal nociceptin-NOP system which is also altered in regions known to play a role in emotional aspects of pain.
Collapse
Affiliation(s)
- M Palmisano
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - D Mercatelli
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - F F Caputi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - D Carretta
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - P Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - S Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
37
|
Anand P, Yiangou Y, Anand U, Mukerji G, Sinisi M, Fox M, McQuillan A, Quick T, Korchev YE, Hein P. Nociceptin/orphanin FQ receptor expression in clinical pain disorders and functional effects in cultured neurons. Pain 2016; 157:1960-1969. [PMID: 27127846 DOI: 10.1097/j.pain.0000000000000597] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The nociceptin/orphanin FQ peptide receptor (NOP), activated by its endogenous peptide ligand nociceptin/orphanin FQ (N/OFQ), exerts several effects including modulation of pain signalling. We have examined, for the first time, the tissue distribution of the NOP receptor in clinical visceral and somatic pain disorders by immunohistochemistry and assessed functional effects of NOP and μ-opioid receptor activation in cultured human and rat dorsal root ganglion (DRG) neurons. Quantification of NOP-positive nerve fibres within the bladder suburothelium revealed a remarkable several-fold increase in detrusor overactivity (P < 0.0001) and painful bladder syndrome patient specimens (P = 0.0014) compared with controls. In postmortem control human DRG, 75% to 80% of small/medium neurons (≤50 μm diameter) in the lumbar (somatic) and sacral (visceral) DRG were positive for NOP, and fewer large neurons; avulsion-injured cervical human DRG neurons showed similar numbers. NOP immunoreactivity was significantly decreased in injured peripheral nerves (P = 0.0004), and also in painful neuromas (P = 0.025). Calcium-imaging studies in cultured rat DRG neurons demonstrated dose-dependent inhibition of capsaicin responses in the presence of N/OFQ, with an IC50 of 8.6 pM. In cultured human DRG neurons, 32% inhibition of capsaicin responses was observed in the presence of 1 pM N/OFQ (P < 0.001). The maximum inhibition of capsaicin responses was greater with N/OFQ than μ-opioid receptor agonist DAMGO. Our findings highlight the potential of NOP agonists, particularly in urinary bladder overactivity and pain syndromes. The regulation of NOP expression in visceral and somatic sensory neurons by target-derived neurotrophic factors deserves further study, and the efficacy of NOP selective agonists in clinical trials.
Collapse
Affiliation(s)
- Praveen Anand
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Yiangos Yiangou
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Uma Anand
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Gaurav Mukerji
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Marco Sinisi
- Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| | - Michael Fox
- Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| | - Anthony McQuillan
- Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| | - Tom Quick
- Peripheral Nerve Injury Unit, Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| | - Yuri E Korchev
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Peter Hein
- Grünenthal Innovation, Translational Science and Strategy, Grünenthal GmbH, Aachen, Germany
| |
Collapse
|
38
|
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: 8.9] [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
|
39
|
Bird MF, Cerlesi MC, Brown M, Malfacini D, Vezzi V, Molinari P, Micheli L, Mannelli LDC, Ghelardini C, Guerrini R, Calò G, Lambert DG. Characterisation of the Novel Mixed Mu-NOP Peptide Ligand Dermorphin-N/OFQ (DeNo). PLoS One 2016; 11:e0156897. [PMID: 27272042 PMCID: PMC4896453 DOI: 10.1371/journal.pone.0156897] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/20/2016] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Opioid receptors are currently classified as Mu (μ), Delta (δ), Kappa (κ) plus the opioid related nociceptin/orphanin FQ (N/OFQ) peptide receptor (NOP). Despite compelling evidence for interactions and benefits of targeting more than one receptor type in producing analgesia, clinical ligands are Mu agonists. In this study we have designed a Mu-NOP agonist named DeNo. The Mu agonist component is provided by dermorphin, a peptide isolated from the skin of Phyllomedusa frogs and the NOP component by the endogenous agonist N/OFQ. METHODS We have assessed receptor binding profile of DeNo and compared with dermorphin and N/OFQ. In a series of functional screens we have assessed the ability to (i) increase Ca2+ in cells coexpressing recombinant receptors and a the chimeric protein Gαqi5, (ii) stimulate the binding of GTPγ[35S], (iii) inhibit cAMP formation, (iv) activate MAPKinase, (v) stimulate receptor-G protein and arrestin interaction using BRET, (vi) electrically stimulated guinea pig ileum (gpI) assay and (vii) ability to produce analgesia via the intrathecal route in rats. RESULTS DeNo bound to Mu (pKi; 9.55) and NOP (pKi; 10.22) and with reasonable selectivity. This translated to increased Ca2+ in Gαqi5 expressing cells (pEC50 Mu 7.17; NOP 9.69), increased binding of GTPγ[35S] (pEC50 Mu 7.70; NOP 9.50) and receptor-G protein interaction in BRET (pEC50 Mu 8.01; NOP 9.02). cAMP formation was inhibited and arrestin was activated (pEC50 Mu 6.36; NOP 8.19). For MAPK DeNo activated p38 and ERK1/2 at Mu but only ERK1/2 at NOP. In the gpI DeNO inhibited electrically-evoked contractions (pEC50 8.63) that was sensitive to both Mu and NOP antagonists. DeNo was antinociceptive in rats. CONCLUSION Collectively these data validate the strategy used to create a novel bivalent Mu-NOP peptide agonist by combining dermorphin (Mu) and N/OFQ (NOP). This molecule behaves essentially as the parent compounds in vitro. In the antonocicoeptive assays employed in this study DeNo displays only weak antinociceptive properties.
Collapse
MESH Headings
- Animals
- CHO Cells
- Calcium/metabolism
- Cricetulus
- Guinea Pigs
- HEK293 Cells
- Humans
- Male
- Opioid Peptides/chemistry
- Peptides/chemical synthesis
- Peptides/chemistry
- Peptides/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid/agonists
- Receptors, Opioid/chemistry
- Receptors, Opioid/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/chemistry
- Receptors, Opioid, mu/metabolism
- Nociceptin Receptor
Collapse
Affiliation(s)
- Mark F. Bird
- Department of Cardiovascular Sciences, University of Leicester, Division of Anaesthesia, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester, LE2 7LX, United Kingdom
| | - Maria Camilla Cerlesi
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Mark Brown
- Department of Cardiovascular Sciences, University of Leicester, Division of Anaesthesia, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester, LE2 7LX, United Kingdom
| | - Davide Malfacini
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - Vanessa Vezzi
- Department of Pharmacology, Istituto Superiore di Sanità, Rome, 00161, Italy
| | - Paola Molinari
- Department of Pharmacology, Istituto Superiore di Sanità, Rome, 00161, Italy
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health—Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health—Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health—Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Ferrara, Italy
| | - Girolamo Calò
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - David G. Lambert
- Department of Cardiovascular Sciences, University of Leicester, Division of Anaesthesia, Critical Care and Pain Management, Leicester Royal Infirmary, Leicester, LE2 7LX, United Kingdom
- * E-mail:
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW This article aims to discuss the multitarget concept for opioid receptor ligands framed on early observations that activating MOP (mu:μ) receptor whilst simultaneously blocking DOP (delta:δ) receptors reduces the onset of morphine tolerance. The review period is ostensibly calendar year 2014 but the new work in 2013 is also covered. RECENT FINDINGS Two molecules of interest with MOP agonist/DOP agonist and MOP agonist/DOP antagonist profiles were described: Rv-Jim-C3 and 3-[(2R,6R,11R)-8-hydroxy-6,11-dimethyl-1,4,5,6-tetrahydro-2,6-methano-3-benzazocin-3(2H)-yl]-N-phenylpropanamide (LP1), respectively. Both were effective in neuropathic pain (wherein classical single target opioids have low efficacy) with the latter having a predicted reduced tolerance profile. BU0807 is a buprenorphine derivative with mixed MOP/NOP agonist activity and this was shown to be effective in abdominal pain. SR16435 and GRT6005 (cebranopadol) are mixed MOP/MOP agonists with varying degrees of partial agonism. Both displayed significant antinociceptive activity and reduced tolerance potential in preclinical models. SUMMARY There is growing evidence for and interest in the design and evaluation of mixed opioids that extend beyond the MOP/DOP pairing to now include NOP. Indeed, a mixed MOP/NOP ligand is close to the clinic; this will reinvigorate the search for other mixed molecules with reduced side-effect profiles.
Collapse
|
41
|
Kwiatkowski K, Piotrowska A, Rojewska E, Makuch W, Jurga A, Slusarczyk J, Trojan E, Basta-Kaim A, Mika J. Beneficial properties of maraviroc on neuropathic pain development and opioid effectiveness in rats. Prog Neuropsychopharmacol Biol Psychiatry 2016; 64:68-78. [PMID: 26190414 DOI: 10.1016/j.pnpbp.2015.07.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/10/2015] [Accepted: 07/12/2015] [Indexed: 01/02/2023]
Abstract
Targeting chemokine signaling pathways is crucial in neuropathy development. In this study, we investigated the influence of chronic administration of maraviroc (CCR5 antagonist) on nociception and opioid effectiveness during neuropathy, which develops as a result of chronic constriction injury (CCI) of the sciatic nerve. To investigate the mechanism of action of maraviroc, we measured the expression of glial cell markers, CCR5 and certain CCR5 ligands (CCL3, CCL4, CCL5, CCL7, CCL11), in the spinal cord and dorsal root ganglia (DRG) of vehicle- and maraviroc-treated, CCI-exposed rats. Our results demonstrate that chronic intrathecal administration of maraviroc diminished neuropathic pain symptoms on day 7 post-CCI. Western blot analysis showed that maraviroc diminished protein level of Iba-1 and GFAP and reversed the up-regulated CCR5 expression observed in spinal cord and DRG after CCI. Additionally, using qRT-PCR, we demonstrated that CCR5 and some of its pronociceptive ligands (CCL3, CCL4, CCL5) increased in the spinal cord after nerve injury, and maraviroc effectively diminished those changes. However, CCL11 spinal expression was undetectable, even after injury. In vitro primary culture studies showed that CCL3, CCL4, CCL5 and CCL7 (but not CCL11) were of microglial and astroglial origin and were up-regulated after LPS stimulation. Our results indicate that maraviroc not only attenuated the development of neuropathic pain symptoms due to significant modulation of neuroimmune interactions but also intensified the analgesic properties of morphine and buprenorphine. In sum, our results suggest the pharmacological modulation of CCR5 by maraviroc as a novel therapeutic approach for co-treatment of patients receiving opioid therapy for neuropathy.
Collapse
Affiliation(s)
- Klaudia Kwiatkowski
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Anna Piotrowska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ewelina Rojewska
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wioletta Makuch
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Agnieszka Jurga
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Joanna Slusarczyk
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ewa Trojan
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Agnieszka Basta-Kaim
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Joanna Mika
- Department of Pain Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| |
Collapse
|
42
|
Kiguchi N, Ding H, Ko MC. Central N/OFQ-NOP Receptor System in Pain Modulation. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2015; 75:217-43. [PMID: 26920014 PMCID: PMC4944813 DOI: 10.1016/bs.apha.2015.10.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Two decades have passed since the peptide, nociceptin/orphanin FQ (N/OFQ), and its cognate (NOP) receptor were discovered. Although NOP receptor activation causes a similar pattern of intracellular actions as mu-opioid (MOP) receptors, NOP receptor-mediated pain modulation in rodents are more complicated than MOP receptor activation. This review highlights the functional evidence of spinal, supraspinal, and systemic actions of NOP receptor agonists for regulating pain. In rodents, effects of the N/OFQ-NOP receptor system in spinal and supraspinal sites for modulating pain are bidirectional depending on the doses, assays, and pain modalities. The net effect of systemically administered NOP receptor agonists may depend on relative contribution of spinal and supraspinal actions of the N/OFQ-NOP receptor signaling in rodents under different pain states. In stark contrast, NOP receptor agonists produce only antinociception and antihypersensitivity in spinal and supraspinal regions of nonhuman primates regardless of doses and assays. More importantly, NOP receptor agonists and a few bifunctional NOP/MOP receptor agonists do not exhibit reinforcing effects (abuse liability), respiratory depression, itch pruritus, nor do they delay the gastrointestinal transit function (constipation) in nonhuman primates. Depending upon their intrinsic efficacies for activating NOP and MOP receptors, bifunctional NOP/MOP receptor agonists warrant additional investigation in primates regarding their side effect profiles. Nevertheless, NOP receptor-related agonists display a much wider therapeutic window as compared to that of MOP receptor agonists in primates. Both selective NOP receptor agonists and bifunctional NOP/MOP receptor agonists hold great potential as effective and safe analgesics without typical opioid-associated side effects in humans.
Collapse
Affiliation(s)
- Norikazu Kiguchi
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Huiping Ding
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA.
| |
Collapse
|
43
|
Di Cesare Mannelli L, Micheli L, Ghelardini C. Nociceptin/orphanin FQ receptor and pain: Feasibility of the fourth opioid family member. Eur J Pharmacol 2015; 766:151-4. [PMID: 26277324 DOI: 10.1016/j.ejphar.2015.08.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 08/10/2015] [Indexed: 10/23/2022]
Abstract
The pharmacological management of chronic pain is a major therapeutic problem. The need of repeated treatments reduces the usefulness of classical analgesic drugs, like μ opioid receptor (MOP) agonists, characterized by tolerance development, side effects and abuse. Moreover, the pathological persistence of pain modifies nociceptive signals and pain-devoted structure activity weakening MOP agonists and making difficult the research of new active molecules. Nociceptine/orphanin FQ (N/OFQ) and its receptor (NOP) offers a peculiar opioid system able to interact with MOP receptors and made more sensitive by chronic pain conditions. The pain reliever efficacy of NOP agonists against persistent pain, mainly neuropathic pain, has been highlighted after intrathecal infusions in rats and non human primates (NHPs). The differences emerged between the effects of NOP stimulation in rodents and NHPs allow to hypothesize the relevance of NOP modulators in higher organisms strongly encouraging the development of compounds active by a systemic route. Possible applicative perspectives are (i) selective NOP agonists as such, (ii) NOP modulation as adjuvant of MOP-based treatments, or (iii) mixed non-selective agonists vs NOP and classical opioid receptors.
Collapse
Affiliation(s)
- Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy.
| | - Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health - NEUROFARBA - Pharmacology and Toxicology Section, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
| |
Collapse
|
44
|
Pessoa BL, Escudeiro G, Nascimento OJM. Emerging Treatments for Neuropathic Pain. Curr Pain Headache Rep 2015; 19:56. [DOI: 10.1007/s11916-015-0530-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
45
|
Vang D, Paul JA, Nguyen J, Tran H, Vincent L, Yasuda D, Zaveri NT, Gupta K. Small-molecule nociceptin receptor agonist ameliorates mast cell activation and pain in sickle mice. Haematologica 2015; 100:1517-25. [PMID: 26294734 DOI: 10.3324/haematol.2015.128736] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/17/2015] [Indexed: 01/04/2023] Open
Abstract
Treatment of pain with morphine and its congeners in sickle cell anemia is suboptimal, warranting the need for analgesics devoid of side effects, addiction and tolerance liability. Small-molecule nociceptin opioid receptor ligands show analgesic efficacy in acute and chronic pain models. We show that AT-200, a high affinity nociceptin opioid receptor agonist with low efficacy at the mu opioid receptor, ameliorated chronic and hypoxia/reoxygenation-induced mechanical, thermal and deep tissue/musculoskeletal hyperalgesia in HbSS-BERK sickle mice. The antinociceptive effect of AT-200 was antagonized by SB-612111, a nociceptin opioid receptor antagonist, but not naloxone, a non-selective mu opioid receptor antagonist. Daily 7-day treatment with AT-200 did not develop tolerance and showed a sustained anti-nociceptive effect, which improved over time and led to reduced plasma serum amyloid protein, neuropeptides, inflammatory cytokines and mast cell activation in the periphery. These data suggest that AT-200 ameliorates pain in sickle mice via the nociceptin opioid receptor by reducing inflammation and mast cell activation without causing tolerance. Thus, nociceptin opioid receptor agonists are promising drugs for treating pain in sickle cell anemia.
Collapse
Affiliation(s)
- Derek Vang
- Vascular Biology Center and Division of Hematology-Oncology-Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Jinny A Paul
- Vascular Biology Center and Division of Hematology-Oncology-Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Julia Nguyen
- Vascular Biology Center and Division of Hematology-Oncology-Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Huy Tran
- Vascular Biology Center and Division of Hematology-Oncology-Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Lucile Vincent
- Vascular Biology Center and Division of Hematology-Oncology-Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | | | - Kalpna Gupta
- Vascular Biology Center and Division of Hematology-Oncology-Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
46
|
Rizzi A, Sukhtankar DD, Ding H, Hayashida K, Ruzza C, Guerrini R, Calò G, Ko MC. Spinal antinociceptive effects of the novel NOP receptor agonist PWT2-nociceptin/orphanin FQ in mice and monkeys. Br J Pharmacol 2015; 172:3661-70. [PMID: 25828800 PMCID: PMC4507167 DOI: 10.1111/bph.13150] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 03/18/2015] [Accepted: 03/30/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE Using an innovative chemical approach, peptide welding technology (PWT), a tetrabranched derivative of nociceptin/orphanin FQ (N/OFQ) has been generated and pharmacologically characterized. Both in vitro and in vivo PWT2-N/OFQ displayed the same pharmacological profile to the natural ligand. It was more potent and produced longer-lasting effects. The aim of the present study was to investigate the spinal effects of PWT2-N/OFQ in nociceptive and neuropathic pain models in mice and non-human primates. EXPERIMENTAL APPROACH Tail withdrawal assay in mice and monkeys was used as a nociceptive pain model and mechanical threshold in mice subjected to chronic constriction injury was used as a neuropathic pain model. The antinociceptive effects of spinally administered N/OFQ and PWT2-N/OFQ were assessed in these models. KEY RESULTS PWT2-N/OFQ mimicked the spinal antinociceptive effects of N/OFQ both in nociceptive and neuropathic pain models in mice as well as in non-human primates displaying 40-fold higher potency and a markedly prolonged duration of action. The effects of N/OFQ and PWT2-N/OFQ were sensitive to the N/OFQ receptor (NOP) antagonist SB-612111, but not to opioid receptor antagonists. CONCLUSIONS AND IMPLICATIONS The present study has demonstrated that PWT2-N/OFQ mimicked the antinociceptive effects of the natural peptide in rodents and non-human primates acting as a potent and longer-lasting NOP-selective agonist. More generally, PWT derivatives of biologically active peptides can be viewed as innovative pharmacological tools for investigating those conditions and states in which selective and prolonged receptor stimulation promotes beneficial effects.
Collapse
Affiliation(s)
- A Rizzi
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - D D Sukhtankar
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - H Ding
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - K Hayashida
- Department of Chemical and Pharmaceutical Sciences and LTTA, University of Ferrara, Ferrara, Italy
| | - C Ruzza
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - R Guerrini
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - G Calò
- Department of Medical Sciences, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, Ferrara, Italy
| | - M C Ko
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
47
|
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: 2.0] [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
|
48
|
Mechanical hyperalgesia in rats with diabetic polyneuropathy is selectively inhibited by local peripheral nociceptin/orphanin FQ receptor and µ-opioid receptor agonism. Eur J Pharmacol 2015; 754:61-5. [DOI: 10.1016/j.ejphar.2015.01.049] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 01/08/2015] [Accepted: 01/26/2015] [Indexed: 01/31/2023]
|
49
|
Abstract
This paper is the thirty-sixth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2013 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, and the roles of these opioid peptides and receptors in pain and analgesia; stress and social status; tolerance and dependence; learning and memory; eating and drinking; alcohol and drugs of abuse; sexual activity and hormones, pregnancy, development and endocrinology; mental illness and mood; seizures and neurologic disorders; electrical-related activity and neurophysiology; general activity and locomotion; gastrointestinal, renal and hepatic functions; cardiovascular responses; respiration and thermoregulation; and immunological responses.
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
|
50
|
Sukhtankar DD, Lagorio CH, Ko MC. Effects of the NOP agonist SCH221510 on producing and attenuating reinforcing effects as measured by drug self-administration in rats. Eur J Pharmacol 2014; 745:182-9. [PMID: 25446568 DOI: 10.1016/j.ejphar.2014.10.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 10/15/2014] [Accepted: 10/16/2014] [Indexed: 10/24/2022]
Abstract
Nociceptin/orphanin FQ peptide (NOP) receptor agonists attenuate morphine-induced conditioned place preference in rodents. However, it is not known whether NOP agonists have reinforcing properties or can inhibit mu opioid receptor (MOP)-mediated reinforcement as measured by drug self-administration in rodents. Further understanding the behavioral effects of NOP agonists could suggest them as having potential in attenuating reinforcing effects of opioids. In the first part of the study, reinforcing properties of selective NOP agonist SCH221510 were determined and compared with the full MOP agonist remifentanil under fixed-ratio 5 (FR5) and progressive-ratio (PR) schedules of drug self-administration. In the second part, effects of systemic and intracisternal pretreatment of SCH221510 were determined and compared with MOP antagonist naltrexone in attenuating reinforcing effects of remifentanil and a non-drug reinforcer (sucrose pellets). Remifentanil self-administration (0.3-10 µg/kg/infusion) generated a biphasic dose-response curve, characteristic of drugs with reinforcing properties. SCH221510 (3-300 µg/kg/infusion) self-administration resulted in flat dose-response curves and early break-points under the PR, indicative of drugs lacking reinforcing value. Intracisternally, but not systemically, administered SCH221510 (0.3-3 µg) attenuated remifentanil self-administration, comparable with systemic naltrexone (0.03-0.3 mg/kg). SCH221510 (1-3 µg), unlike naltrexone (0.03-1 mg/kg), attenuated responding for sucrose pellets. Both effects of SCH221510 were reversed by the NOP antagonist J-113397 (0.3-3 µg). These results suggest that SCH221510 does not function as a reinforcer in rats, and that it can attenuate the reinforcing value of MOP agonists; therefore, the potential utility of NOP agonists for the treatment of drug addiction warrants further evaluation.
Collapse
Affiliation(s)
- Devki D Sukhtankar
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | - Carla H Lagorio
- Department of Psychology, University of Wisconsin-Eau Claire, Eau Claire, WI 54702, USA
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA; Center for Comparative Medicine Research, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|