1
|
Cannella N, Lunerti V, Shen Q, Li H, Benvenuti F, Soverchia L, Narendran R, Weiss F, Ciccocioppo R. Cebranopadol, a novel long-acting opioid agonist with low abuse liability, to treat opioid use disorder: Preclinical evidence of efficacy. Neuropharmacology 2024; 257:110048. [PMID: 38901642 DOI: 10.1016/j.neuropharm.2024.110048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/09/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
Maintenance therapy with buprenorphine and methadone is the gold standard pharmacological treatment for opioid use disorder (OUD). Despite these compounds demonstrating substantial efficacy, a significant number of patients do not show optimal therapeutic responses. The abuse liability of these medications is also a concern. Here we used rats to explore the therapeutic potential of the new long-acting pan-opioid agonist Cebranopadol in OUD. We tested the effect of cebranopadol on heroin self-administration and yohimbine-induced reinstatement of heroin seeking. In addition, we evaluated the abuse liability potential of cebranopadol in comparison to that of heroin under fixed ratio 1 (FR1) and progressive ratio (PR) operant self-administration contingencies. Oral administration of cebranopadol (0, 25, 50 μg/kg) significantly attenuated drug self-administration independent of heroin dose (1, 7, 20, 60μg/inf). Cebranopadol also reduced the break point for heroin (20 μg/inf). Finally, pretreatment with cebranopadol significantly attenuated yohimbine-induced reinstatement of drug seeking. In abuse liability experiments under FR1 contingency, rats maintained responding for heroin (1, 7, 20, 60μg/inf) to a larger extent than cebranopadol (0.03, 0.1, 0.3, 1.0, 6.0μg/inf). Under PR contingency, heroin maintained responding at high levels at all except the lowest dose, while the break point (BP) for cebranopadol did not differ from that of saline. Together, these data indicate that cebranopadol is highly efficacious in attenuating opioid self-administration and stress-induced reinstatement, while having limited abuse liability properties. Overall, the data suggest clinical potential of this compound for OUD treatment.
Collapse
Affiliation(s)
- Nazzareno Cannella
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy
| | - Veronica Lunerti
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy
| | - Qianwei Shen
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy
| | - Hongwu Li
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy; School of Chemical Engineering, Changchun University of Changchung, 130012, China
| | - Federica Benvenuti
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy
| | - Laura Soverchia
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy
| | - Rajesh Narendran
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Friedbert Weiss
- Department of Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - Roberto Ciccocioppo
- School of Pharmacy, Center for Neuroscience, Pharmacology Unit, University of Camerino, Italy.
| |
Collapse
|
2
|
Qin F, Wang Q, Wang Y, Li Z, Liu A, Liu Q, Lin W, Mu X, Liu X, Wang Q, Lu Z. Exoticin as a selective agonist of 6TM μ opioid receptors identifies endogenous chaperones essential for its activity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155898. [PMID: 39154526 DOI: 10.1016/j.phymed.2024.155898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/11/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Classical opioids are effective analgesics but carry various side effects, necessitating safer alternatives. Truncated six-transmembrane mu opioid receptors (6TM-μORs) mediate potent analgesia with fewer side effects and are a promising therapeutic target. However, few ligands known selectively target 6TM-μORs. Moreover, endogenous chaperones are believed essential for 6TM-μOR ligand binding and function. PURPOSE To identify a 6TM-μOR selective agonist and elucidate requisite endogenous chaperones. METHODS Virtual screening was used to identify promising selective 6TM-μOR agonists from traditional Chinese medicines. The role of 6TM-μOR in Exoticin analgesia was validated in loss- and gain-of-function models. APEX2 proteomics profiled proximal proteins under Exoticin or IBNtxA. Interactions were further characterized in vivo and in vitro. RESULTS Exoticin was shortlisted for its selective binding to 6TM-μOR and ability to induce 6TM-μOR-dependent signal transduction. Exoticin analgesia was sensitive to β-FNA and absent in E11 KO mice, but restored in mice infected with AAV-μOR1G. Slc3a2, Lrrc59, and Ppp1cb co-interacted with 6TM-μOR1G and were equally essential for Exoticin binding and 6TM-μOR1G activity. CONCLUSION Exoticin is a promising selective agonist of 6TM μ opioid receptors with broad-spectrum analgesic efficacy but few side effects. Slc3a2, Lrrc59, Ppp1cb are endogenous chaperones essential for 6TM-μOR ligand binding and function.
Collapse
Affiliation(s)
- Fenfen Qin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qisheng Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuxuan Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhonghao Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Anlong Liu
- Nanjing Hospital of Traditional Chinese Medicine Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Qingyang Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weixin Lin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xinru Mu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xingjun Liu
- School of Pharmacy, Nantong University, Nantong 226001, China.
| | - Qian Wang
- International Education college, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Zhigang Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
3
|
Breault É, Desgagné M, Neve JD, Côté J, Barlow TMA, Ballet S, Sarret P. Multitarget ligands that comprise opioid/nonopioid pharmacophores for pain management: Current state of the science. Pharmacol Res 2024; 209:107408. [PMID: 39307212 DOI: 10.1016/j.phrs.2024.107408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Chronic pain, which affects more than one-third of the world's population, represents one of the greatest medical challenges of the 21st century, yet its effective management remains sub-optimal. The 'gold standard' for the treatment of moderate to severe pain consists of opioid ligands, such as morphine and fentanyl, that target the µ-opioid receptor (MOP). Paradoxically, these opioids also cause serious side effects, including constipation, respiratory depression, tolerance, and addiction. In addition, the development of opioid-use disorders, such as opioid diversion, misuse, and abuse, has led to the current opioid crisis, with dramatic increases in addiction, overdoses, and ultimately deaths. As pain is a complex, multidimensional experience involving a variety of pathways and mediators, dual or multitarget ligands that can bind to more than one receptor and exert complementary analgesic effects, represent a promising avenue for pain relief. Indeed, unlike monomodal therapeutic approaches, the modulation of several endogenous nociceptive systems can often result in an additive or even synergistic effect, thereby improving the analgesic-to-side-effect ratio. Here, we provide a comprehensive overview of research efforts towards the development of dual- or multi-targeting opioid/nonopioid hybrid ligands for effective and safer pain management. We reflect on the underpinning discovery rationale by discussing the design, medicinal chemistry, and in vivo pharmacological effects of multitarget antinociceptive compounds.
Collapse
Affiliation(s)
- Émile Breault
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Michael Desgagné
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, Brussels 1050, Belgium
| | - Jérôme Côté
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Thomas M A Barlow
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, Brussels 1050, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, Brussels 1050, Belgium
| | - Philippe Sarret
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
4
|
De Neve J, Breault É, Previti S, Vangeloven E, Loranger B, Chartier M, Brouillette R, Lanoie A, Holleran BJ, Longpré JM, Gendron L, Tourwé D, Sarret P, Ballet S. Design, Synthesis, and In Vitro Characterization of Proteolytically-Stable Opioid-Neurotensin Hybrid Peptidomimetics. ACS Pharmacol Transl Sci 2024; 7:2784-2798. [PMID: 39296263 PMCID: PMC11406707 DOI: 10.1021/acsptsci.4c00236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 09/21/2024]
Abstract
Linking an opioid to a nonopioid pharmacophore represents a promising approach for reducing opioid-induced side effects during pain management. Herein, we describe the optimization of the previously reported opioid-neurotensin hybrids (OPNT-hybrids), SBL-OPNT-05 & -10, containing the μ-/δ-opioid agonist H-Dmt-d-Arg-Aba-β-Ala-NH2 and NT(8-13) analogs optimized for NTS2 affinity. In the present work, the constrained dipeptide Aba-β-Ala was modified to investigate the optimal linker length between the two pharmacophores, as well as the effect of expanding the aromatic moiety within constrained dipeptide analogs, via the inclusion of a naphthyl moiety. Additionally, the N-terminal Arg residue of the NT(8-13) pharmacophore was substituted with β3 hArg. For all analogs, affinity was determined at the MOP, DOP, NTS1, and NTS2 receptors. Several of the hybrid ligands showed a subnanomolar affinity for MOP, improved binding for DOP compared to SBL-OPNT-05 & -10, as well as an excellent NTS2-affinity with high selectivity over NTS1. Subsequently, the Gαi1 and β-arrestin-2 pathways were evaluated for all hybrids, along with their stability in rat plasma. Upon MOP activation, SBL-OPNT-13 and -18 were the least effective at recruiting β-arrestin-2 (E max = 17 and 12%, respectively), while both compounds were also found to be partial agonists at the Gαi1 pathway, despite improved potency compared to DAMGO. Importantly, these analogs also showed a half-life in rat plasma in excess of 48 h, making them valuable tools for future in vivo investigations.
Collapse
Affiliation(s)
- Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Émile Breault
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Santo Previti
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Esaü Vangeloven
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Bobbi Loranger
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Magali Chartier
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Rebecca Brouillette
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Annik Lanoie
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Brian J Holleran
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Jean-Michel Longpré
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Louis Gendron
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Dirk Tourwé
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Philippe Sarret
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4 Sherbrooke, Quebec, Canada
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| |
Collapse
|
5
|
Wu Y, Song X, Ji Y, Chen G, Zhao L. A synthetic peptide exerts nontolerance-forming antihyperalgesic and antidepressant effects in mice. Neurotherapeutics 2024; 21:e00377. [PMID: 38777742 PMCID: PMC11284537 DOI: 10.1016/j.neurot.2024.e00377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Chronic pain is a prevalent and persistent ailment that affects individuals worldwide. Conventional medications employed in the treatment of chronic pain typically demonstrate limited analgesic effectiveness and frequently give rise to debilitating side effects, such as tolerance and addiction, thereby diminishing patient compliance with medication. Consequently, there is an urgent need for the development of efficacious novel analgesics and innovative methodologies to address chronic pain. Recently, a growing body of evidence has suggested that multireceptor ligands targeting opioid receptors (ORs) are favorable for improving analgesic efficacy, decreasing the risk of adverse effects, and occasionally yielding additional advantages. In this study, the intrathecal injection of a recently developed peptide (VYWEMEDKN) at nanomolar concentrations decreased pain sensitivity in naïve mice and effectively reduced pain-related behaviors in nociceptive pain model mice with minimal opioid-related side effects. Importantly, the compound exerted significant rapid-acting antidepressant effects in both the forced swim test and tail suspension test. It is possible that the rapid antihyperalgesic and antidepressant effects of the peptide are mediated through the OR pathway. Overall, this peptide could both effectively provide pain relief and alleviate depression with fewer side effects, suggesting that it is a potential agent for chronic pain and depression comorbidities from the perspective of pharmaceutical development.
Collapse
Affiliation(s)
- Yongjiang Wu
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China
| | - Xiaofei Song
- Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - YanZhe Ji
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China
| | - Gang Chen
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China; Key Laboratory of Neuroregeneration of Jiangsu and the Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China; Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China.
| | - Long Zhao
- Center for Basic Medical Research, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China.
| |
Collapse
|
6
|
Gao N, Xu X, Ye F, Li XY, Lin C, Shen XW, Qian J. Crizotinib inhibits the metabolism of tramadol by non-competitive suppressing the activities of CYP2D1 and CYP3A2. PeerJ 2024; 12:e17446. [PMID: 38827306 PMCID: PMC11144398 DOI: 10.7717/peerj.17446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/02/2024] [Indexed: 06/04/2024] Open
Abstract
Objectives To investigate the interaction between tramadol and representative tyrosine kinase inhibitors, and to study the inhibition mode of drug-interaction. Methods Liver microsomal catalyzing assay was developed. Sprague-Dawley rats were administrated tramadol with or without selected tyrosine kinase inhibitors. Samples were prepared and ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) was used for analysis. Besides, liver, kidney, and small intestine were collected and morphology was examined by hematoxyline-eosin (H&E) staining. Meanwhile, liver microsomes were prepared and carbon monoxide differential ultraviolet radiation (UV) spectrophotometric quantification was performed. Results Among the screened inhibitors, crizotinib takes the highest potency in suppressing the metabolism of tramadol in rat/human liver microsome, following non-competitive inhibitory mechanism. In vivo, when crizotinib was co-administered, the AUC value of tramadol increased compared with the control group. Besides, no obvious pathological changes were observed, including cell morphology, size, arrangement, nuclear morphology with the levels of alanine transaminase (ALT) and aspartate transaminase (AST) increased after multiple administration of crizotinib. Meanwhile, the activities of CYP2D1 and CYP3A2 as well as the total cytochrome P450 abundance were found to be decreased in rat liver of combinational group. Conclusions Crizotinib can inhibit the metabolism of tramadol. Therefore, this recipe should be vigilant to prevent adverse reactions.
Collapse
Affiliation(s)
- Nanyong Gao
- Yueqing Maternity and Child Health Hospital, Wenzhou, China
- Wenzhou Medical University, Wenzhou, China
| | - Xiaoyu Xu
- Wenzhou Medical University, Wenzhou, China
| | - Feng Ye
- Wenzhou Medical University, Wenzhou, China
| | - Xin-yue Li
- Wenzhou Medical University, Wenzhou, China
| | | | | | | |
Collapse
|
7
|
De Neve J, Elhabazi K, Gonzalez S, Herby C, Schneider S, Utard V, Fellmann-Clauss R, Petit-Demouliere N, Lecat S, Kremer M, Ces A, Daubeuf F, Martin C, Ballet S, Bihel F, Simonin F. Multitarget μ-Opioid Receptor Agonists─Neuropeptide FF Receptor Antagonists Induce Potent Antinociception with Reduced Adverse Side Effects. J Med Chem 2024. [PMID: 38687204 DOI: 10.1021/acs.jmedchem.4c00442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The design of bifunctional compounds is a promising approach toward the development of strong analgesics with reduced side effects. We here report the optimization of the previously published lead peptide KGFF09, which contains opioid receptor agonist and neuropeptide FF receptor antagonist pharmacophores and is shown to induce potent antinociception and reduced side effects. We evaluated the novel hybrid peptides for their in vitro activity at MOP, NPFFR1, and NPFFR2 and selected four of them (DP08/14/32/50) for assessment of their acute antinociceptive activity in mice. We further selected DP32 and DP50 and observed that their antinociceptive activity is mostly peripherally mediated; they produced no respiratory depression, no hyperalgesia, significantly less tolerance, and strongly attenuated withdrawal syndrome, as compared to morphine and the recently FDA-approved TRV130. Overall, these data suggest that MOP agonist/NPFF receptor antagonist hybrids might represent an interesting strategy to develop novel analgesics with reduced side effects.
Collapse
Affiliation(s)
- Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Khadija Elhabazi
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Simon Gonzalez
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Claire Herby
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Séverine Schneider
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Valérie Utard
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Rosine Fellmann-Clauss
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Nathalie Petit-Demouliere
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Sandra Lecat
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Mélanie Kremer
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (INCI), 67000 Strasbourg, France
| | - Aurelia Ces
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives (INCI), 67000 Strasbourg, France
| | - François Daubeuf
- Plateforme de Chimie Biologique Intégrative de Strasbourg, UAR 3286, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Charlotte Martin
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Frédéric Bihel
- Laboratoire d'Innovation Thérapeutique, Faculté de Pharmacie, UMR 7200, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR 7242, Centre National de la Recherche Scientifique, Université de Strasbourg, 67400 Illkirch, France
| |
Collapse
|
8
|
Ujcikova H, Lee YS, Roubalova L, Svoboda P. The impact of multifunctional enkephalin analogs and morphine on the protein changes in crude membrane fractions isolated from the rat brain cortex and hippocampus. Peptides 2024; 174:171165. [PMID: 38307418 DOI: 10.1016/j.peptides.2024.171165] [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: 12/11/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Endogenous opioid peptides serve as potent analgesics through the opioid receptor (OR) activation. However, they often suffer from poor metabolic stability, low lipophilicity, and low blood-brain barrier permeability. Researchers have developed many strategies to overcome the drawbacks of current pain medications and unwanted biological effects produced by the interaction with opioid receptors. Here, we tested multifunctional enkephalin analogs LYS739 (MOR/DOR agonist and KOR partial antagonist) and LYS744 (MOR/DOR agonist and KOR full antagonist) under in vivo conditions in comparison with MOR agonist, morphine. We applied 2D electrophoretic resolution to investigate differences in proteome profiles of crude membrane (CM) fractions isolated from the rat brain cortex and hippocampus exposed to the drugs (10 mg/kg, seven days). Our results have shown that treatment with analog LYS739 induced the most protein changes in cortical and hippocampal samples. The identified proteins were mainly associated with energy metabolism, cell shape and movement, apoptosis, protein folding, regulation of redox homeostasis, and signal transduction. Among these, the isoform of mitochondrial ATP synthase subunit beta (ATP5F1B) was the only protein upregulation in the hippocampus but not in the brain cortex. Contrarily, the administration of analog LYS744 caused a small number of protein alterations in both brain parts. Our results indicate that the KOR full antagonism, together with MOR/DOR agonism of multifunctional opioid ligands, can be beneficial in treating chronic pain states by reducing changes in protein expression levels but retaining analgesic efficacy.
Collapse
Affiliation(s)
- Hana Ujcikova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic.
| | - Yeon Sun Lee
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA
| | - Lenka Roubalova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Petr Svoboda
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| |
Collapse
|
9
|
Kan BF, Liu XY, Han MM, Yang CW, Zhu X, Jin Y, Wang D, Huang X, Wu WJ, Fu T, Kang F, Zhang Z, Li J. Nerve Growth Factor/Tyrosine Kinase A Receptor Pathway Enhances Analgesia in an Experimental Mouse Model of Bone Cancer Pain by Increasing Membrane Levels of δ-Opioid Receptors. Anesthesiology 2024; 140:765-785. [PMID: 38118180 DOI: 10.1097/aln.0000000000004880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
BACKGROUND The role of nerve growth factor (NGF)/tyrosine kinase A receptor (TrKA) signaling, which is activated in a variety of pain states, in regulating membrane-associated δ-opioid receptor (mDOR) expression is poorly understood. The hypothesis was that elevated NGF in bone cancer tumors could upregulate mDOR expression in spinal cord neurons and that mDOR agonism might alleviate bone cancer pain. METHODS Bone cancer pain (BCP) was induced by inoculating Lewis lung carcinoma cells into the femoral marrow cavity of adult C57BL/6J mice of both sexes. Nociceptive behaviors were evaluated by the von Frey and Hargreaves tests. Protein expression in the spinal dorsal horn of animals was measured by biochemical analyses, and excitatory synaptic transmission was recorded in miniature excitatory synaptic currents. RESULTS The authors found that mDOR expression was increased in BCP mice (BCP vs. sham, mean ± SD: 0.18 ± 0.01 g vs. mean ± SD: 0.13 ± 0.01 g, n = 4, P < 0.001) and that administration of the DOR agonist deltorphin 2 (Del2) increased nociceptive thresholds (Del2 vs. vehicle, median [25th, 75th percentiles]: 1.00 [0.60, 1.40] g vs. median [25th, 75th percentiles]: 0.40 [0.16, 0.45] g, n = 10, P = 0.001) and reduced miniature excitatory synaptic current frequency in lamina II outer neurons (Del2 vs. baseline, mean ± SD: 2.21 ± 0.81 Hz vs. mean ± SD: 2.43 ± 0.90 Hz, n = 12, P < 0.001). Additionally, NGF expression was increased in BCP mice (BCP vs. sham, mean ± SD: 0.36 ± 0.03 vs. mean ± SD: 0.16 ± 0.02, n = 4, P < 0.001), and elevated NGF was associated with enhanced mDOR expression via TrKA signaling. CONCLUSIONS Activation of mDOR produces analgesia that is dependent on the upregulation of the NGF/TrKA pathway by increasing mDOR levels under conditions of BCP in mice. EDITOR’S PERSPECTIVE
Collapse
Affiliation(s)
- Bu-Fan Kan
- Department of Anesthesiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xing-Yun Liu
- Department of Anesthesiology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ming-Ming Han
- Department of Anesthesiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Cheng-Wei Yang
- Department of Anesthesiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xia Zhu
- Department of Anesthesiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yan Jin
- Stroke Center and Department of Neurology, Department of Anesthesiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Di Wang
- Department of Anesthesiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiang Huang
- Department of Anesthesiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wen-Jie Wu
- Department of Anesthesiology, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei, China
| | - Tong Fu
- Graduate School of Wannan Medical College, Wuhu, China
| | - Fang Kang
- Department of Anesthesiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhi Zhang
- Department of Anesthesiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; and Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, University of Science and Technology of China, Hefei, China
| | - Juan Li
- Department of Anesthesiology, the First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
10
|
Zhang YZ, Wang SY, Guo XC, Liu XH, Wang XF, Wang MM, Qiu TT, Han FT, Zhang Y, Wang CL. Novel endomorphin analogues CEMR-1 and CEMR-2 produce potent and long-lasting antinociception with a favourable side effect profile at the spinal level. Br J Pharmacol 2024; 181:1268-1289. [PMID: 37990825 DOI: 10.1111/bph.16287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 10/09/2023] [Accepted: 11/08/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND AND PURPOSE Endomorphins have shown great promise as pharmaceutics for the treatment of pain. We have previously confirmed that novel endomorphin analogues CEMR-1 and CEMR-2 behaved as potent μ agonists and displayed potent antinociceptive activities at the supraspinal and peripheral levels. The present study was undertaken to evaluate the antinociceptive properties of CEMR-1 and CEMR-2 following intrathecal (i.t.) administration. Furthermore, their antinociceptive tolerance and opioid-like side effects were also determined. EXPERIMENTAL APPROACH The spinal antinociceptive effects of CEMR-1 and CEMR-2 were determined in a series of pain models, including acute radiant heat paw withdrawal test, spared nerve injury-induced neuropathic pain, complete Freund's adjuvant-induced inflammatory pain, visceral pain and formalin pain. Antinociceptive tolerance was evaluated in radiant heat paw withdrawal test. KEY RESULTS Spinal administration of CEMR-1 and CEMR-2 produced potent and prolonged antinociceptive effects in acute pain. CEMR-1 and CEMR-2 may produce their antinociception through distinct μ receptor subtypes. These two analogues also exhibited significant analgesic activities in neuropathic, inflammatory, visceral and formalin pain at the spinal level. It is noteworthy that CEMR-1 showed non-tolerance-forming analgesic properties, while CEMR-2 exhibited substantially reduced antinociceptive tolerance. Furthermore, both analogues displayed no or reduced side effects on conditioned place preference response, physical dependence, locomotor activity and gastrointestinal transit. CONCLUSIONS AND IMPLICATIONS The present investigation demonstrated that CEMR-1 and CEMR-2 displayed potent and long-lasting antinociception with a favourable side effect profile at the spinal level. Therefore, CEMR-1 and CEMR-2 might serve as promising analgesic compounds with minimal opioid-like side effects.
Collapse
Affiliation(s)
- Yu-Zhe Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Si-Yu Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xue-Ci Guo
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xiao-Han Liu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | | | - Meng-Meng Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Ting-Ting Qiu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Feng-Tong Han
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yao Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Chang-Lin Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, China
| |
Collapse
|
11
|
Gach-Janczak K, Biernat M, Kuczer M, Adamska-Bartłomiejczyk A, Kluczyk A. Analgesic Peptides: From Natural Diversity to Rational Design. Molecules 2024; 29:1544. [PMID: 38611824 PMCID: PMC11013236 DOI: 10.3390/molecules29071544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Pain affects one-third of the global population and is a significant public health issue. The use of opioid drugs, which are the strongest painkillers, is associated with several side effects, such as tolerance, addiction, overdose, and even death. An increasing demand for novel, safer analgesic agents is a driving force for exploring natural sources of bioactive peptides with antinociceptive activity. Since the G protein-coupled receptors (GPCRs) play a crucial role in pain modulation, the discovery of new peptide ligands for GPCRs is a significant challenge for novel drug development. The aim of this review is to present peptides of human and animal origin with antinociceptive potential and to show the possibilities of their modification, as well as the design of novel structures. The study presents the current knowledge on structure-activity relationship in the design of peptide-based biomimetic compounds, the modification strategies directed at increasing the antinociceptive activity, and improvement of metabolic stability and pharmacodynamic profile. The procedures employed in prolonged drug delivery of emerging compounds are also discussed. The work summarizes the conditions leading to the development of potential morphine replacements.
Collapse
Affiliation(s)
- Katarzyna Gach-Janczak
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.G.-J.); (A.A.-B.)
| | - Monika Biernat
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (M.B.); (M.K.)
| | - Mariola Kuczer
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (M.B.); (M.K.)
| | - Anna Adamska-Bartłomiejczyk
- Department of Biomolecular Chemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.G.-J.); (A.A.-B.)
| | - Alicja Kluczyk
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland; (M.B.); (M.K.)
| |
Collapse
|
12
|
Ettitaou A, Kabdy H, Oubella K, Raoui K, Oubahmane M, Aboufatima R, Elyazouli L, Garzoli S, Chait A. Molecular docking of quercetin: a promising approach for the development of new anti-inflammatory and analgesic drugs. Nat Prod Res 2024:1-10. [PMID: 38520257 DOI: 10.1080/14786419.2024.2333053] [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: 02/02/2024] [Accepted: 03/12/2024] [Indexed: 03/25/2024]
Abstract
The aim of this study is to investigate the antinociceptive, anti-inflammatory and antipyretic effects of quercetin. Additionally, molecular docking studies were conducted to evaluate potential interactions between quercetin and various molecular targets. Animal models were used to conduct a comprehensive pharmacological investigation of quercetin. Evaluation of analgesic activity revealed a reduction in the number of abdominal cramps during the twisting test and inhibition of pain during the second phase of the formaldehyde test. Additionally, evaluation of its anti-inflammatory activity showed a reduction in ear oedema. However, it is important to note that quercetin administration has not been shown to significantly reduce yeast-induced hyperthermia. The docking study revealed the high inhibitory potential of quercetin against the COX-2 receptor.
Collapse
Affiliation(s)
- Amina Ettitaou
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Hamid Kabdy
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Khadija Oubella
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Karima Raoui
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Mehdi Oubahmane
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech, Morocco
| | - Rachida Aboufatima
- Laboratory of Genie Biologic, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Loubna Elyazouli
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Stefania Garzoli
- Department of Chemistry and Technologies of Drug, Sapienza University, Rome, Italy
| | - Abderrahman Chait
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| |
Collapse
|
13
|
Huang P, Ho CK, Cao D, Inan S, Rawls SM, Li M, Huang B, Pagare PP, Townsend EA, Poklis JL, Halquist MS, Banks M, Zhang Y, Liu-Chen LY. NCP, a dual kappa and mu opioid receptor agonist, is a potent analgesic against inflammatory pain without reinforcing or aversive properties. J Pharmacol Exp Ther 2024; 389:JPET-AR-2023-001870. [PMID: 38409113 PMCID: PMC10949162 DOI: 10.1124/jpet.123.001870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/28/2024] Open
Abstract
While agonists of mu (MOR) and kappa (KOR) opioid receptors have analgesic effects, they produce euphoria and dysphoria, respectively. Other side effects include respiratory depression and addiction for MOR agonists and sedation for KOR agonists. We reported that 17-cyclopropylmethyl-3,14β-dihydroxy-4,5α-epoxy-6β-{[4'-(2'-cyanopyridyl)]carboxamido}cmorphinan (NCP) displayed potent KOR full agonist and MOR partial agonist activities (58%) with 6.5x KOR-over-MOR selectivity in vitro Herein, we characterized pharmacological effects of NCP in rodents. In mice, NCP exerted analgesic effects against inflammatory pain in both the formalin test and the acetic acid writhing test, with A50 values of 47.6 and 14.4 microg/kg (s.c.), respectively. The analgesic effects in the acetic acid writhing test were mediated by the KOR. NCP at doses much higher than those effective in reducing inflammatory pain did not produce antinociception in the hot plate and tail flick tests, inhibit compound 48/80-induced scratching, cause conditioned place aversion (CPA) or preference, impair rotarod performance, inhibit locomotor activity, cause respiratory depression, or precipitate morphine withdrawal. However, NCP (10~100 microg/kg) inhibited gastrointestinal transit with a maximum of ~40% inhibition. In MOR knockout mice, NCP caused CPA, demonstrating that its lack of CPA is due to combined actions on the MOR and KOR. Following s.c. injection, NCP penetrated into the mouse brain. In rats trained to self-administer heroin, NCP (1~320 microg/kg/infusion) did not function as a reinforcer. Thus, NCP produces potent analgesic effects via KOR without side effects except constipation. Therefore, dual full KOR/partial MOR agonists with moderate KOR-over-MOR selectivity may be promising as non-addictive analgesics for inflammatory pain. Significance Statement Developing non-addictive analgesics is crucial for reducing opioid overdose deaths, minimizing drug misuse, and promoting safer pain management practices. Herein, pharmacology of a potential non-addictive analgesic, NCP, is reported. NCP has full KOR agonist / partial MOR agonist activities with a 6.5 x selectivity for KOR over MOR. Unlike MOR agonists, analgesic doses of NCP do not lead to self-administration or respiratory depression. Furthermore, NCP does not produce aversion, hypolocomotion, or motor incoordination, side effects typically associated with KOR activation.
Collapse
Affiliation(s)
- Peng Huang
- Temple University Lewis Katz School of Medicine, United States
| | - Conrad K Ho
- Temple University Lewis Katz School of Medicine, United States
| | - Danni Cao
- Temple University Lewis Katz School of Medicine, United States
| | - Saadet Inan
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Lewis Katz School of Medicine, Temple University, United States
| | - Scott M Rawls
- Temple University Lewis Katz School of Medicine, United States
| | - Mengchu Li
- Virginia Commonwealth University School of Pharmacy, United States
| | - Boshi Huang
- Virginia Commonwealth University School of Pharmacy, United States
| | - Piyusha P Pagare
- Virginia Commonwealth University School of Pharmacy, United States
| | | | | | | | - Matthew Banks
- Virginia Commonwealth University School of Medicine, United States
| | - Yan Zhang
- Virginia Commonwealth University School of Pharmacy, United States
| | - Lee-Yuan Liu-Chen
- Center for Substance Abuse Research, Temple University Lewis Katz School of Medicine, United States
| |
Collapse
|
14
|
Zhang Q, Xu B, Chen D, Wu S, Hu X, Zhang X, Yu B, Zhang S, Yang Z, Zhang M, Fang Q. Structure-Activity Relationships of a Novel Cyclic Hexapeptide That Exhibits Multifunctional Opioid Agonism and Produces Potent Antinociceptive Activity. J Med Chem 2024; 67:272-288. [PMID: 38118143 DOI: 10.1021/acs.jmedchem.3c01347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The cyclic peptide c[d-Lys2, Asp5]-DN-9 has recently been identified as a multifunctional opioid/neuropeptide FF receptor agonist, displaying potent analgesic activity with reduced side effects. This study utilized Tyr-c[d-Lys-Gly-Phe-Asp]-d-Pro-NH2 (0), a cyclic hexapeptide derived from the opioid pharmacophore of c[d-Lys2, Asp5]-DN-9, as a chemical template. We designed, synthesized, and characterized 22 analogs of 0 with a single amino acid substitution to investigate its structure-activity relationship. Most of these cyclic hexapeptide analogs exhibited multifunctional activity at μ and δ opioid receptors (MOR and DOR, respectively) and produced antinociceptive effects following subcutaneous administration. The lead compound analog 15 showed potent agonistic activities at the MOR, κ opioid receptor (KOR), and DOR in vitro and produced a strong and long-lasting analgesic effect through peripheral MOR and KOR in the tail-flick test. Further biological evaluation identified that analog 15 did not cause significant side effects such as tolerance, withdrawal, or reward liability.
Collapse
Affiliation(s)
- Qinqin Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Biao Xu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Dan Chen
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Shuyuan Wu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Xuanran Hu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Xiaodi Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Bowen Yu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Shichao Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Zhenyun Yang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Mengna Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Quan Fang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| |
Collapse
|
15
|
Zhang M, Xu B, Li N, Zhang Q, Chen D, Wu S, Yu B, Zhang X, Hu X, Zhang S, Jing Y, Yang Z, Jiang J, Fang Q. All-Hydrocarbon Stapled Peptide Multifunctional Agonists at Opioid and Neuropeptide FF Receptors: Highly Potent, Long-Lasting Brain Permeant Analgesics with Diminished Side Effects. J Med Chem 2023; 66:17138-17154. [PMID: 38095323 DOI: 10.1021/acs.jmedchem.3c02093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Our previous study reported the multifunctional agonist for opioid and neuropeptide FF receptors DN-9, along with its cyclic peptide analogues c[D-Cys2, Cys5]-DN-9 and c[D-Lys2, Asp5]-DN-9. These analogues demonstrated potent antinociceptive effects with reduced opioid-related side effects. To develop more stable and effective analgesics, we designed, synthesized, and evaluated seven hydrocarbon-stapled cyclic peptides based on DN-9. In vitro calcium mobilization assays revealed that most of the stapled peptides, except 3, displayed multifunctional agonistic activities at opioid and neuropeptide FF receptors. Subcutaneous administration of all stapled peptides resulted in effective and long-lasting antinociceptive activities lasting up to 360 min. Among these stapled peptides, 1a and 1b emerged as the optimized compounds, producing potent central antinociception following subcutaneous, intracerebroventricular, and oral administrations. Additionally, subcutaneous administration of 1a and 1b caused nontolerance antinociception, with limited occurrence of constipation and addiction. Furthermore, 1a was selected as the final optimized compound due to its wider safety window compared to 1b.
Collapse
Affiliation(s)
- Mengna Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Biao Xu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Ning Li
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Qinqin Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Dan Chen
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Shuyuan Wu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Bowen Yu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Xiaodi Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Xuanran Hu
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Shichao Zhang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Yuhong Jing
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Zhenyun Yang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| | - Jinhong Jiang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, Jiangsu 221004, China
| | - Quan Fang
- Institute of Physiology, School of Basic Medical Sciences, and State Key Laboratory of Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou 730000, PR China
| |
Collapse
|
16
|
He Y, Su Q, Zhao L, Zhang L, Yu L, Shi J. Historical perspectives and recent advances in small molecule ligands of selective/biased/multi-targeted μ/δ/κ opioid receptor (2019-2022). Bioorg Chem 2023; 141:106869. [PMID: 37797454 DOI: 10.1016/j.bioorg.2023.106869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023]
Abstract
The opioids have been used for more than a thousand years and are not only the most widely prescribed drugs for moderate to severe pain and acute pain, but also the preferred drugs. However, their non-analgesic effects, especially respiratory depression and potential addiction, are important factors that plague the safety of clinical use and are an urgent problem for pharmacological researchers to address. Current research on analgesic drugs has evolved into different directions: de-opioidization; application of pharmacogenomics to individualize the use of opioids; development of new opioids with less adverse effects. The development of new opioid drugs remains a hot research topic, and with the in-depth study of opioid receptors and intracellular signal transduction mechanisms, new research ideas have been provided for the development of new opioid analgesics with less side effects and stronger analgesic effects. The development of novel opioid drugs in turn includes selective opioid receptor ligands, biased opioid receptor ligands, and multi-target opioid receptor ligands and positive allosteric modulators (PAMs) or antagonists and the single compound as multi-targeted agnoists/antagonists for different receptors. PAMs strategies are also getting newer and are the current research hotspots, including the BMS series of compounds and others, which are extensive and beyond the scope of this review. This review mainly focuses on the selective/biased/multi-targeted MOR/DOR/KOR (mu opioid receptor/delta opioid receptor/kappa opioid receptor) small molecule ligands and involves some cryo-electron microscopy (cryoEM) and structure-based approaches as well as the single compound as multi-targeted agnoists/antagonists for different receptors from 2019 to 2022, including discovery history, activities in vitro and vivo, and clinical studies, in an attempt to provide ideas for the development of novel opioid analgesics with fewer side effects.
Collapse
Affiliation(s)
- Ye He
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Qian Su
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Liyun Zhao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lijuan Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Lu Yu
- Department of Respiratory Medicine, Sichuan Academy of Medical Sciences and Sichuan provincial People's Hospital, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
17
|
Illuminati D, Trapella C, Zanirato V, Guerrini R, Albanese V, Sturaro C, Stragapede S, Malfacini D, Compagnin G, Catani M, Fantinati A. (L)-Monomethyl Tyrosine (Mmt): New Synthetic Strategy via Bulky 'Forced-Traceless' Regioselective Pd-Catalyzed C(sp 2)-H Activation. Pharmaceuticals (Basel) 2023; 16:1592. [PMID: 38004457 PMCID: PMC10675785 DOI: 10.3390/ph16111592] [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: 10/03/2023] [Revised: 10/27/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
The enormous influence in terms of bioactivity, affinity, and selectivity represented by the replacement of (L)-2,6-dimethyl tyrosine (Dmt) instead of Phenylalanine (Phe) into Nociceptin/orphanin (N/OFQ) neuropeptide analogues has been well documented in the literature. More recently, the non-natural amino acid (L)-2-methyl tyrosine (Mmt), with steric hindrance included between Tyr and Dmt, has been studied because of the modulation of steric effects in opioid peptide chains. Here, we report a new synthetic strategy to obtain Mmt based on the well-known Pd-catalyzed ortho-C(sp2)-H activation approach, because there is a paucity of other synthetic routes in the literature to achieve it. The aim of this work was to force only the mono-ortho-methylation process over the double ortho-methylation one. In this regard, we are pleased to report that the introduction of the dibenzylamine moiety on a Tyr aromatic nucleus is a convenient and traceless solution to achieve such a goal. Interestingly, our method provided the aimed Mmt either as N-Boc or N-Fmoc derivatives ready to be inserted into peptide chains through solid-phase peptide synthesis (SPPS). Importantly, the introduction of Mmt in place of Phe1 in the sequence of N/OFQ(1-13)-NH2 was very well tolerated in terms of pharmacological profile and bioactivity.
Collapse
Affiliation(s)
- Davide Illuminati
- Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi 213/d, 41125 Modena, Italy;
| | - Claudio Trapella
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy; (C.T.); (V.Z.); (R.G.); (G.C.); (M.C.)
| | - Vinicio Zanirato
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy; (C.T.); (V.Z.); (R.G.); (G.C.); (M.C.)
| | - Remo Guerrini
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy; (C.T.); (V.Z.); (R.G.); (G.C.); (M.C.)
| | - Valentina Albanese
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Chiara Sturaro
- U.O. Neurological Clinic, University Hospital of Ferrara, Via Aldo Moro, 8, 44124 Ferrara, Italy; (C.S.)
| | - Simona Stragapede
- U.O. Neurological Clinic, University Hospital of Ferrara, Via Aldo Moro, 8, 44124 Ferrara, Italy; (C.S.)
| | - Davide Malfacini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via 8 Febbraio, 2, 35131 Padova, Italy;
| | - Greta Compagnin
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy; (C.T.); (V.Z.); (R.G.); (G.C.); (M.C.)
| | - Martina Catani
- Department of Chemical and Pharmaceutical Sciences, University of Ferrara, Via Fossato di Mortara 17, 44121 Ferrara, Italy; (C.T.); (V.Z.); (R.G.); (G.C.); (M.C.)
| | - Anna Fantinati
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy;
| |
Collapse
|
18
|
Todorov P, Assenov B, Angelov D, Dzhambazova E, Pechlivanova D. Behavioral Effects and Analgesic Profile of Hemoglobin-Derived Valorphin and Its Synthetic Analog in Rodents. Biomedicines 2023; 11:2783. [PMID: 37893157 PMCID: PMC10603931 DOI: 10.3390/biomedicines11102783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/09/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Valorphin (V1) is a naturally occurring peptide derived from hemoglobin that has been found to have an affinity for opioid receptors and exhibits antinociceptive and anticonvulsant activity. Some of its synthetic analogs containing an aminophosphonate moiety show structure-dependent potent antinociceptive effects. This study aimed to reveal a detailed picture of the antinociceptive mechanisms and behavioral effects of V1 and its recently synthesized phosphopeptide analog V2p in rodents using a range of methods. The studied peptides significantly reduced acute (mean V1-9.0, V2p-5.8 vs. controls-54.1 s) and inflammatory (mean V1-57.9 and V2p-53.3 vs. controls-107.6 s) nociceptive pain in the formalin test, as well as carrageenan-induced hyperalgesia (mean V1-184.7 and V2p-107.3 vs. controls-61.8 g) in the paw pressure test. These effects are mediated by activation of opioid receptors with a predominance of kappa in V1 antinociception and by delta, kappa, and mu receptors in V2p-induced antinociception. Both peptides did not change the levels of TNF-alpha and IL-1-beta in blood serum. V1 induces depression-like behavior, and V2p shows a tendency toward anxiolysis and short-term impairment of motor coordination without affecting exploratory behavior. The results characterize valorphin and its derivative as promising analgesics that exert their effects both centrally and peripherally, without causing severe behavioral changes in experimental animals. These encouraging data are a foundation for future studies focusing on the effects of hemorphins after long-term treatment.
Collapse
Affiliation(s)
- Petar Todorov
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, 1756 Sofia, Bulgaria;
| | - Borislav Assenov
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 23, 1113 Sofia, Bulgaria; (B.A.); (D.A.)
- Faculty of Medicine, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria;
| | - Dimo Angelov
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 23, 1113 Sofia, Bulgaria; (B.A.); (D.A.)
| | - Elena Dzhambazova
- Faculty of Medicine, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria;
| | - Daniela Pechlivanova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 23, 1113 Sofia, Bulgaria; (B.A.); (D.A.)
- Faculty of Medicine, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria;
| |
Collapse
|
19
|
Bao SS, Tang PF, Gao NY, Xiao ZX, Qian JC, Zheng L, Hu GX, Xu HH. Effect of apatinib on the pharmacokinetics of tramadol and O-desmethyltramadol in rats. PeerJ 2023; 11:e16051. [PMID: 37719112 PMCID: PMC10501372 DOI: 10.7717/peerj.16051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
Since the combination of anticancer drugs and opioids is very common, apatinib and tramadol are likely to be used in combination clinically. This study evaluated the effects of apatinib on the pharmacokinetics of tramadol and its main metabolite O-desmethyltramadol in Sprague-Dawley (SD) rats and the inhibitory effects of apatinib on tramadol in rat liver microsomes (RLMs), human liver microsomes (HLMs) and recombinant human CYP2D6.1. The samples were determined by ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). The in vivo results showed that compared with the control group, apatinib increased the AUC(0-t), AUC(0-∞) and Cmax values of tramadol and O-desmethyltramadol, and decreased the values of VZ/F and CLz/F. In addition, the MRT(0-t), MRT(0-∞) values of O-desmethyltramadol were increased. In vitro, apatinib inhibited the metabolism of tramadol by a mixed way with IC50 of 1.927 µM in RLMs, 2.039 µM in HLMs and 15.32 µM in CYP2D6.1. In summary, according to our findings, apatinib has a strong in vitro inhibitory effect on tramadol, and apatinib can increase the analgesic effect of tramadol and O-desmethyltramadol in rats.
Collapse
Affiliation(s)
- Su-su Bao
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Peng-fei Tang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Nan-yong Gao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhong-xiang Xiao
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jian-chang Qian
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Long Zheng
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| | - Guo-xin Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huan-hai Xu
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
20
|
Lunerti V, Shen Q, Li H, Benvenuti F, Soverchia L, Narendran R, Weiss F, Cannella N, Ciccocioppo R. Cebranopadol, a novel long-acting opioid agonist with low abuse liability, to treat opioid use disorder: Preclinical evidence of efficacy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.21.550008. [PMID: 37546836 PMCID: PMC10401954 DOI: 10.1101/2023.07.21.550008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The gold standard pharmacological treatment for opioid use disorder (OUD) consists of maintenance therapy with long-acting opioid agonists such as buprenorphine and methadone. Despite these compounds having demonstrated substantial efficacy, a significant number of patients do not show optimal therapeutic responses. Moreover, the abuse liability of these medications remains a major concern. Cebranopadol, is a new, long-acting pan-opioid agonist that also activates the nociception/orphanin FQ NOP receptor. Here we used rats to explore the therapeutic potential of this agent in OUD. First, in operant intravenous self-administration experiments we compared the potential abuse liability of cebranopadol with the prototypical opioid heroin. Under a fixed ratio 1 (FR1) contingency, rats maintained responding for heroin (1, 7, 20, 60 μg/inf) to a larger extent than cebranopadol (0.03, 0.1, 0.3, 1.0, 6.0 μg/inf). When the contingency was switched to a progressive ratio (PR) reinforcement schedule, heroin maintained responding at high levels at all except the lowest dose. Conversely, in the cebranopadol groups responding decreased drastically and the break point (BP) did not differ from saline controls. Next, we demonstrated that oral administration of cebranopadol (0, 25, 50 μg/kg) significantly attenuated drug self-administration independent of heroin dose (1, 7, 20, 60 μg/inf). Cebranopadol also reduced the break point for heroin (20 μg/inf). Furthermore, in a heroin self-administration training extinction/reinstatement paradigm, pretreatment with cebranopadol significantly attenuated yohimbine stress-induced reinstatement of drug seeking. Together, these data indicate that cebranopadol has limited abuse liability compared to heroin and is highly efficacious in attenuating opioid self-administration and stress-induced reinstatement, suggesting clinical potential of this compound for OUD treatment.
Collapse
|
21
|
Ullah Q, Ali Z, Rashid U, Ali G, Ahmad N, Khan R, Ullah S, Ayaz M, Murthy HCA. Involvement of the Opioidergic Mechanism in the Analgesic Potential of a Novel Indazolone Derivative: Efficacy in the Management of Pain, Neuropathy, and Inflammation Using In Vivo and In Silico Approaches. ACS OMEGA 2023; 8:22809-22819. [PMID: 37396203 PMCID: PMC10308391 DOI: 10.1021/acsomega.3c01717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/26/2023] [Indexed: 07/04/2023]
Abstract
Indazolones possess interesting pharmacological activities. The search for indazole and indazolone-containing nuclei as drugs is an important research area of medicinal chemistry. The current work aims to evaluate a novel indazolone derivative against in vivo and in silico targets of pain, neuropathy, and inflammation. An indazolone derivative (ID) was synthesized and characterized using advanced spectroscopic techniques. Well-established animal models of abdominal constriction, hot plate, tail immersion, carrageenan paw edema, and Brewer's yeast-induced pyrexia were employed for evaluating the potential of the ID at different doses (20-60 mg kg-1). Nonselective GABA antagonists, opioid antagonist naloxone (NLX) and pentylenetetrazole (PTZ), were employed to assess the potential role of GABAergic and opioidergic processes. The antineuropathic potential of the drug was evaluated using a vincristine-induced neuropathic pain model. In silico studies were performed to assess any possible interactions of the ID with pain target sites like cyclooxygenases (COX-I/II), GABAA, and opioid receptors. This study revealed that the selected ID (doses of 20-60 mg kg-1) efficiently hampered chemically and thermally induced nociceptive responses, producing significant anti-inflammatory and antipyretic effects. These effects produced by the ID were dose-dependent (i.e., 20-60 mg kg-1 and p range of 0.001-0.01) and significant in comparison to standards (p < 0.001). Antagonistic studies with NLX (1.0 mg kg-1) and PTZ (15.0 mg kg-1) revealed the involvement of the opioidergic mechanism rather than the GABAergic mechanism. The ID showed promising anti-static allodynia effects as well. In silico studies revealed preferential binding interactions of the ID with cyclooxygenases (COX-I/II), GABAA, and opioid receptors. According to the results of the current investigation, the ID may serve in the future as a therapeutic agent for the treatment of pyrexia, chemotherapy-induced neuropathic pain, and nociceptive inflammatory pain.
Collapse
Affiliation(s)
- Qarib Ullah
- Department of Chemistry, Hazara University, Mansehra 21310, Pakistan
| | - Zarshad Ali
- Department of Chemistry, Hazara University, Mansehra 21310, Pakistan
| | - Umer Rashid
- Department of Chemistry, COMSATS University Islamabad-Abbottabad Campus, 22060 Abbottabad, Pakistan
| | - Gowhar Ali
- Department of Pharmacy, University of Peshawar, Peshawar 25120, Pakistan
| | - Nisar Ahmad
- School of Pharmacy, Institute of Health Sciences Mardan, Mardan, KPK 23200, Pakistan
| | - Rasool Khan
- Institute of Chemical Sciences, University of Peshawar, Peshawar 25120, Pakistan
| | - Sami Ullah
- Department of Pharmacy, University of Peshawar, Peshawar 25120, Pakistan
| | - Muhammad Ayaz
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Dir (L), Chakdara, KP 18000, Pakistan
| | - H C Ananda Murthy
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, P O Box 1888, Adama, Oromia 1888, Ethiopia
- Department of Prosthodontics, Saveetha Dental College & Hospital, Saveetha Institute of Medical and Technical Science (SIMATS), Saveetha University, Chennai 600077, Tamil Nadu, India
| |
Collapse
|
22
|
Alam MR, Singh S. Neuromodulation in Parkinson's disease targeting opioid and cannabinoid receptors, understanding the role of NLRP3 pathway: a novel therapeutic approach. Inflammopharmacology 2023:10.1007/s10787-023-01259-0. [PMID: 37318694 DOI: 10.1007/s10787-023-01259-0] [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: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 06/16/2023]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disorder characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta, resulting in motor and non-motor symptoms. Although levodopa is the primary medication for PD, its long-term use is associated with complications such as dyskinesia and drug resistance, necessitating novel therapeutic approaches. Recent research has highlighted the potential of targeting opioid and cannabinoid receptors as innovative strategies for PD treatment. Modulating opioid transmission, particularly through activating µ (MOR) and δ (DOR) receptors while inhibiting κ (KOR) receptors, shows promise in preventing motor complications and reducing L-DOPA-induced dyskinesia. Opioids also possess neuroprotective properties and play a role in neuroprotection and seizure control. Similar to this, endocannabinoid signalling via CB1 and CB2 receptors influences the basal ganglia and may contribute to PD pathophysiology, making it a potential therapeutic target. In addition to opioid and cannabinoid receptor targeting, the NLRP3 pathway, implicated in neuroinflammation and neurodegeneration, emerges as another potential therapeutic avenue for PD. Recent studies suggest that targeting this pathway holds promise as a therapeutic strategy for PD management. This comprehensive review focuses on neuromodulation and novel therapeutic approaches for PD, specifically highlighting the targeting of opioid and cannabinoid receptors and the NLRP3 pathway. A better understanding of these mechanisms has the potential to enhance the quality of life for PD patients.
Collapse
Affiliation(s)
- Md Reyaz Alam
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
23
|
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: 1.0] [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
|
24
|
Imam MZ, Kuo A, Ghassabian S, Cai Y, Qin Y, Li T, Smith MT. CYX-5, a G-protein biassed MOP receptor agonist, DOP receptor antagonist and KOP receptor agonist, evokes constipation but not respiratory depression relative to morphine in rats. Pharmacol Rep 2023; 75:634-646. [PMID: 36637684 PMCID: PMC10227131 DOI: 10.1007/s43440-023-00446-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/14/2023]
Abstract
BACKGROUND Strong opioid analgesics such as morphine alleviate moderate to severe acute nociceptive pain (e.g. post-surgical or post-trauma pain) as well as chronic cancer pain. However, they evoke many adverse effects and so there is an unmet need for opioid analgesics with improved tolerability. Recently, a prominent hypothesis has been that opioid-related adverse effects are mediated by β-arrestin2 recruitment at the µ-opioid (MOP) receptor and this stimulated research on discovery of G-protein biassed opioid analgesics. In other efforts, opioids with MOP agonist and δ-opioid (DOP) receptor antagonist profiles are promising for reducing side effects c.f. morphine. Herein, we report on the in vivo pharmacology of a novel opioid peptide (CYX-5) that is a G-protein biassed MOP receptor agonist, DOP receptor antagonist and kappa opioid (KOP) receptor agonist. METHODS Male Sprague-Dawley received intracerebroventricular bolus doses of CYX-5 (3, 10, 20 nmol), morphine (100 nmol) or vehicle, and antinociception (tail flick) was assessed relative to constipation (charcoal meal and castor oil-induced diarrhoea tests) and respiratory depression (whole body plethysmography). RESULTS CYX-5 evoked naloxone-sensitive, moderate antinociception, at the highest dose tested. Although CYX-5 did not inhibit gastrointestinal motility, it reduced stool output markedly in the castor oil-induced diarrhoea test. In contrast to morphine that evoked respiratory depression, CYX-5 increased tidal volume, thereby stimulating respiration. CONCLUSION Despite its lack of recruitment of β-arrestin2 at MOP, DOP and KOP receptors, CYX-5 evoked constipation, implicating a mechanism other than β-arrestin2 recruitment at MOP, DOP and KOP receptors, mediating constipation evoked by CYX-5 and potentially other opioid ligands.
Collapse
Affiliation(s)
- Mohammad Zafar Imam
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Andy Kuo
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Sussan Ghassabian
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia Campus, Brisbane, QLD, 4072, Australia
| | - Yunxin Cai
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yajuan Qin
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tingyou Li
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Maree T Smith
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia Campus, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
25
|
Gaborit M, Massotte D. Therapeutic potential of opioid receptor heteromers in chronic pain and associated comorbidities. Br J Pharmacol 2023; 180:994-1013. [PMID: 34883528 DOI: 10.1111/bph.15772] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/07/2021] [Accepted: 07/21/2021] [Indexed: 11/27/2022] Open
Abstract
Chronic pain affects 20% to 45% of the global population and is often associated with the development of anxio-depressive disorders. Treatment of this debilitating condition remains particularly challenging with opioids prescribed to alleviate moderate to severe pain. However, despite strong antinociceptive properties, numerous adverse effects limit opioid use in the clinic. Moreover, opioid misuse and abuse have become a major health concern worldwide. This prompted efforts to design original strategies that would efficiently and safely relieve pain. Targeting of opioid receptor heteromers is one of these. This review summarizes our current knowledge on the role of heteromers involving opioid receptors in the context of chronic pain and anxio-depressive comorbidities. It also examines how heteromerization in native tissue affects ligand binding, receptor signalling and trafficking properties. Finally, the therapeutic potential of ligands designed to specifically target opioid receptor heteromers is considered. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Marion Gaborit
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Dominique Massotte
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| |
Collapse
|
26
|
Smith MT, Kong D, Kuo A, Imam MZ, Williams CM. Multitargeted Opioid Ligand Discovery as a Strategy to Retain Analgesia and Reduce Opioid-Related Adverse Effects. J Med Chem 2023; 66:3746-3784. [PMID: 36856340 DOI: 10.1021/acs.jmedchem.2c01695] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
The global "opioid crisis" has placed enormous pressure on the opioid ligand discovery community to produce novel opioid analgesics with superior opioid-related adverse-effect profiles compared with morphine. In this Perspective, the multitargeted opioid ligand strategy for the discovery of opioid analgesics with superior preclinical therapeutic indices relative to morphine is reviewed and discussed. Dual-targeted μ-opioid (MOP)/δ-opioid (DOP) ligands in which the in vitro DOP antagonist potency at least equals that of the MOP agonist activity, and are devoid of DOP or κ-opioid (KOP) agonist activity, are sufficiently promising candidates to warrant further investigation. Dual-targeted MOP/NOP partial agonists have superior preclinical therapeutic indices to morphine and/or fentanyl in nonhuman primates and are also considered promising. Based on the poor preclinical and clinical therapeutic indices of cebranopadol, which is a full agonist at MOP, DOP, and NOP receptors and a partial agonist at the KOP receptor, this pharmacologic template should be avoided.
Collapse
|
27
|
Goff J, Hina M, Malik N, McLardy H, Reilly F, Robertson M, Ruddy L, Willox F, Forget P. Can Opioid-Free Anaesthesia Be Personalised? A Narrative Review. J Pers Med 2023; 13:jpm13030500. [PMID: 36983682 PMCID: PMC10056629 DOI: 10.3390/jpm13030500] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Background: A significant amount of evidence suggests that Opioid-Free Anaesthesia (OFA) may provide better outcomes for patients undergoing surgery, sparing patients who are particularly vulnerable to adverse side effects of opioids. However, to what extent personalizing OFA is feasible and beneficial has not been adequately described. Methods: We conducted a narrative literature review aiming to provide a comprehensive understanding of nociception and pain and its context within the field of OFA. Physiological (including monitoring), pharmacological, procedural (type of surgery), genetical and phenotypical (including patients’ conditions) were considered. Results: We did not find any monitoring robustly associated with improved outcomes. However, we found evidence supporting particular OFA indications, such as bariatric and cancer surgery. We found that vulnerable patients may benefit more from OFA, with an interesting field of research in patients suffering from vascular disease. We found a variety of techniques and medications making it impossible to consider OFA as a single technique. Our findings suggest that a vast field of research remains unexplored. In particular, a deeper understanding of nociception with an interest in its genetic and acquired contributors would be an excellent starting point paving the way for personalised OFA. Conclusion: Recent developments in OFA may present a more holistic approach, challenging the use of opioids. Understanding better nociception, given the variety of OFA techniques, may help to maximize their potential in different contexts and potential indications.
Collapse
Affiliation(s)
- Jenna Goff
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Morgan Hina
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Nayaab Malik
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Hannah McLardy
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Finley Reilly
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Matthew Robertson
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
- Correspondence:
| | - Louis Ruddy
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Faith Willox
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Patrice Forget
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
- Department of Anaesthesia, NHS Grampian, Aberdeen AB25 2ZD, UK
- Pain AND Opioids after Surgery (PANDOS) European Society of Anaesthesia and Intensive Care (ESAIC) Research Group, 1000 Brussels, Belgium
| |
Collapse
|
28
|
Hennessy MR, Gutridge AM, French AR, Rhoda ES, Meqbil YJ, Gill M, Kashyap Y, Appourchaux K, Paul B, Wang ZJ, van Rijn RM, Riley AP. Modified Akuamma Alkaloids with Increased Potency at the Mu-opioid Receptor. J Med Chem 2023; 66:3312-3326. [PMID: 36827198 PMCID: PMC10037270 DOI: 10.1021/acs.jmedchem.2c01707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Akuammine (1) and pseudoakuammigine (2) are indole alkaloids found in the seeds of the akuamma tree (Picralima nitida). Both alkaloids are weak agonists of the mu opioid receptor (μOR); however, they produce minimal effects in animal models of antinociception. To probe the interactions of 1 and 2 at the opioid receptors, we have prepared a collection of 22 semisynthetic derivatives. Evaluation of this collection at the μOR and kappa opioid receptor (κOR) revealed structural-activity relationship trends and derivatives with improved potency at the μOR. Most notably, the introduction of a phenethyl moiety to the N1 of 2 produces a 70-fold increase in potency and a 7-fold increase in selectivity for the μOR. The in vitro potency of this compound resulted in increased efficacy in the tail-flick and hot-plate assays of antinociception. The improved potency of these derivatives highlights the promise of exploring natural product scaffolds to probe the opioid receptors.
Collapse
Affiliation(s)
- Madeline R Hennessy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612 USA
| | - Anna M Gutridge
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907 USA
| | - Alexander R French
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907 USA
- Departments of Neurology and Bioengineering, University of Illinois Chicago, Chicago, IL 60612 USA
| | - Elizabeth S Rhoda
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907 USA
| | - Yazan J. Meqbil
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907 USA
| | - Meghna Gill
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612 USA
| | - Yavnika Kashyap
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612 USA
| | - Kevin Appourchaux
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Barnali Paul
- Center for Clinical Pharmacology, University of Health Sciences & Pharmacy at St. Louis and Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Zaijie Jim Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612 USA
- Departments of Neurology and Bioengineering, University of Illinois Chicago, Chicago, IL 60612 USA
| | - Richard M. van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907 USA
- Purdue Institute for Drug Discovery, West Lafayette, IN 47907 USA
- Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907 USA
- Purdue Interdisciplinary Life Sciences Graduate Program, Purdue University, West Lafayette, IN 47907 USA
| | - Andrew P. Riley
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612 USA
| |
Collapse
|
29
|
Frolov NA, Vereshchagin AN. Piperidine Derivatives: Recent Advances in Synthesis and Pharmacological Applications. Int J Mol Sci 2023; 24:2937. [PMID: 36769260 PMCID: PMC9917539 DOI: 10.3390/ijms24032937] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
Piperidines are among the most important synthetic fragments for designing drugs and play a significant role in the pharmaceutical industry. Their derivatives are present in more than twenty classes of pharmaceuticals, as well as alkaloids. The current review summarizes recent scientific literature on intra- and intermolecular reactions leading to the formation of various piperidine derivatives: substituted piperidines, spiropiperidines, condensed piperidines, and piperidinones. Moreover, the pharmaceutical applications of synthetic and natural piperidines were covered, as well as the latest scientific advances in the discovery and biological evaluation of potential drugs containing piperidine moiety. This review is designed to help both novice researchers taking their first steps in this field and experienced scientists looking for suitable substrates for the synthesis of biologically active piperidines.
Collapse
Affiliation(s)
| | - Anatoly N. Vereshchagin
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky Prospect 47, 119991 Moscow, Russia
| |
Collapse
|
30
|
Han Z, Jin G, Tang J, Wang H, Guo D, Zhang J. Analgesic tolerance and cross-tolerance to the bifunctional opioid/neuropeptide FF receptors agonist EN-9 and μ-opioid receptor ligands at the supraspinal level in mice. Neuropeptides 2023; 97:102309. [PMID: 36410163 DOI: 10.1016/j.npep.2022.102309] [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: 05/07/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022]
Abstract
The chimeric peptide EN-9 was reported as a κ-opioid/neuropeptide FF receptors bifunctional agonist that modulated chronic pain with no tolerance. Many lines of evidence have shown that the effect of the κ-opioid receptor is mediated by not only its specific activation but also downstream events participation, especially interaction with the μ-opioid receptor pathway in antinociception and tolerance on most occasions. The present study investigated the acute and chronic cross-tolerance of EN-9 with μ-opioid receptor agonist EM-2, DAMGO, and morphine after intracerebroventricularly (i.c.v) injection in the mouse tail-flick test. In the acute tolerance test, EN-9 showed symmetrical acute cross-tolerance to DAMGO but no cross-tolerance to EM2. In the chronic tolerance test, EN-9 had no tolerance after eight days of repeated administration. However, EN-9 illustrated complete cross-tolerance to morphine and symmetrical cross-tolerance to EM2. In addition, inhibition of NPFF receptor could induce the tolerance development of EN-9. These findings indicated that supraspinal EN-9-induced antinociception contains additional components, which are mediated by the downstream μ-opioid receptor pathway both in acute and chronic treatment, whereas the subtypes of μ-opioid receptor or NPFF system pathway involved in antinociceptive effects induced by EN-9 are complex. Identifying the receptor mechanism could help design preferable bifunctional opioid compounds.
Collapse
Affiliation(s)
- Zhenglan Han
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, 55 Dongshun Road, Nanchong 637100, PR China
| | - Guofei Jin
- Nanchong Key Laboratory of Metabolic Drugs and Biological Products, 55 Dongshun Road, Nanchong 637100, PR China
| | - Jiancai Tang
- Nanchong Key Laboratory of Metabolic Drugs and Biological Products, 55 Dongshun Road, Nanchong 637100, PR China
| | - Hanyan Wang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, 55 Dongshun Road, Nanchong 637100, PR China
| | - Dongmei Guo
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, 55 Dongshun Road, Nanchong 637100, PR China
| | - Jingping Zhang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, 55 Dongshun Road, Nanchong 637100, PR China.
| |
Collapse
|
31
|
Wang Y, Zhuang Y, DiBerto JF, Zhou XE, Schmitz GP, Yuan Q, Jain MK, Liu W, Melcher K, Jiang Y, Roth BL, Xu HE. Structures of the entire human opioid receptor family. Cell 2023; 186:413-427.e17. [PMID: 36638794 DOI: 10.1016/j.cell.2022.12.026] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/11/2022] [Accepted: 12/13/2022] [Indexed: 01/13/2023]
Abstract
Opioids are effective analgesics, but their use is beset by serious side effects, including addiction and respiratory depression, which contribute to the ongoing opioid crisis. The human opioid system contains four opioid receptors (μOR, δOR, κOR, and NOPR) and a set of related endogenous opioid peptides (EOPs), which show distinct selectivity toward their respective opioid receptors (ORs). Despite being key to the development of safer analgesics, the mechanisms of molecular recognition and selectivity of EOPs to ORs remain unclear. Here, we systematically characterize the binding of EOPs to ORs and present five structures of EOP-OR-Gi complexes, including β-endorphin- and endomorphin-bound μOR, deltorphin-bound δOR, dynorphin-bound κOR, and nociceptin-bound NOPR. These structures, supported by biochemical results, uncover the specific recognition and selectivity of opioid peptides and the conserved mechanism of opioid receptor activation. These results provide a structural framework to facilitate rational design of safer opioid drugs for pain relief.
Collapse
Affiliation(s)
- Yue Wang
- The CAS Key Laboratory of Receptor Research and the State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Youwen Zhuang
- The CAS Key Laboratory of Receptor Research and the State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Jeffrey F DiBerto
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - X Edward Zhou
- Department of Structural Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Gavin P Schmitz
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Qingning Yuan
- The CAS Key Laboratory of Receptor Research and the State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; The Shanghai Advanced Electron Microscope Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Manish K Jain
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA
| | - Weiyi Liu
- The CAS Key Laboratory of Receptor Research and the State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Karsten Melcher
- Department of Structural Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Yi Jiang
- The CAS Key Laboratory of Receptor Research and the State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Lingang Laboratory, Shanghai 200031, China
| | - Bryan L Roth
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599, USA.
| | - H Eric Xu
- The CAS Key Laboratory of Receptor Research and the State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
32
|
Ma H, Pagare PP, Li M, Neel LT, Mendez RE, Gillespie JC, Stevens DL, Dewey WL, Selley DE, Zhang Y. Structural Alterations of the "Address" Moiety of NAN Leading to the Discovery of a Novel Opioid Receptor Modulator with Reduced hERG Toxicity. J Med Chem 2023; 66:577-595. [PMID: 36538027 PMCID: PMC10546487 DOI: 10.1021/acs.jmedchem.2c01499] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The search for selective opioid ligands with desired pharmacological potency and improved safety profile has always been an area of interest. Our previous effort yielded a potent opioid modulator, NAN, a 6α-N-7'-indolyl-substituted naltrexamine derivative, which exhibited promising pharmacological activities both in vitro and in vivo. However, significant human ether-a-go-go-related gene (hERG) liability limited its further development. Therefore, a systematic structural modification on NAN was conducted in order to alleviate hERG toxicity while preserving pharmacological properties, which led to the discovery of 2'-methylindolyl derivative compound 21. Compared to NAN, compound 21 manifested overall improved pharmacological profiles. Follow-up hERG channel inhibition evaluation revealed a seven-fold decreased potency of compound 21 compared to NAN. Furthermore, several fundamental drug-like property evaluations suggested a reasonable ADME profile of 21. Collectively, compound 21 appeared to be a promising opioid modulator for further development as a novel therapeutic agent toward opioid use disorder treatments.
Collapse
Affiliation(s)
- Hongguang Ma
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia23298, United States
| | - Piyusha P Pagare
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia23298, United States
| | - Mengchu Li
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia23298, United States
| | - Logan T Neel
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia23298, United States
| | - Rolando E Mendez
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia23298, United States
| | - James C Gillespie
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia23298, United States
| | - David L Stevens
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia23298, United States
| | - William L Dewey
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia23298, United States
| | - Dana E Selley
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 410 North 12th Street, Richmond, Virginia23298, United States
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, 800 E Leigh Street, Richmond, Virginia23298, United States
- Institute for Drug and Alcohol Studies, 203 East Cary Street, Richmond, Virginia23298-0059, United States
| |
Collapse
|
33
|
A Novel Multi-Target Mu/Delta Opioid Receptor Agonist, HAGD, Produced Potent Peripheral Antinociception with Limited Side Effects in Mice and Minimal Impact on Human Sperm Motility In Vitro. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28010427. [PMID: 36615612 PMCID: PMC9824695 DOI: 10.3390/molecules28010427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023]
Abstract
Pain is a common clinical symptom among patients. Although various opioid analgesics have been developed, their side effects hinder their application. This study aimed to develop a novel opioid analgesic, HAGD (H-Tyr-D-AIa-GIy-Phe-NH2), with limited side effects. In vivo studies on mouse models as well as in vitro studies on Chinese hamster ovary (CHO) cells expressing human mu, delta, or kappa opioid receptors (CHOhMOP, CHOhDOP, and CHOhKOP, respectively) and human sperm were conducted. Compared with subcutaneous morphine (10 mg/kg), subcutaneous HAGD (10 mg/kg) produced equipotent or even greater antinociception with a prolonged duration by activating mu/delta opioid receptors in preclinical mouse pain models. The analgesic tolerance, rewarding effects (i.e., conditioned place preference and acute hyperlocomotion), and gastrointestinal transit inhibition of HAGD were significantly reduced compared with those of morphine. Both HAGD and morphine exhibited a withdrawal response and had no impacts on motor coordination. In CHOhMOP and CHOhDOP, HAGD showed specific and efficient intracellular Ca2+ stimulation. HAGD had minimal impact on human sperm motility in vitro, whereas 1 × 10-7 and 1 × 10-8 mol/L of morphine significantly declined sperm motility at 3.5 h. Overall, HAGD may serve as a promising antinociceptive compound.
Collapse
|
34
|
Neale KJ, Weimer MB, Davis MP, Jones KF, Kullgren JG, Kale SS, Childers J, Broglio K, Merlin JS, Peck S, Francis SY, Bango J, Jones CA, Sager Z, Ho JJ. Top Ten Tips Palliative Care Clinicians Should Know About Buprenorphine. J Palliat Med 2023; 26:120-130. [PMID: 36067137 DOI: 10.1089/jpm.2022.0399] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Pain management in palliative care (PC) is becoming more complex as patients survive longer with life-limiting illnesses and population-wide trends involving opioid misuse become more common in serious illness. Buprenorphine, a generally safe partial mu-opioid receptor agonist, has been shown to be effective for both pain management and opioid use disorder. It is critical that PC clinicians become comfortable with indications for its use, strategies for initiation while understanding risks and benefits. This article, written by a team of PC and addiction-trained specialists, including physicians, nurse practitioners, social workers, and a pharmacist, offers 10 tips to demystify buprenorphine use in serious illness.
Collapse
Affiliation(s)
- Kyle J Neale
- The Lois U and Harry R Horvitz Palliative Medicine Program, Department of Palliative Medicine and Supportive Care, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Melissa B Weimer
- Program in Addiction Medicine, Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Mellar P Davis
- Department of Palliative Care, Geisinger Medical Center, Danville, Pennsylvania, USA
| | | | - Justin G Kullgren
- Palliative Medicine Clinical Pharmacy, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Sachin S Kale
- Division of Palliative Medicine, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Julie Childers
- Section of Palliative Care and Medical Ethics, Division of General Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kathleen Broglio
- Section of Palliative Medicine, Geisel School of Medicine at Dartmouth, Collaboratory for Implementation Sciences at Dartmouth, Lebanon, New Hampshire, USA
| | - Jessica S Merlin
- Section of Palliative Care and Medical Ethics and Palliative Research Center, Division of General Internal Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sarah Peck
- Division of Palliative Medicine, Emory University Healthcare Midtown, Atlanta, Georgia, USA
| | - Sheria Y Francis
- Collaborative Care Management, University of Pittsburgh Medical Center Presbyterian Shadyside, Pittsburgh, Pennsylvania, USA
| | | | - Christopher A Jones
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Zachary Sager
- Department of Psychosocial Oncology and Palliative Care, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - J Janet Ho
- Division of Palliative Medicine, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
35
|
Li M, Stevens DL, Arriaga M, Townsend EA, Mendez RE, Blajkevch NA, Selley DE, Banks ML, Negus SS, Dewey WL, Zhang Y. Characterization of a Potential KOR/DOR Dual Agonist with No Apparent Abuse Liability via a Complementary Structure-Activity Relationship Study on Nalfurafine Analogues. ACS Chem Neurosci 2022; 13:3608-3628. [PMID: 36449691 PMCID: PMC10243363 DOI: 10.1021/acschemneuro.2c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Discovery of analgesics void of abuse liability is critical to battle the opioid crisis in the United States. Among many strategies to achieve this goal, targeting more than one opioid receptor seems promising to minimize this unwanted side effect while achieving a reasonable therapeutic profile. In the process of understanding the structure-activity relationship of nalfurafine, we identified a potential analgesic agent, NMF, as a dual kappa opioid receptor/delta opioid receptor agonist with minimum abuse liability. Further characterizations, including primary in vitro ADMET studies (hERG toxicity, plasma protein binding, permeability, and hepatic metabolism), and in vivo pharmacodynamic and toxicity profiling (time course, abuse liability, tolerance, withdrawal, respiratory depression, body weight, and locomotor activity) further confirmed NMF as a promising drug candidate for future development.
Collapse
Affiliation(s)
- Mengchu Li
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298
| | - David L. Stevens
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - Michelle Arriaga
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - E. Andrew Townsend
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - Rolando E. Mendez
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - Nadejda A. Blajkevch
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - Dana E. Selley
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - Matthew L. Banks
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - S. Stevens Negus
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - William L. Dewey
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298
| | - Yan Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298
- Institute for Drug and Alcohol Studies, 203 East Cary Street, Virginia Commonwealth University, Richmond, VA 23298
| |
Collapse
|
36
|
Li F, Kopajtic TA, Katz JL, Luo D, Prisinzano TE, Imler GH, Deschamps JR, Jacobson AE, Rice KC. Synthesis and Pharmacological Evaluation of Enantiopure N-Substituted Ortho-c Oxide-Bridged 5-Phenylmorphans. Molecules 2022; 27:molecules27248808. [PMID: 36557961 PMCID: PMC9785231 DOI: 10.3390/molecules27248808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/15/2022] Open
Abstract
The design of enantiopure stereoisomers of N-2-phenylcyclopropylmethyl-substituted ortho-c oxide-bridged phenylmorphans, the E and Z isomers of an N-cinnamyl moiety, and N-propyl enantiomers were based on combining the most potent oxide-bridged phenylmorphan (the ortho-c isomer) with the most potent N-substituent that we previously found with a 5-(3-hydroxy)phenylmorphan (i.e., N-2-phenylcyclopropyl methyl moieties, N-cinnamyl, and N-propyl substituents). The synthesis of the eight enantiopure N-2-phenylcyclopropylmethyl ortho-c oxide-bridged phenylmorphans and six additional enantiomers of the N-substituted ortho-c oxide-bridged phenylmorphans (N-E and Z-cinnamyl compounds, and N-propyl compounds) was accomplished. The synthesis started from common intermediates (3R,6aS,11aS)-10-methoxy-1,3,4,5,6,11a-hexahydro-2H-3,6a-methano-benzofuro[2,3-c]azocine (+)-6 and its enantiomer, (3S, 6aR, 11aR)-(-)-6, respectively. The enantiomers of ±-6 were obtained through salt formation with (S)-(+)- and (R)-(-)-p-methylmandelic acid, and the absolute configuration of the (R)-(-)-p-methylmandelate salt of (3S, 6aR, 11aR)-(-)-6 was determined by single-crystal X-ray analysis. The enantiomeric secondary amines were reacted with N-(2-phenylcyclopropyl)methyl derivatives, 2-(E)-cinnamyl bromide, and (Z)-3-phenylacrylic acid. These products led to all of the desired N-derivatives of the ortho-c oxide-bridged phenylmorphans. Their opioid receptor binding affinity was measured. The compounds with MOR affinity < 50 nM were examined for their functional activity in the forskolin-induced cAMP accumulation assay. Only the enantiomer of the N-phenethyl ortho-c oxide-bridged phenylmorphan ((-)-1), and only the (3S,6aR,11aR)-2-(((1S,2S)-2-phenylcyclopropyl)methyl)-1,3,4,5,6,11a-hexahydro-2H-3,6a-methanobenzofuro[2,3-c]azocin-10-ol isomer ((+)-17), and the N-phenylpropyl derivative ((-)-25) had opioid binding affinity < 50 nM. Both (-)-1 and (-)-25 were partial agonists in the cAMP assay, with the former showing high potency and low efficacy, and the latter with lower potency and less efficacy. Most interesting was the N-2-phenylcyclopropylmethyl (3S,6aR,11aR)-2-(1S,2S)-enantiomer ((+)-17). That compound had good MOR binding affinity (Ki = 11.9 nM) and was found to have naltrexone-like potency as a MOR antagonist (IC50 = 6.92 nM).
Collapse
Affiliation(s)
- Fuying Li
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, Rockville, MD 20852, USA
| | - Theresa A. Kopajtic
- Psychobiology Section, Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jonathan L. Katz
- Psychobiology Section, Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Dan Luo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Thomas E. Prisinzano
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Gregory H. Imler
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA
| | - Jeffrey R. Deschamps
- Center for Biomolecular Science and Engineering, Naval Research Laboratory, Washington, DC 20375, USA
| | - Arthur E. Jacobson
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, Rockville, MD 20852, USA
- Correspondence: (A.E.J.); (K.C.R.); Tel.: +1-301-451-5028 (A.E.J.); +1-301-451-4799 (K.C.R.)
| | - Kenner C. Rice
- Drug Design and Synthesis Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Department of Health and Human Services, Rockville, MD 20852, USA
- Correspondence: (A.E.J.); (K.C.R.); Tel.: +1-301-451-5028 (A.E.J.); +1-301-451-4799 (K.C.R.)
| |
Collapse
|
37
|
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
|
38
|
Gerak LR, Maguire DR, Cami-Kobeci G, Olson KM, Traynor JR, Husbands SM, France CP, Acevedo L, Belli B, Flynn P. OREX-1038: a potential new treatment for pain with low abuse liability and limited adverse effects. Behav Pharmacol 2022; 33:377-394. [PMID: 35947066 PMCID: PMC9371589 DOI: 10.1097/fbp.0000000000000684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Drugs targeting mu opioid receptors are the mainstay of clinical practice for treating moderate-to-severe pain. While they can offer excellent analgesia, their use can be limited by adverse effects, including constipation, respiratory depression, tolerance, and abuse liability. Multifunctional ligands acting at mu opioid and nociceptin/orphanin FQ peptide receptors might provide antinociception with substantially improved adverse-effect profiles. This study explored one of these ligands, OREX-1038 (BU10038), in several assays in rodents and nonhuman primates. Binding and functional studies confirmed OREX-1038 to be a low-efficacy agonist at mu opioid and nociceptin/orphanin FQ peptide receptors and an antagonist at delta and kappa opioid receptors with selectivity for opioid receptors over other proteins. OREX-1038 had long-acting antinociceptive effects in postsurgical and complete Freund's adjuvant (CFA)-induced thermal hyperalgesia assays in rats and a warm water tail-withdrawal assay in monkeys. OREX-1038 was active for at least 24 h in each antinociception assay, and its effects in monkeys did not diminish over 22 days of daily administration. This activity was coupled with limited effects on physiological signs (arterial pressure, heart rate, and body temperature) and no evidence of withdrawal after administration of naltrexone or discontinuation of treatment in monkeys receiving OREX-1038 daily. Over a range of doses, OREX-1038 was only transiently self-administered, which diminished rapidly to nonsignificant levels; overall, both OREX-1038 and buprenorphine maintained less responding than remifentanil. These results support the concept of dual mu and nociceptin/orphanin FQ peptide receptor partial agonists having improved pharmacological profiles compared with opioids currently used to treat pain.
Collapse
Affiliation(s)
- Lisa R Gerak
- Department of Pharmacology, University of Texas Health Science Center at San Antonio
- Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David R Maguire
- Department of Pharmacology, University of Texas Health Science Center at San Antonio
- Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Gerta Cami-Kobeci
- Department of Pharmacy and Pharmacology, University of Bath, Bath
- Current address: School of Life Sciences, University of Bedfordshire, Luton, UK
| | - Keith M Olson
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - John R Traynor
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Stephen M Husbands
- Department of Pharmacy and Pharmacology, University of Bath, Bath
- Centre for Therapeutic Innovation, University of Bath, Bath, UK
| | - Charles P France
- Department of Pharmacology, University of Texas Health Science Center at San Antonio
- Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
- Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | | | - Barbara Belli
- Orexigen Therapeutics Inc., La Jolla, California, USA
| | - Peter Flynn
- Orexigen Therapeutics Inc., La Jolla, California, USA
| |
Collapse
|
39
|
McLaughlin JP, Rayala R, Bunnell AJ, Tantak MP, Eans SO, Nefzi K, Ganno ML, Dooley CT, Nefzi A. Bis-Cyclic Guanidine Heterocyclic Peptidomimetics as Opioid Ligands with Mixed μ-, κ- and δ-Opioid Receptor Interactions: A Potential Approach to Novel Analgesics. Int J Mol Sci 2022; 23:9623. [PMID: 36077029 PMCID: PMC9455983 DOI: 10.3390/ijms23179623] [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: 08/11/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/25/2022] Open
Abstract
The design and development of analgesics with mixed-opioid receptor interactions has been reported to decrease side effects, minimizing respiratory depression and reinforcing properties to generate safer analgesic therapeutics. We synthesized bis-cyclic guanidine heterocyclic peptidomimetics from reduced tripeptides. In vitro screening with radioligand competition binding assays demonstrated variable affinity for the mu-opioid receptor (MOR), delta-opioid receptor (DOR), and kappa-opioid receptor (KOR) across the series, with compound 1968-22 displaying good affinity for all three receptors. Central intracerebroventricular (i.c.v.) administration of 1968-22 produced dose-dependent, opioid receptor-mediated antinociception in the mouse 55 °C warm-water tail-withdrawal assay, and 1968-22 also produced significant antinociception up to 80 min after oral administration (10 mg/kg, p.o.). Compound 1968-22 was detected in the brain 5 min after intravenous administration and was shown to be stable in the blood for at least 30 min. Central administration of 1968-22 did not produce significant respiratory depression, locomotor effects or conditioned place preference or aversion. The data suggest these bis-cyclic guanidine heterocyclic peptidomimetics with multifunctional opioid receptor activity may hold potential as new analgesics with fewer liabilities of use.
Collapse
Affiliation(s)
- Jay P. McLaughlin
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - Ramanjaneyulu Rayala
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Ashley J. Bunnell
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Mukund P. Tantak
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| | - Shainnel O. Eans
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - Khadija Nefzi
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - Michelle L. Ganno
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Colette T. Dooley
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, FL 34987, USA
| | - Adel Nefzi
- Center for Translational Science, Florida International University, Port St. Lucie, FL 34987, USA
| |
Collapse
|
40
|
Dalefield ML, Scouller B, Bibi R, Kivell BM. The Kappa Opioid Receptor: A Promising Therapeutic Target for Multiple Pathologies. Front Pharmacol 2022; 13:837671. [PMID: 35795569 PMCID: PMC9251383 DOI: 10.3389/fphar.2022.837671] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Kappa-opioid receptors (KOR) are widely expressed throughout the central nervous system, where they modulate a range of physiological processes depending on their location, including stress, mood, reward, pain, inflammation, and remyelination. However, clinical use of KOR agonists is limited by adverse effects such as dysphoria, aversion, and sedation. Within the drug-development field KOR agonists have been extensively investigated for the treatment of many centrally mediated nociceptive disorders including pruritis and pain. KOR agonists are potential alternatives to mu-opioid receptor (MOR) agonists for the treatment of pain due to their anti-nociceptive effects, lack of abuse potential, and reduced respiratory depressive effects, however, dysphoric side-effects have limited their widespread clinical use. Other diseases for which KOR agonists hold promising therapeutic potential include pruritis, multiple sclerosis, Alzheimer's disease, inflammatory diseases, gastrointestinal diseases, cancer, and ischemia. This review highlights recent drug-development efforts targeting KOR, including the development of G-protein-biased ligands, mixed opioid agonists, and peripherally restricted ligands to reduce side-effects. We also highlight the current KOR agonists that are in preclinical development or undergoing clinical trials.
Collapse
Affiliation(s)
| | | | | | - Bronwyn M. Kivell
- Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
41
|
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: 0.5] [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
|
42
|
Castroman P, Quiroga O, Mayoral Rojals V, Gómez M, Moka E, Pergolizzi Jr J, Varrassi G. Reimagining How We Treat Acute Pain: A Narrative Review. Cureus 2022; 14:e23992. [PMID: 35547466 PMCID: PMC9084930 DOI: 10.7759/cureus.23992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/06/2022] [Indexed: 11/25/2022] Open
Abstract
Acute pain may be influenced by biopsychosocial factors. Conditioned pain modulation, distraction, peripheral nerve stimulation, and cryoneurolysis may be helpful in its treatment. New developments in opioids, such as opioids with bifunctional targets and oliceridine, may be particularly suited for acute pain care. Allosteric modulators can enhance receptor subtype selectivity, offering analgesia with fewer and/or less severe side effects. Neuroinflammation in acute pain is caused by direct insult to the central nervous system and is distinct from neuroinflammation in degenerative disorders. Pharmacologic agents targeting the neuroinflammatory process are limited at this time. Postoperative pain is a prevalent form of acute pain and must be recognized as a global public health challenge. This type of pain may be severe, impede rehabilitation, and is often under-treated. A subset of surgical patients develops chronic postsurgical pain. Acute pain is not just temporally limited pain that often resolves on its own. It is an important subject for further research as acute pain may transition into more damaging and debilitating chronic pain. Reimagining how we treat acute pain will help us better address this urgent unmet medical need.
Collapse
|
43
|
Mizobuchi Y, Miyano K, Manabe S, Uezono E, Komatsu A, Kuroda Y, Nonaka M, Matsuoka Y, Sato T, Uezono Y, Morimatsu H. Ketamine Improves Desensitization of µ-Opioid Receptors Induced by Repeated Treatment with Fentanyl but Not with Morphine. Biomolecules 2022; 12:426. [PMID: 35327617 PMCID: PMC8946650 DOI: 10.3390/biom12030426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
The issue of tolerance to continuous or repeated administration of opioids should be addressed. The ability of ketamine to improve opioid tolerance has been reported in clinical studies, and its mechanism of tolerance may involve improved desensitization of μ-opioid receptors (MORs). We measured changes in MOR activity and intracellular signaling induced by repeated fentanyl and morphine administration and investigated the effects of ketamine on these changes with human embryonic kidney 293 cells expressing MOR using the CellKey™, cADDis cyclic adenosine monophosphate, and PathHunter® β-arrestin recruitment assays. Repeated administration of fentanyl or morphine suppressed the second MOR responses. Administration of ketamine before a second application of opioids within clinical concentrations improved acute desensitization and enhanced β-arrestin recruitment elicited by fentanyl but not by morphine. The effects of ketamine on fentanyl were suppressed by co-treatment with an inhibitor of G-protein-coupled receptor kinase (GRK). Ketamine may potentially reduce fentanyl tolerance but not that of morphine through modulation of GRK-mediated pathways, possibly changing the conformational changes of β-arrestin to MOR.
Collapse
Affiliation(s)
- Yusuke Mizobuchi
- Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikatacho, Kita-ku, Okayama-shi 700-8558, Japan; (Y.M.); (H.M.)
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.M.); (E.U.); (M.N.)
- Department of Anesthesiology and Critical Care Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan;
| | - Kanako Miyano
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.M.); (E.U.); (M.N.)
| | - Sei Manabe
- Department of Anesthesiology and Resuscitology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-shi 700-8558, Japan; (S.M.); (Y.M.)
| | - Eiko Uezono
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.M.); (E.U.); (M.N.)
- Department of Anesthesiology and Pain Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (A.K.); (Y.K.)
| | - Akane Komatsu
- Department of Anesthesiology and Pain Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (A.K.); (Y.K.)
| | - Yui Kuroda
- Department of Anesthesiology and Pain Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan; (A.K.); (Y.K.)
| | - Miki Nonaka
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.M.); (E.U.); (M.N.)
| | - Yoshikazu Matsuoka
- Department of Anesthesiology and Resuscitology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-shi 700-8558, Japan; (S.M.); (Y.M.)
| | - Tetsufumi Sato
- Department of Anesthesiology and Critical Care Medicine, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan;
| | - Yasuhito Uezono
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan; (K.M.); (E.U.); (M.N.)
- Supportive and Palliative Care Research Support Office, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa-shi 277-8577, Japan
| | - Hiroshi Morimatsu
- Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikatacho, Kita-ku, Okayama-shi 700-8558, Japan; (Y.M.); (H.M.)
- Department of Anesthesiology and Resuscitology, Okayama University Hospital, 2-5-1 Shikatacho, Kita-ku, Okayama-shi 700-8558, Japan; (S.M.); (Y.M.)
| |
Collapse
|
44
|
Mayoral V. An overview of the use and misuse/abuse of opioid analgesics in different world regions and future perspectives. Pain Manag 2022; 12:535-555. [PMID: 35118876 DOI: 10.2217/pmt-2021-0094] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Opioids are an important therapeutic option for severe resistant chronic pain but, in the absence of proper oversight, their use has risks. The level of prescription opioid misuse/abuse differs among countries, due to differences in healthcare systems and pain management approaches. However, evaluating the true dimension of prescription opioid misuse/abuse is complicated by statistical reporting which often does not differentiate between prescription and illicit opioid use, or between prescription opioid use by patients and nonpatients, highlighting a need for greater uniformity. Parallel efforts to educate patients and the general public about opioid risks, facilitate appropriate analgesic prescribing and identify alternative formulations or options to use instead of or with opioids, may contribute to optimizing prescription opioid use for pain management.
Collapse
Affiliation(s)
- Victor Mayoral
- Pain Unit, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
45
|
Puls K, Schmidhammer H, Wolber G, Spetea M. Mechanistic Characterization of the Pharmacological Profile of HS-731, a Peripherally Acting Opioid Analgesic, at the µ-, δ-, κ-Opioid and Nociceptin Receptors. Molecules 2022; 27:919. [PMID: 35164182 PMCID: PMC8840597 DOI: 10.3390/molecules27030919] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
Accumulated preclinical and clinical data show that peripheral restricted opioids provide pain relief with reduced side effects. The peripherally acting opioid analgesic HS-731 is a potent dual μ-/δ-opioid receptor (MOR/DOR) full agonist, and a weak, partial agonist at the κ-opioid receptor (KOR). However, its binding mode at the opioid receptors remains elusive. Here, we present a comprehensive in silico evaluation of HS-731 binding at all opioid receptors. We provide insights into dynamic interaction patterns explaining the different binding and activity of HS-731 on the opioid receptors. For this purpose, we conducted docking, performed molecular dynamics (MD) simulations and generated dynamic pharmacophores (dynophores). Our results highlight two residues important for HS-731 recognition at the classical opioid receptors (MOR, DOR and KOR), particular the conserved residue 5.39 (K) and the non-conserved residue 6.58 (MOR: K, DOR: W and KOR: E). Furthermore, we assume a salt bridge between the transmembrane helices (TM) 5 and 6 via K2275.39 and E2976.58 to be responsible for the partial agonism of HS-731 at the KOR. Additionally, we experimentally demonstrated the absence of affinity of HS-731 to the nociceptin/orphanin FQ peptide (NOP) receptor. We consider the morphinan phenol Y1303.33 responsible for this affinity lack. Y1303.33 points deep into the NOP receptor binding pocket preventing HS-731 binding to the orthosteric binding pocket. These findings provide significant structural insights into HS-731 interaction pattern with the opioid receptors that are important for understanding the pharmacology of this peripheral opioid analgesic.
Collapse
Affiliation(s)
- Kristina Puls
- Department of Pharmaceutical Chemistry, Institute of Pharmcy, Freie Universität Berlin, Königin-Luise-Str. 2+4, D-14195 Berlin, Germany;
| | - Helmut Schmidhammer
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria;
| | - Gerhard Wolber
- Department of Pharmaceutical Chemistry, Institute of Pharmcy, Freie Universität Berlin, Königin-Luise-Str. 2+4, D-14195 Berlin, Germany;
| | - Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria;
| |
Collapse
|
46
|
Massaly N, Markovic T, Creed M, Al-Hasani R, Cahill CM, Moron JA. Pain, negative affective states and opioid-based analgesics: Safer pain therapies to dampen addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 157:31-68. [PMID: 33648672 DOI: 10.1016/bs.irn.2020.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Across centuries and civilizations opioids have been used to relieve pain. In our modern societies, opioid-based analgesics remain one of the most efficient treatments for acute pain. However, the long-term use of opioids can lead to the development of analgesic tolerance, opioid-induced hyperalgesia, opioid use disorders, and overdose, which can ultimately produce respiratory depressant effects with fatal consequences. In addition to the nociceptive sensory component of pain, negative affective states arising from persistent pain represent a risk factor for developing an opioid use disorder. Several studies have indicated that the increase in prescribed opioid analgesics since the 1990s represents the root of our current opioid epidemic. In this review, we will present our current knowledge on the endogenous opioid system within the pain neuroaxis and the plastic changes occurring in this system that may underlie the occurrence of pain-induced negative affect leading to misuse and abuse of opioid medications. Dissecting the allostatic neuronal changes occurring during pain is the most promising avenue to uncover novel targets for the development of safer pain medications. We will discuss this along with current and potential approaches to treat pain-induced negative affective states that lead to drug misuse. Moreover, this chapter will provide a discussion on potential avenues to reduce the abuse potential of new analgesic drugs and highlight a basis for future research and drug development based on recent advances in this field.
Collapse
Affiliation(s)
- Nicolas Massaly
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States; Washington University in St Louis, Pain Center, St. Louis, MO, United States; Washington University in St Louis, School of Medicine, St. Louis, MO, United States.
| | - Tamara Markovic
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States; Washington University in St Louis, Pain Center, St. Louis, MO, United States; Washington University in St Louis, School of Medicine, St. Louis, MO, United States
| | - Meaghan Creed
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States; Washington University in St Louis, Pain Center, St. Louis, MO, United States; Washington University in St Louis, School of Medicine, St. Louis, MO, United States; Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, United States; Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - Ream Al-Hasani
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States; Washington University in St Louis, Pain Center, St. Louis, MO, United States; Washington University in St Louis, School of Medicine, St. Louis, MO, United States; Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, United States; Center for Clinical Pharmacology, St. Louis College of Pharmacy and Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Catherine M Cahill
- Department of Psychiatry and Biobehavioural Sciences, University of California, Los Angeles, CA, United States; Shirley and Stefan Hatos Center for Neuropharmacology, University of California Los Angeles, Los Angeles, CA, United States; Jane & Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, United States
| | - Jose A Moron
- Department of Anesthesiology, Washington University in St. Louis, St. Louis, MO, United States; Washington University in St Louis, Pain Center, St. Louis, MO, United States; Washington University in St Louis, School of Medicine, St. Louis, MO, United States; Department of Neuroscience, Washington University in St. Louis, St. Louis, MO, United States; Department of Psychiatry, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
47
|
Quirk K, Stevenson M. Buprenorphine Microdosing for the Pain and Palliative Care Clinician. J Palliat Med 2022; 25:145-154. [PMID: 34978915 DOI: 10.1089/jpm.2021.0378] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Buprenorphine (BUP) can be a safe and effective alternative to traditional opioids for many patients with chronic pain. For patients on higher doses of opioids, rotation to BUP is complicated by the requirement of an opioid-free interval or withdrawal during the transition. Microdosing inductions, in which BUP is gradually titrated, while full agonist opioids are continued, are a viable alternative to traditional inductions. The objective of this article is to review the current literature on BUP microdosing induction, with a focus on patients using opioids for pain. A literature review of the PubMed database was performed in the United States on articles published from inception to May 2021. A total of 34 publications were included. The most commonly utilized microdosing strategy involved administering divided doses of sublingual (SL) products marketed for opioid use disorder treatment, with 25 (73.5%) articles reporting use of partial SL tablets or films (ranging from 1/8 to 1/2 of a 2 mg product) at some point during the induction. Transdermal patches, low-dose SL BUP available in Europe, intravenous BUP, and buccal BUP have also been used. Beyond the products used, the speed of the microinduction, setting, final BUP dosing, and management of concomitant full agonists vary widely in the literature. Microdosing regimens should be individualized based on local guidelines and patient-specific factors. Further studies comparing the safety and efficacy of different protocols are warranted.
Collapse
Affiliation(s)
- Kyle Quirk
- Department of Palliative Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Maximillian Stevenson
- Department of Palliative Medicine, Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
48
|
Zhang YZ, Wang MM, Wang SY, Wang XF, Yang WJ, Zhao YN, Han FT, Zhang Y, Gu N, Wang CL. Novel Cyclic Endomorphin Analogues with Multiple Modifications and Oligoarginine Vector Exhibit Potent Antinociception with Reduced Opioid-like Side Effects. J Med Chem 2021; 64:16801-16819. [PMID: 34781680 DOI: 10.1021/acs.jmedchem.1c01631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Endomorphins (EMs) are potent pharmaceuticals for the treatment of pain. Herein, we investigated several novel EM analogues with multiple modifications and oligoarginine conjugation. Our results showed that analogues 1-6 behaved as potent μ-opioid agonists and enhanced stability and lipophilicity. Analogues 5 and 6 administered centrally and peripherally induced significant and prolonged antinociceptive effects in acute pain. Both analogues also produced long-acting antiallodynic effects against neuropathic and inflammatory pain. Furthermore, they showed a reduced acute antinociceptive tolerance. Analogue 6 decreased the extent of chronic antinociceptive tolerance, and analogue 5 exhibited no tolerance at the supraspinal level. Particularly, they displayed nontolerance-forming antinociception at the peripheral level. In addition, analogues 5 and 6 exhibited reduced or no opioid-like side effects on gastrointestinal transit, conditioned place preference (CPP), and motor impairment. The present investigation established that multiple modifications and oligoarginine-vector conjugation of EMs would be helpful in developing novel analgesics with fewer side effects.
Collapse
Affiliation(s)
- Yu-Zhe Zhang
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin 150001, China
| | - Meng-Meng Wang
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin 150001, China
| | - Si-Yu Wang
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin 150001, China
| | - Xiao-Fang Wang
- Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Wen-Jiao Yang
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin 150001, China
| | - Ya-Nan Zhao
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin 150001, China
| | - Feng-Tong Han
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin 150001, China
| | - Yao Zhang
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin 150001, China
| | - Ning Gu
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin 150001, China
| | - Chang-Lin Wang
- School of Life Science and Technology, Harbin Institute of Technology, 92 West Dazhi Street, Harbin 150001, China.,Stake Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China
| |
Collapse
|
49
|
Agonistic activity of fentanyl analogs and their metabolites on opioid receptors. Forensic Toxicol 2021; 40:156-162. [DOI: 10.1007/s11419-021-00602-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/26/2021] [Indexed: 11/30/2022]
|
50
|
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: 1.7] [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
|