1
|
Birring SS, Dicpinigaitis PV, Maher TM, Mazzone SB, Page CP, Hawi A, Sciascia T, Morice AH. Kappa and Mu Opioid Receptors in Chronic Cough: Current Evidence and Future Treatment. Lung 2025; 203:62. [PMID: 40358749 PMCID: PMC12075272 DOI: 10.1007/s00408-025-00812-8] [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: 02/28/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025]
Abstract
Chronic cough is a significant burden on patient quality of life and is associated with poor health outcomes. Chronic cough may be a result of neural hypersensitivity due to changes in both the peripheral and the central nervous systems, although the exact mechanisms underlying its pathogenesis are not completely understood. Opioid receptors, specifically kappa and mu, are potential therapeutic targets in the management of chronic cough because they play a pivotal role in both the peripheral and the central neural pathways implicated in the act of coughing. Morphine, a mu opioid receptor agonist, is an effective cough modulator; however, mu receptor agonists are part of a drug class that can induce respiratory depression and euphoria, with strong reinforcing properties that may lead to excessive use and abuse. Drugs with a dual-acting mechanism of kappa receptor agonism and mu receptor antagonism may be effective in the management of chronic cough without the potential for abuse. This review summarizes the current understanding of the mechanisms of cough hypersensitivity, the role of the kappa and mu receptors in the neurophysiology of cough, and the clinical potential of targeting these receptors as a novel way of managing chronic cough.
Collapse
MESH Headings
- Humans
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Receptors, Opioid, mu/antagonists & inhibitors
- Cough/drug therapy
- Cough/physiopathology
- Cough/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, kappa/antagonists & inhibitors
- Chronic Disease
- Animals
- Antitussive Agents/therapeutic use
- Analgesics, Opioid/therapeutic use
- Chronic Cough
Collapse
Affiliation(s)
- Surinder S Birring
- Centre for Human & Applied Physiological Sciences, School of Basic & Medical Biosciences, King's College London, London, UK
| | | | - Toby M Maher
- University of Southern California, Los Angeles, CA, USA
| | - Stuart B Mazzone
- Fibrosis Research Group, National Heart and Lung Institute, Imperial College London, London, UK
| | - Clive P Page
- University of Melbourne, Melbourne, VIC, Australia
| | - Amale Hawi
- Trevi Therapeutics, Inc, New Haven, CT, USA
| | | | - Alyn H Morice
- Respiratory Medicine, Hull York Medical School, Castle Hill Hospital, Castle Rd, E Yorkshire, Cottingham, HU16 5JQ, UK.
| |
Collapse
|
2
|
Tang S, Hu S, Feng L, Kong L, Gui J, Zhang Y, Liu ZH, Zhang D, Liu AA, Liu X, Hu C, Lan Y, Liu X, Li Z, Liu P, Duan S, Du Z, Liu M, Xie Q, Liu J, Shao L, Fu W, Wang Y, Li W. Structure-activity relationship analysis of meta-substituted N-cyclopropylmethyl-nornepenthones with mixed KOR/MOR activities. Eur J Med Chem 2025; 289:117449. [PMID: 40068406 DOI: 10.1016/j.ejmech.2025.117449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Substance Use Disorder (SUD) remains a significant global challenge, with current treatment options offering limited efficacy. Agonists targeting the kappa opioid receptor (KOR), especially those with additional mu opioid receptor (MOR) antagonistic activity, have shown promise in addressing SUD. In this study, a series of meta-substituted N-cyclopropylmethyl-nornepenthone derivatives were designed and synthesized, and their biological activities were assessed, leading to the identification of a KOR/MOR dual modulator, compound 10a. Unlike its para-positional isomer SLL-1062, where KOR activity is completely abolished, compound 10a displayed a single-digit nanomolar affinity for KOR, while its binding profiles for MOR and delta opioid receptor (DOR) were comparable to those of SLL-1062. Functional assays in vitro confirmed that compound 10a exhibited agonistic activity at KOR and antagonistic activity at MOR. The molecular basis for the introduction of a KOR component into compound 10a was further elucidated. Although compound 10a did not produce apparent antinociception in vivo, it effectively blocked morphine-induced antinociception and intestinal motility inhibition in rodent models. This study provides valuable insights into the development of MOR/KOR dual modulators and presents new lead compounds for potential treatments for SUD.
Collapse
MESH Headings
- Structure-Activity Relationship
- Animals
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/antagonists & inhibitors
- Mice
- Receptors, Opioid, mu/metabolism
- Receptors, Opioid, mu/agonists
- Molecular Structure
- Humans
- Male
- Dose-Response Relationship, Drug
- Rats
- Gastrointestinal Motility/drug effects
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Siyuan Tang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China; School of Physical Science and Technology, ShanghaiTech University, No. 393 Huaxiazhong Road, Shanghai, 201210, China
| | - Shuyang Hu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Lijing Feng
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China
| | - Linghui Kong
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Jiangwen Gui
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China; School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ying Zhang
- School of Physical Science and Technology, ShanghaiTech University, No. 393 Huaxiazhong Road, Shanghai, 201210, China
| | - Zi-Han Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Denggao Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - An-An Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiao Liu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Chuyuan Hu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China; University of Chinese Academy of Sciences, No. 19 A Yuquan Road, 100049, Beijing, China
| | - Yingjie Lan
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Xiaoning Liu
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China
| | - Zixiang Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Panwen Liu
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China
| | - Shaoliang Duan
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Zeyi Du
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Min Liu
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Jinggen Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China.
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China.
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China
| | - Yujun Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai, 201203, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China; University of Chinese Academy of Sciences, No. 19 A Yuquan Road, 100049, Beijing, China.
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai, 201203, China.
| |
Collapse
|
3
|
Lewis T, Haouzi P. Nalfurafine reverses fentanyl-induced muscle rigidity and respiratory depression without affecting sedation in rats: decoupling respiration from sedation. J Appl Physiol (1985) 2025; 138:1161-1172. [PMID: 40215161 DOI: 10.1152/japplphysiol.00952.2024] [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: 12/05/2024] [Revised: 01/18/2025] [Accepted: 03/26/2025] [Indexed: 05/01/2025] Open
Abstract
This study examines whether nalfurafine, a κ-opioid receptor (κOR) agonist, reverses fentanyl-induced muscle rigidity and respiratory depression without impacting sedation. Adult, nonsedated Sprague-Dawley rats received intravenous fentanyl bolus injection (150 μg/kg), which induced immediate coma and muscle rigidity along with a transient central apnea, followed by sustained respiratory depression. Five minutes postfentanyl, rats were treated with saline, nalfurafine (0.1 or 1 mg/kg), or the κOR agonist U50488 (1 or 10 mg/kg). Muscle rigidity, sedation, and response to painful stimuli were assessed using blinded clinical scoring. Ventilation and pulmonary gas exchange were measured using open-flow body plethysmography. Nalfurafine at 1 mg/kg quickly and effectively reduced fentanyl-induced muscle rigidity, with a sustained effect throughout the 30-min observation period. Sedation scores remained consistent across all groups. In contrast to saline, nalfurafine (1 mg/kg) sharply increased minute ventilation V̇e, frequency (f), and tidal volume (Vt), which all plateaued at prefentanyl levels despite the animals remaining in a coma. V̇e/V̇o2 and V̇e/V̇co2 ratios also increased significantly compared with saline injection. U50488 at 1 mg/kg reversed muscle rigidity. Both nalfurafine (0.1 mg/kg) and U50488 at 1 mg/kg had a significantly weaker effect on ventilation than nalfurafine (1 mg/kg). At higher doses (10 mg/kg), U50488 after fentanyl was fatal. Nalfurafine is a promising therapeutic candidate able to reverse fentanyl-induced muscle rigidity and respiratory depression without affecting sedation or the response to painful stimuli, potentially improving the safety and efficacy of fentanyl in specific clinical settings. We could not, however, confirm that this effect was mediated via κOR.NEW & NOTEWORTHY Nalfurafine, a κ-opioid receptor (κOR) agonist, can reverse fentanyl-induced muscle rigidity and respiratory depression without affecting sedation. In nonanesthetized rats, nalfurafine (1 mg/kg) effectively reversed muscle rigidity and restored ventilation to prefentanyl levels even though the animals remained fully sedated. U50488, another κOR agonist, did not reproduce the results. These findings suggest that nalfurafine is a promising therapeutic to mitigate fentanyl's side effects while maintaining the benefits.
Collapse
Affiliation(s)
- Tristan Lewis
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, United States
| | - Philippe Haouzi
- Department of Pulmonary Medicine, Cleveland Clinic, Cleveland, Ohio, United States
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States
| |
Collapse
|
4
|
Zhang L, Zhou S, Yan L, Li X, Ju Y, Wu B, Wang H, Wang J, Sun Y. Supraspinal kappa-opioid receptors: new therapeutic strategies for pain, pruritus, and negative emotions. Exp Brain Res 2025; 243:116. [PMID: 40338314 PMCID: PMC12062100 DOI: 10.1007/s00221-025-07066-z] [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: 09/09/2024] [Accepted: 03/24/2025] [Indexed: 05/09/2025]
Abstract
Research on kappa-opioid receptor (KOR) regulation of pain and itching has focused primarily on spinal and peripheral levels. However, the role of central KOR in this process, as well as the mechanisms exacerbating negative emotional responses to pain and itching, remains unknown. Therefore, this study aimed to utilize the advantages of intracerebroventricular (i.c.v.) administration of U50488H to explore supraspinal KOR activation on pain, itching, and negative emotions. U50488H, a prototypical KOR agonist, was administered i.c.v., with physiological saline as the control. The Hargreaves test and intradermal injection of histamine and chloroquine were conducted to assess thermal pain and itch behavior, respectively. The elevated plus maze (EPM), open field test (OFT), and tail suspension test (TST) were performed to evaluate negative emotions. i.c.v. administration of U50488H increased thermal pain latencies, reduced scratching behavior, and decreased locomotor activity in the central zone of the OFT and in the open arms of the EPM, while increasing immobility in the TST. i.c.v. pretreatment with the KOR antagonist nor-Binaltorphimine dihydrochloride reversed all of the above behaviors. In conclusion, central administration of U50488H can exhibit analgesic and antipruritic effects while also inducing negative emotional responses. Our results highlight the potential of supraspinal KOR as a promising therapeutic target in the combined treatment of pain, pruritus, and negative emotions.
Collapse
MESH Headings
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
- Pruritus/drug therapy
- Pruritus/metabolism
- Animals
- Male
- Pain/drug therapy
- Pain/metabolism
- Emotions/drug effects
- Emotions/physiology
- Behavior, Animal/drug effects
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/administration & dosage
- Mice
- Rats
- Analgesics, Opioid/pharmacology
Collapse
Affiliation(s)
- Li Zhang
- School of Basic Medicine, Binzhou Medical University, Yantai, 264003, PR China
| | - Shuai Zhou
- School of Basic Medicine, Binzhou Medical University, Yantai, 264003, PR China
| | - Lujin Yan
- School of First Clinical Medicine, Binzhou Medical University, Yantai, 264003, PR China
| | - Xinyu Li
- School of First Clinical Medicine, Binzhou Medical University, Yantai, 264003, PR China
| | - Yunqi Ju
- School of Medical Imaging, Binzhou Medical University, Yantai, 264003, PR China
| | - Bo Wu
- School of Medical Imaging, Binzhou Medical University, Yantai, 264003, PR China
| | - Hongjie Wang
- School of First Clinical Medicine, Binzhou Medical University, Yantai, 264003, PR China
| | - Jian Wang
- Department of Cardiothoracic Surgery, The General Hospital of Western Theater Command, Chengdu, 610083, PR China.
| | - Yi Sun
- School of Basic Medicine, Binzhou Medical University, Yantai, 264003, PR China.
| |
Collapse
|
5
|
Lobe MMM, Verma S, Patil VM, Iyer MR. A review of kappa opioid receptor antagonists and their clinical trial landscape. Eur J Med Chem 2025; 287:117205. [PMID: 39893986 DOI: 10.1016/j.ejmech.2024.117205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 02/04/2025]
Abstract
Myriad signaling pathways are implicated in neuropsychiatric disorders, yet many mechanisms are unknown and current treatment options are limited. The intriguing dynorphin/kappa opioid receptor (KOR) system that is widely distributed throughout the brain appears to be essential in regulating many physiological and pathophysiological processes. This review explores up to date advances on the relationship between the dynorphin/KOR system with a particular focus on the KOR antagonist compounds tested as clinical candidates that could offer potential treatment options for CNS disorders.
Collapse
Affiliation(s)
- Maloba M M Lobe
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, MD, 20852, USA
| | - Saroj Verma
- Department of Pharmacy, School of Medical and Allied Sciences, K.R. Mangalam University, Gurugram, Haryana, India
| | - Vaishali M Patil
- Charak School of Pharmacy, Chaudhary Charan Singh University, Bharat, Meerut, Uttar Pradesh, 250004, India
| | - Malliga R Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, MD, 20852, USA.
| |
Collapse
|
6
|
Volf A, Brust TF, Kobylski RR, Czekner KM, Stahl EL, Cameron MD, Trojniak AE, Aubé J, Bohn LM. Triazole 187 is a biased KOR agonist that suppresses itch without sedation and induces anxiolytic-like behaviors in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638680. [PMID: 40027836 PMCID: PMC11870565 DOI: 10.1101/2025.02.17.638680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Kappa opioid receptor agonists are clinically used to treat pruritis and have therapeutic potential for the treatment of pain and neuropsychiatric disorders. We have previously shown that triazole 1.1 is a G protein signaling-biased KOR agonist, that can suppress itch without producing signs of sedation in mice. This profile was recapitulated in rats and non-human primates however, triazole 1.1 had limited potency as an antipruritic. Here we describe a more potent, G protein signaling-biased agonist, triazole 187. Triazole 187 is a potent antipruritic agent and does not decrease spontaneous locomotor activity; interestingly, it produces anxiolytic-like behaviors in mice, an effect not observed for triazole 1.1. In addition to curbing sedation, triazole 187 produces only mild diuresis, resulting in 30% of urine output induced by U50,488H at dose that is 188-fold the antipruritic potency dose. Compounds like triazole 187 may present a means to treat anxiety accompanied by persistent chronic itch while avoiding sedation and diuresis accompanied by typical KOR agonists. Abstract Figure
Collapse
|
7
|
Sudmoon R, Tanee T, Wonok W, Ameamsri U, Liehr T, Daduang S, Siripiyasing P, Chaveerach A. Discovery of rhynchophylline and mitraphylline in two Thai Mitragyna species and the investigation of their biological activity via opioid gene expression analysis. Sci Rep 2025; 15:5865. [PMID: 39966542 PMCID: PMC11836046 DOI: 10.1038/s41598-025-89715-5] [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: 11/21/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Mitragyna speciosa (Ms), M. diversifolia (Md), M. hirsuta (Mh) and M. rotundifolia (Mr) were investigated for phytochemicals by GC-MS and GC-FID, cytotoxicity and genotoxicity testing by MTT and comet assay, and biological activity examination through gene expression of human µ, δ, κ, and nociceptin opioid receptors by qRT-PCR. The opioid substances mitragynine, 7-hydroxymitragynine, and mitraphylline were found in all studied species, and, for first time, rhynchophylline was found in Mr, and mitraphylline in Md, Mh and Mr. The MTT and comet assays of the ethanol and hexane leaf extracts on PBMCs revealed no cytotoxicity and no significant genotoxicity compared to the negative control, except for the hexane leaf extract of Mh, which caused significant DNA damage. The biological activity of the ethanolic extract of the four species showed a binding affinity to the µ (MOR) receptor revealing a relative gene expression of 89.54 and 50.41 by Ms and Md at 1.92 and 1.133 mg/ml, 32.42 and 19.97 mg/ml by Md and Ms at 3.77 and 1.76 mg/ml. Mr contained the three opioids mentioned plus rhynchophylline and showed low relative µ (MOR) gene expression of 16.89 at 0.189 mg/ml, while as an additional species, Ipomoea aquatica (Ia) showed higher relative µ (MOR) gene expression of 37.75 and 59.76 at 2.878, and 5.813 mg/ml. A combination of Mr and Ia at 1.227, 2.907 and 0.0123, 0.0291 mg/ml extract showed high relative µ (MOR) gene expression at 71.01 and 21.71. These Mitragyna species and the combination (formula details are patent registered), substances and their biological activities can be used for the innovative production of new medicines and further clinical investigation.
Collapse
Affiliation(s)
| | - Tawatchai Tanee
- Faculty of Environment and Resource Studies, Mahasarakham University, Maha Sarakham, Thailand
| | - Warin Wonok
- Department of Biology, Faculty of Science, Khon Kaen University, Khon Kaen, Thailand
| | - Unchaleeporn Ameamsri
- Department of Biology, Faculty of Science, Khon Kaen University, Khon Kaen, Thailand
| | - Thomas Liehr
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Sakda Daduang
- Division of Pharmacognosy and Toxicology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Pornnarong Siripiyasing
- Faculty of Science and Technology, Rajabhat Mahasarakham University, Maha Sarakham, Thailand
| | - Arunrat Chaveerach
- Department of Biology, Faculty of Science, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
8
|
Schreiber S, Keidan L, Pick CG. A New Trick of Old Dogs: Can Kappa Opioid Receptor Antagonist Properties of Antidepressants Assist in Treating Treatment-Resistant Depression (TRD)? Pharmaceuticals (Basel) 2025; 18:208. [PMID: 40006022 PMCID: PMC11858657 DOI: 10.3390/ph18020208] [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: 12/01/2024] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Approximately one in five individuals will experience major depressive disorder (MDD), and 30% exhibit resistance to standard antidepressant treatments, resulting in a diagnosis of treatment-resistant depression (TRD). Historically, opium was used effectively to treat depression; however, when other medications were introduced, its use was discontinued due to addiction and other hazards. Recently, kappa opioid receptor (KOR) antagonism has been proposed as a potential mechanism for treating TRD. The main research question is whether commonly used psychotropic medications possess KOR antagonist properties and whether this characteristic could contribute to their efficacy in TRD. Methods: We investigated the antinociceptive effects of many psychotropic medications and their interactions with the opioid system. Mice were tested with a hotplate or tail-flick after being injected with different doses of these agents. Results: The antidepressants mianserin and mirtazapine (separately) induced dose-dependent antinociception, each yielding a biphasic dose-response curve. Similarly, the antidepressant venlafaxine produced a potent effect and reboxetine produced a weak effect. The antipsychotics risperidone and amisulpride exhibited a dose-dependent antinociceptive effect. The sedative-hypnotic zolpidem induced a weak bi-phasic dose-dependent antinociceptive effect. All seven psychotropic medications elicited antinociception, which was reversed by the non-selective opiate antagonist naloxone and, separately, by the kappa-selective antagonist Nor-BNI. Conclusions: Clinical studies are mandatory to establish the potential efficacy of augmentation of the treatment with antidepressants with these drugs in persons with treatment-resistant depression and the optimal dosage of medications prescribed. We suggest a possible beneficial effect of antidepressants with kappa antagonistic properties.
Collapse
Affiliation(s)
- Shaul Schreiber
- Department of Psychiatry, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel;
- Dr. Miriam and Sheldon G. Adelson Clinic for Drug Abuse Treatment and Research, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
- Faculty of Medicine & Health Sciences, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6905904, Israel
| | - Lee Keidan
- Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv 6905904, Israel;
- Department of Anatomy and Anthropology, Faculty of Medicine & Health Sciences, Tel Aviv University, Tel Aviv 6905904, Israel
| | - Chaim G. Pick
- Faculty of Medicine & Health Sciences, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6905904, Israel
- Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv 6905904, Israel;
- Department of Anatomy and Anthropology, Faculty of Medicine & Health Sciences, Tel Aviv University, Tel Aviv 6905904, Israel
- Dr. Miriam and Sheldon G. Adelson Center for the Biology of Addictive Diseases, Tel-Aviv University, Tel-Aviv 6905904, Israel
| |
Collapse
|
9
|
van de Wetering R, Vu LY, Kornberger LD, Luo D, Scouller B, Hong S, Paton K, Prisinzano TE, Kivell BM. Effects of Biased Analogues of the Kappa Opioid Receptor Agonist, U50,488, in Preclinical Models of Pain and Side Effects. Molecules 2025; 30:604. [PMID: 39942708 PMCID: PMC11820436 DOI: 10.3390/molecules30030604] [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: 12/17/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Kappa opioid receptor (KOR) agonists have well-established antinociceptive effects. However, many KOR agonists have negative side effects, which limit their therapeutic potential. Some researchers have suggested that the development of biased agonists that preferentially stimulate KOR G-protein pathways over β-arrestin pathways may yield drugs with fewer adverse side effects. This was investigated in the current study. We describe the synthesis and characterization of three U50,488 analogues, 1, 2, and 3. We evaluated the acute and chronic antinociceptive effects of these compounds in mice using the warm-water tail flick assay and in a paclitaxel-induced neuropathic pain model. Side effects were investigated using open-field, passive wire hang, rotarod, elevated zero maze, conditioned place aversion, and whole-body plethysmography, with some tests being conducted in KOR or β-arrestin2 knock out mice. All compounds were highly potent, full agonists of the KOR, with varying signaling biases in vitro. In the warm-water tail withdrawal assay, these agonists were ~10 times more potent than U50,488, but not more efficacious. All KOR agonists reversed paclitaxel-induced neuropathic pain, without tolerance. Compound 3 showed no significant side effects on any test. Signaling bias did not correlate with the antinociceptive or side effects of any compounds and knockout of β-arrestin2 had no effect on U50,488-induced sedation or motor incoordination. These findings highlight the therapeutic potential of 3, with its lack of side effects typically associated with KOR agonists, and also suggest that G-protein signaling bias is a poor predictor of KOR agonist-induced side effects.
Collapse
MESH Headings
- Animals
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
- Mice
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/adverse effects
- 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/chemistry
- Disease Models, Animal
- Neuralgia/drug therapy
- Neuralgia/chemically induced
- Male
- Mice, Knockout
- Paclitaxel/adverse effects
- Humans
- Analgesics/pharmacology
Collapse
Affiliation(s)
- Ross van de Wetering
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Loan Y. Vu
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Lindsay D. Kornberger
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Dan Luo
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Brittany Scouller
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Sheein Hong
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Kelly Paton
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| | - Thomas E. Prisinzano
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA; (L.Y.V.); (L.D.K.); (D.L.)
| | - Bronwyn M. Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand (B.S.)
| |
Collapse
|
10
|
Momotani K, Nojiri R, Uchiyama T, Taniguchi T. [Pharmacological, pharmacokinetic and clinical profiles of Difelikefalin (KORSUVA ® IV Injection Syringe for Dialysis), a peripheral kappa opioid receptor agonist]. Nihon Yakurigaku Zasshi 2025; 160:127-140. [PMID: 40024699 DOI: 10.1254/fpj.24050] [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: 03/04/2025]
Abstract
Difelikefalin (KORSUVA® IV Injection Syringe for Dialysis) is a novel kappa opioid receptor (KOR) agonist. In September 2023, difelikefalin was approved for the treatment of pruritus in hemodialysis patients. Pruritus is a major symptom that significantly reduces the quality of life of hemodialysis patients, even with improved dialysis techniques, dialysis membranes, and dialysate solutions. The factors that contribute to pruritus include dry skin, accumulation of uremic toxins, overproduction of chemical mediators and altered immune function, and disruption of the opioid balance. In nonclinical studies, difelikefalin showed highly selective for KOR and antipruritic effects in animal models of histamine- and substance P-induced itching. It also showed anti-inflammatory effects by suppressing cytokine release in human monocyte-derived macrophages and TNFα and IL-1β induced by lipopolysaccharide administration in mice. In the phase 3 clinical trial in Japanese hemodialysis patients, difelikefalin showed significant improvement compared to placebo in the primary endpoint of the change from baseline in the weekly mean NRS score at week 4. It also improved sleep disturbance and itch-related quality of life, and the improvement in itch was sustained up to 58 weeks. Furthermore, there was no increase in adverse drug reactions with long-term treatment, and no delayed adverse events were observed. In conclusion, the novel KOR agonist difelikefalin is expected to be a new treatment option for pruritus on dialysis.
Collapse
Affiliation(s)
| | - Rumi Nojiri
- Clinical Development Department, Maruishi Pharmaceutical Co., Ltd
| | - Takuma Uchiyama
- Clinical Projects Management, Kissei Pharmaceutical Co., Ltd
| | | |
Collapse
|
11
|
Xu K, Zhang M, Chen D, Xu B, Hu X, Zhang Q, Zhang R, Zhang N, Li N, Fang Q. Conorphin-66 produces peripherally restricted antinociception via the kappa-opioid receptor with limited side effects. Neuropharmacology 2024; 261:110157. [PMID: 39276862 DOI: 10.1016/j.neuropharm.2024.110157] [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: 04/12/2024] [Revised: 08/15/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
With the current unmet demand for effective pain relief, analgesics without major central adverse effects are highly appealing, such as peripherally restricted kappa-opioid receptor (KOR) agonists. In this study, Conorphin-66, an analog of the selective KOR peptide agonist Conorphin T, was pharmacologically characterized in a series of experiments, with CR845 serving as the reference compound. Firstly, in vitro functional assay indicated that Conorphin-66 selectively activates KOR and exhibits weak β-arrestin2 signaling bias (-1.54 versus -4.35 for CR845). Additionally, subcutaneous Conorphin-66 produced potent antinociception in mouse pain models with ED50 values ranged from 0.02 to 3.28 μmol/kg, including tail-flick test, post-operative pain, formalin pain, and acetic acid-induced visceral pain. Similarly, CR845 exert potent antinociception in mouse pain models ranged from 0.15 to 1.47 μmol/kg. Notably, antagonism studies revealed that the analgesic effects of Conorphin-66 were mainly mediated by the peripheral KOR. Furthermore, Conorphin-66 produced non-tolerance-forming antinociception over 8 days. Unlike CR845, subcutaneous Conorphin-66 did not promote the sedation, anxiogenic effects, depressive-like effects, but did exhibit diuretic activity. Further study showed that Conorphin-66 does not have apparent antipruritic effects in an acute itch model. Overall, Conorphin-66 emerges as a novel peripherally restricted KOR agonist that produced potent antinociception with reduced side effects.
Collapse
Affiliation(s)
- Kangtai Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, State Key Laboratory of Veterinary Etiological Biology College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Mengna Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, State Key Laboratory of Veterinary Etiological Biology College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Dan Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, State Key Laboratory of Veterinary Etiological Biology College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Biao Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, State Key Laboratory of Veterinary Etiological Biology College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Xuanran Hu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, State Key Laboratory of Veterinary Etiological Biology College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Qinqin Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, State Key Laboratory of Veterinary Etiological Biology College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Run Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, State Key Laboratory of Veterinary Etiological Biology College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Nan Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, State Key Laboratory of Veterinary Etiological Biology College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Ning Li
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, State Key Laboratory of Veterinary Etiological Biology College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China
| | - Quan Fang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, and Institute of Physiology, School of Basic Medical Sciences, State Key Laboratory of Veterinary Etiological Biology College of Veterinary Medicine, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, PR China.
| |
Collapse
|
12
|
Zhang M, Morice AH. Current and emerging opioids for the treatment of chronic cough: a mini review. Expert Opin Pharmacother 2024; 25:2167-2175. [PMID: 39434699 DOI: 10.1080/14656566.2024.2418983] [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: 09/17/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024]
Abstract
INTRODUCTION Chronic cough has increasingly been recognized as a distinct clinical entity that affects a significant portion of the global population. Despite advancements in understanding its pathophysiology, treatment options remain limited. Opioid analgesics have long been used for cough, and some have proven clear antitussive potential. However, these have yet to be approved by regulatory authorities for the treatment of chronic cough. Several novel synthetic opioid modulators that demonstrated antitussive effects in early-stage studies also failed to translate into clinical practice. AREAS COVERED This mini review aims to summarize the implications of opioid receptors in the development of cough medicines and highlight recent advances in opioid analgesics in cough trials. PUB MED/CINAHL/Web of Science/Scopus was searched (September 2024). EXPERT OPINION Our understanding of the precise sites of action and the involvement of peripheral opioid receptors in cough remains limited. Despite these gaps in knowledge, opioids remain a viable option for some patients until more novel effective treatments are available. Due to the frequent opioid side effects, new opioid derivatives with improved properties are needed. The development of tailored or biased delta-opioid receptor ligands and mixed agonists of opioid receptor-like 1/mu receptors may offer hope for new opioid-based drug discovery for chronic cough.
Collapse
Affiliation(s)
- Mengru Zhang
- Centre for Clinical Science, Respiratory Medicine, Hull York Medical School, University of Hull, Castle Hill Hospital, Cottingham, UK
| | - Alyn H Morice
- Centre for Clinical Science, Respiratory Medicine, Hull York Medical School, University of Hull, Castle Hill Hospital, Cottingham, UK
| |
Collapse
|
13
|
Rullo L, Morosini C, Lacorte A, Cristani M, Coluzzi F, Candeletti S, Romualdi P. Opioid system and related ligands: from the past to future perspectives. JOURNAL OF ANESTHESIA, ANALGESIA AND CRITICAL CARE 2024; 4:70. [PMID: 39390585 PMCID: PMC11468104 DOI: 10.1186/s44158-024-00201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024]
Abstract
Chronic pain is a pathological condition affecting about 30% of population. It represents a relevant social-health issue worldwide, and it is considered a significant source of human suffering and disability, strongly affecting patients' quality of life. Despite several pharmacological strategies to guarantee an adequate pain management have been proposed over the years, opioids still represent one of the primary choices for treating moderate-to-severe pain in both cancer and non-cancer patients. However, chronic use of opioids often leads to numerous side effects, including respiratory depression, constipation, analgesic tolerance, and opioid-induced hyperalgesia (OIH), which can strongly limit their use. Given the fundamental role of opioid system in pain relief, this review provides a general overview about the main actors (endogenous opioid peptides and receptors) involved in its modulation. Furthermore, this review explores the action and the limitations of conventional clinically used opioids and describes the efficacy and safety profile of some promising analgesic compounds. A deeper understanding of the molecular mechanisms behind both analgesic effects and adverse events could advance knowledge in this field, thus improving chronic pain treatment.
Collapse
Affiliation(s)
- Laura Rullo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy
| | - Camilla Morosini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy
| | - Antonio Lacorte
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy
| | - Marco Cristani
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy
| | - Flaminia Coluzzi
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
- Unit of Anaesthesia, Intensive Care and Pain Medicine, Sant'Andrea University Hospital, Rome, Italy
| | - Sanzio Candeletti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy
| | - Patrizia Romualdi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Irnerio 48, Bologna, 40126, Italy.
| |
Collapse
|
14
|
Trojniak AE, Dang VQ, Czekner KM, Russo RJ, Mather LM, Stahl EL, Cameron MD, Bohn LM, Aubé J. Synthesis and evaluation of 3,4,5-trisubstituted triazoles as G protein-biased kappa opioid receptor agonists. Eur J Med Chem 2024; 276:116627. [PMID: 38971050 PMCID: PMC11316643 DOI: 10.1016/j.ejmech.2024.116627] [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: 05/08/2024] [Revised: 06/14/2024] [Accepted: 06/24/2024] [Indexed: 07/08/2024]
Abstract
Kappa opioid receptor (KOR) agonists represent promising therapeutics for pain relief due to their analgesic properties along with lower abuse potential than opioids that act at the mu opioid receptor. However, typical KOR agonists produce sedation and dysphoria. Previous studies have shown that G protein signaling-biased KOR agonists may present a means to untangle the desired analgesic properties from undesired side effects. In this paper, we report a new series of G protein signaling-biased KOR agonists entailing -S- → -CH2- replacement in a previously reported KOR agonist, triazole 1.1. With an optimized carbon linker in hand, further development of the scaffold was undertaken to investigate the appendages of the triazole core. The structure-activity relationship study of this series is described, including several analogues that display enhanced potency while maintaining G protein-signaling bias compared to triazole 1.1.
Collapse
Affiliation(s)
- Ashley E Trojniak
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7363, USA
| | - Vuong Q Dang
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Kerri M Czekner
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA; The Skaggs Graduate School of Chemical and Biological Sciences at Scripps Research, Jupiter, FL, 33458, USA
| | - Robin J Russo
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA; The Skaggs Graduate School of Chemical and Biological Sciences at Scripps Research, Jupiter, FL, 33458, USA
| | - Lilyan M Mather
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7363, USA
| | - Edward L Stahl
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Michael D Cameron
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA
| | - Laura M Bohn
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 130 Scripps Way, Jupiter, FL, 33458, USA; The Skaggs Graduate School of Chemical and Biological Sciences at Scripps Research, Jupiter, FL, 33458, USA
| | - Jeffrey Aubé
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7363, USA.
| |
Collapse
|
15
|
Radomsky T, Anderson RC, Millar RP, Newton CL. Restoring function to inactivating G protein-coupled receptor variants in the hypothalamic-pituitary-gonadal axis 1. J Neuroendocrinol 2024; 36:e13418. [PMID: 38852954 DOI: 10.1111/jne.13418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 03/30/2024] [Accepted: 05/15/2024] [Indexed: 06/11/2024]
Abstract
G protein-coupled receptors (GPCRs) are central to the functioning of the hypothalamic-pituitary-gonadal axis (HPG axis) and include the rhodopsin-like GPCR family members, neurokinin 3 receptor, kappa-opioid receptor, kisspeptin 1 receptor, gonadotropin-releasing hormone receptor, and the gonadotropin receptors, luteinizing hormone/choriogonadotropin receptor and follicle-stimulating hormone receptor. Unsurprisingly, inactivating variants of these receptors have been implicated in a spectrum of reproductive phenotypes, including failure to undergo puberty, and infertility. Clinical induction of puberty in patients harbouring such variants is possible, but restoration of fertility is not always a realisable outcome, particularly for those patients suffering from primary hypogonadism. Thus, novel pharmaceuticals and/or a fundamental change in approach to treating these patients are required. The increasing wealth of data describing the effects of coding-region genetic variants on GPCR function has highlighted that the majority appear to be dysfunctional as a result of misfolding of the encoded receptor protein, which, in turn, results in impaired receptor trafficking through the secretory pathway to the cell surface. As such, these intracellularly retained receptors may be amenable to 'rescue' using a pharmacological chaperone (PC)-based approach. PCs are small, cell permeant molecules hypothesised to interact with misfolded intracellularly retained proteins, stabilising their folding and promoting their trafficking through the secretory pathway. In support of the use of this approach as a viable therapeutic option, it has been observed that many rescued variant GPCRs retain at least a degree of functionality when 'rescued' to the cell surface. In this review, we examine the GPCR PC research landscape, focussing on the rescue of inactivating variant GPCRs with important roles in the HPG axis, and describe what is known regarding the mechanisms by which PCs restore trafficking and function. We also discuss some of the merits and obstacles associated with taking this approach forward into a clinical setting.
Collapse
Affiliation(s)
- Tarryn Radomsky
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Ross C Anderson
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Robert P Millar
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- School of Medicine, University of St Andrews, St Andrews, UK
| | - Claire L Newton
- Centre for Neuroendocrinology, Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
16
|
Puls K, Olivé-Marti AL, Hongnak S, Lamp D, Spetea M, Wolber G. Discovery of Novel, Selective, and Nonbasic Agonists for the Kappa-Opioid Receptor Determined by Salvinorin A-Based Virtual Screening. J Med Chem 2024; 67:13788-13801. [PMID: 39088801 PMCID: PMC11345774 DOI: 10.1021/acs.jmedchem.4c00590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 08/03/2024]
Abstract
Modulating the kappa-opioid receptor (KOR) is a promising strategy for treating various human diseases. KOR agonists show potential for treating pain, pruritus, and epilepsy, while KOR antagonists show potential for treating depression, anxiety, and addiction. The diterpenoid Salvinorin A (SalA), a secondary metabolite of Salvia divinorum, is a potent and selective KOR agonist. Unlike typical opioids, SalA lacks a basic nitrogen, which encouraged us to search for nonbasic KOR ligands. Through structure-based virtual screening using 3D pharmacophore models based on the binding mode of SalA, we identified novel, nonbasic, potent, and selective KOR agonists. In vitro studies confirmed two virtual hits, SalA-VS-07 and SalA-VS-08, as highly selective for the KOR and showing G protein-biased KOR agonist activity. Both KOR ligands share a novel spiro-moiety and a nonbasic scaffold. Our findings provide novel starting points for developing therapeutics aimed at treating pain and other conditions in which KOR is a central player.
Collapse
Affiliation(s)
- Kristina Puls
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| | - Aina-Leonor Olivé-Marti
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Siriwat Hongnak
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - David Lamp
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Mariana Spetea
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy and Center for
Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Gerhard Wolber
- Department
of Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| |
Collapse
|
17
|
Govender D, Moloko L, Papathanasopoulos M, Tumba N, Owen G, Calvey T. Ibogaine administration following repeated morphine administration upregulates myelination markers 2', 3'-cyclic nucleotide 3'-phosphodiesterase (CNP) and myelin basic protein (MBP) mRNA and protein expression in the internal capsule of Sprague Dawley rats. Front Neurosci 2024; 18:1378841. [PMID: 39114487 PMCID: PMC11303312 DOI: 10.3389/fnins.2024.1378841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
Ibogaine is a psychedelic alkaloid being investigated as a possible treatment for opioid use disorder. Ibogaine has a multi-receptor profile with affinities for mu and kappa opioid as well as NMDA receptors amongst others. Due to the sparsity of research into ibogaine's effects on white matter integrity and given the growing evidence that opioid use disorder is characterized by white matter pathology, we set out to investigate ibogaine's effects on two markers of myelination, 2', 3'-cyclic nucleotide 3'-phosphodiesterase (CNP) and myelin basic protein (MBP). Fifty Sprague Dawley rats were randomly assigned to five experimental groups of n = 10; (1) a saline control group received daily saline injections for 10 days, (2) a morphine control group received escalating morphine doses from 5 to 15 mg/kg over 10 days, (3) an ibogaine control group that received 10 days of saline followed by 50 mg/kg ibogaine hydrochloride, (4) a combination morphine and ibogaine group 1 that received the escalating morphine regime followed by 50 mg/kg ibogaine hydrochloride and (5) a second combination morphine and ibogaine group 2 which followed the same morphine and ibogaine regimen yet was terminated 72 h after administration compared to 24 h in the other groups. White matter from the internal capsule was dissected and qPCR and western blotting determined protein and gene expression of CNP and MBP. Morphine upregulated CNPase whereas ibogaine alone had no effect on CNP mRNA or protein expression. However, ibogaine administration following repeated morphine administration had an immediate effect by increasing CNP mRNA expression. This effect diminished after 72 h and resulted in a highly significant upregulation of CNPase protein at 72 h post administration. Ibogaine administration alone significantly upregulated protein expression yet downregulated MBP mRNA expression. Ibogaine administration following repeated morphine administration significantly upregulated MBP mRNA expression which increased at 72 h post administration resulting in a highly significant upregulation of MBP protein expression at 72 h post administration. These findings indicate that ibogaine is able to upregulate genes and proteins involved in the process of remyelination following opioid use and highlights an important mechanism of action of ibogaine's ability to treat substance use disorders.
Collapse
Affiliation(s)
- Demi Govender
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Leila Moloko
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Maria Papathanasopoulos
- HIV Pathogenesis Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nancy Tumba
- HIV Pathogenesis Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Gavin Owen
- HIV Pathogenesis Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Tanya Calvey
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
18
|
Saboori Amleshi R, Soltaninejad M, Ilaghi M. Potential Involvement of Apelin/APJ System in Addiction and Neuroprotection Against Drugs of Abuse. ADDICTION & HEALTH 2024; 16:198-204. [PMID: 39439853 PMCID: PMC11491857 DOI: 10.34172/ahj.1479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 04/15/2024] [Indexed: 10/25/2024]
Abstract
Addiction, characterized by compulsive drug-seeking behavior and impaired self-control, remains a significant public health concern. Understanding the neurobiology of addiction is crucial for identifying novel therapeutic targets and further developing effective treatments. Recently, the apelin/APJ system, an emerging signaling pathway, has attracted attention for its involvement in various neuropsychiatric disorders. The cross-talk between the apelin/APJ system and hypothalamic mu opioid signaling, as well as its heterodimerization with kappa opioid receptors (KORs), supports the potential relevance of this system to addiction. Moreover, several protective effects of apelin against various addictive substances, including methamphetamine, morphine, and alcohol, underscore the need for further investigation into its role in substance use disorder. Understanding the contribution of the apelin/APJ system in addiction may offer valuable insights into the underlying neurobiology and pave the way for novel therapeutic interventions in substance use disorders. This review provides a concise overview of the apelin/APJ system, emphasizing its physiological roles and highlighting its relevance to addiction research.
Collapse
Affiliation(s)
| | | | - Mehran Ilaghi
- Institute of Neuropharmacology, Kerman Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
19
|
Cayir S, Zhornitsky S, Barzegary A, Sotomayor-Carreño E, Sarfo-Ansah W, Funaro MC, Matuskey D, Angarita G. A review of the kappa opioid receptor system in opioid use. Neurosci Biobehav Rev 2024; 162:105713. [PMID: 38733895 DOI: 10.1016/j.neubiorev.2024.105713] [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: 03/04/2024] [Revised: 04/23/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
The kappa opioid receptor (KOR) system is implicated in dysphoria and as an "anti-reward system" during withdrawal from opioids. However, no clear consensus has been made in the field, as mixed findings have been reported regarding the relationship between the KOR system and opioid use. This review summarizes the studies to date on the KOR system and opioids. A systematic scoping review was reported following PRISMA guidelines and conducted based on the published protocol. Comprehensive searches of several databases were done in the following databases: MEDLINE, Embase, PsycINFO, Web of Science, Scopus, and Cochrane. We included preclinical and clinical studies that tested the administration of KOR agonists/antagonists or dynorphin and/or measured dynorphin levels or KOR expression during opioid intoxication or withdrawal from opioids. One hundred studies were included in the final analysis. Preclinical administration of KOR agonists decreased drug-seeking/taking behaviors and opioid withdrawal symptoms. KOR antagonists showed mixed findings, depending on the agent and/or type of withdrawal symptom. Administration of dynorphins attenuated opioid withdrawal symptoms both in preclinical and clinical studies. In the limited number of available studies, dynorphin levels were found to increase in cerebrospinal fluid (CSF) and peripheral blood lymphocytes (PBL) of opioid use disorder subjects (OUD). In animals, dynorphin levels and/or KOR expression showed mixed findings during opioid use. The KOR/dynorphin system appears to have a multifaceted and complex nature rather than simply functioning as an anti-reward system. Future research in well-controlled study settings is necessary to better understand the clinical role of the KOR system in opioid use.
Collapse
Affiliation(s)
- Salih Cayir
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06510, USA
| | - Simon Zhornitsky
- Department of Psychology, Southern Connecticut State University, New Haven, CT 06515, USA
| | - Alireza Barzegary
- Islamic Azad University Tehran Medical Sciences School of Medicine, Iran
| | | | | | - Melissa C Funaro
- Harvey Cushing/John Hay Whitney Medical Library, Yale University, New Haven, CT 06510, USA
| | - David Matuskey
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06510, USA; Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA; Department of Neurology, Yale University, New Haven, CT 06510, USA
| | - Gustavo Angarita
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA; Clinical Neuroscience Research Unit, Connecticut Mental Health Center, 34 Park Street, New Haven, CT 06519, USA.
| |
Collapse
|
20
|
Li Z, Huang R, Xia M, Chang N, Guo W, Liu J, Dong F, Liu B, Varghese A, Aslam A, Patterson TA, Hong H. Decoding the κ Opioid Receptor (KOR): Advancements in Structural Understanding and Implications for Opioid Analgesic Development. Molecules 2024; 29:2635. [PMID: 38893511 PMCID: PMC11173883 DOI: 10.3390/molecules29112635] [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: 04/27/2024] [Revised: 05/25/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
The opioid crisis in the United States is a significant public health issue, with a nearly threefold increase in opioid-related fatalities between 1999 and 2014. In response to this crisis, society has made numerous efforts to mitigate its impact. Recent advancements in understanding the structural intricacies of the κ opioid receptor (KOR) have improved our knowledge of how opioids interact with their receptors, triggering downstream signaling pathways that lead to pain relief. This review concentrates on the KOR, offering crucial structural insights into the binding mechanisms of both agonists and antagonists to the receptor. Through comparative analysis of the atomic details of the binding site, distinct interactions specific to agonists and antagonists have been identified. These insights not only enhance our understanding of ligand binding mechanisms but also shed light on potential pathways for developing new opioid analgesics with an improved risk-benefit profile.
Collapse
Affiliation(s)
- Zoe Li
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (Z.L.); (W.G.); (J.L.); (F.D.); (B.L.); (A.V.); (A.A.)
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA; (R.H.); (M.X.)
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA; (R.H.); (M.X.)
| | - Nancy Chang
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA;
| | - Wenjing Guo
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (Z.L.); (W.G.); (J.L.); (F.D.); (B.L.); (A.V.); (A.A.)
| | - Jie Liu
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (Z.L.); (W.G.); (J.L.); (F.D.); (B.L.); (A.V.); (A.A.)
| | - Fan Dong
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (Z.L.); (W.G.); (J.L.); (F.D.); (B.L.); (A.V.); (A.A.)
| | - Bailang Liu
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (Z.L.); (W.G.); (J.L.); (F.D.); (B.L.); (A.V.); (A.A.)
| | - Ann Varghese
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (Z.L.); (W.G.); (J.L.); (F.D.); (B.L.); (A.V.); (A.A.)
| | - Aasma Aslam
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (Z.L.); (W.G.); (J.L.); (F.D.); (B.L.); (A.V.); (A.A.)
| | - Tucker A. Patterson
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA; (Z.L.); (W.G.); (J.L.); (F.D.); (B.L.); (A.V.); (A.A.)
| | - Huixiao Hong
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892, USA; (R.H.); (M.X.)
| |
Collapse
|
21
|
Li Z, Ye R, He Q, Lu J, Sun Y, Sun X, Tang S, Hu S, Chai J, Kong L, Liu X, Chen J, Fang Y, Lan Y, Xie Q, Liu J, Shao L, Fu W, Wang Y, Li W. Discovery of an Ortho-Substituted N-Cyclopropylmethyl-7α-phenyl-6,14- endoethano-tetrahydronorthebaine Derivative as a Selective and Potent Kappa Opioid Receptor Agonist with Subsided Sedative Effect. J Med Chem 2024; 67:7112-7129. [PMID: 38647397 DOI: 10.1021/acs.jmedchem.3c02439] [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: 04/25/2024]
Abstract
Research into kappa opioid receptor (KOR) agonists with attenuated central-nervous-system side effects is a critical focus for developing productive and safe analgesics. Herein, a series of ortho-substituted N-cyclopropylmethyl-7α-phenyl-6,14-endoethano-tetrahydronorthebaines were designed, synthesized, and subjected to bioassays. Compound 7a exhibited high subtype selectivity and potent agonistic activity toward KOR (KOR, Ki = 3.9 nM, MOR/KOR = 270, DOR/KOR = 1075; [35S]GTPγS binding, EC50 = 3.4 nM). Additionally, this compound exhibited robust and persistent antinociceptive effects in rodent models with different animal strains (hot plate test, ED50 = 0.20-0.30 mg/kg, i.p.; abdominal constriction test, ED50 = 0.20-0.60 mg/kg, i.p.), with its KOR-mediated mechanism for antinociception firmly established. Notably, compound 7a, unlike conventional KOR agonists, displayed minimal sedation and aversion at the antinociceptive ED50 dose. This feature addresses a crucial limitation in existing KOR agonists, positioning compound 7a as a promising novel therapeutic agent.
Collapse
Affiliation(s)
- Zixiang Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Rufeng Ye
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Qian He
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Jiashuo Lu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
- Department of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Yanting Sun
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China
| | - Xiujian Sun
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China
| | - Siyuan Tang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Shuyang Hu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Jingrui Chai
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Linghui Kong
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Xiaoning Liu
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, Shandong, China
| | - Jing Chen
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yun Fang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Yingjie Lan
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Jinggen Liu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurobiology of Zhejiang Province, Hangzhou 310053, China
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Yujun Wang
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, No. 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, Shandong, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
22
|
Uniyal A, Tiwari V, Tsukamoto T, Dong X, Guan Y, Raja SN. Targeting sensory neuron GPCRs for peripheral neuropathic pain. Trends Pharmacol Sci 2023; 44:1009-1027. [PMID: 37977131 PMCID: PMC10657387 DOI: 10.1016/j.tips.2023.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 11/19/2023]
Abstract
Despite the high prevalence of peripheral neuropathic pain (NP) conditions and significant progress in understanding its underlying mechanisms, the management of peripheral NP remains inadequate. Existing pharmacotherapies for NP act primarily on the central nervous system (CNS) and are often associated with CNS-related adverse effects, limiting their clinical effectiveness. Mounting preclinical evidence indicates that reducing the heightened activity in primary sensory neurons by targeting G-protein-coupled receptors (GPCRs), without activating these receptors in the CNS, relieves pain without central adverse effects. In this review, we focus on recent advancements in GPCR-mediated peripheral pain relief and discuss strategies to advance the development of more effective and safer therapies for peripheral NP by shifting from traditional CNS modulatory approaches toward selective targeting of GPCRs on primary sensory neurons.
Collapse
Affiliation(s)
- Ankit Uniyal
- Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA
| | - Vinod Tiwari
- Neuroscience and Pain Research Laboratory, Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (B.H.U), Varanasi, India
| | - Takashi Tsukamoto
- Department of Neurology and Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xinzhong Dong
- Department of Neuroscience, The Johns Hopkins University, Baltimore, MD, USA
| | - Yun Guan
- Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA; Department of Neurological Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Srinivasa N Raja
- Division of Pain Medicine, Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, USA; Department of Neurology and Johns Hopkins Drug Discovery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
23
|
Ide S, Hirai T, Muto T, Yamakawa T, Ikeda K. Effects of the novel selective κ-opioid receptor agonist NP-5497-KA on morphine-induced reward-related behaviors. Sci Rep 2023; 13:18164. [PMID: 37875567 PMCID: PMC10598265 DOI: 10.1038/s41598-023-45584-4] [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: 06/26/2023] [Accepted: 10/21/2023] [Indexed: 10/26/2023] Open
Abstract
Opioid addiction and the opioid overdose epidemic are becoming more serious, and the development of therapeutic agents is essential for the pharmacological treatment of substance use disorders. The κ-opioid receptor (KOP) is a member of the opioid receptor system that has been gaining attention as a promising molecular target for the treatment of numerous human disorders, including pain, depression, anxiety, and drug addiction. Here, we biologically and pharmacologically evaluated a novel azepane-derived ligand, NP-5497-KA, as a selective KOP agonist. NP-5497-KA had 1000-fold higher selectivity for the KOP over the μ-opioid receptor (MOP), which was higher than nalfurafine (KOP/MOP: 65-fold), and acted as a selective KOP full agonist in the 3',5'-cyclic adenosine monophosphate assay. The oral administration of NP-5497-KA (1-10 mg/kg) dose-dependently suppressed morphine-induced conditioned place preference in C57BL/6 J mice, and its effects were comparable to an intraperitoneal injection of nalfurafine (1-10 μg/kg). Nalfurafine (10 μg/kg) significantly inhibited rotarod performance, whereas NP-5497-KA (10 mg/kg) exerted no effect on rotarod performance. These results indicate that NP-5497-KA may be a novel option for the treatment of opioid use disorder with fewer side effects.
Collapse
Affiliation(s)
- Soichiro Ide
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan.
| | - Toshitake Hirai
- Discovery Research Laboratories, Nippon Chemiphar Co., Ltd., 1-22 Hikokawado, Misato, Saitama, 341-0005, Japan
| | - Takafumi Muto
- Discovery Research Laboratories, Nippon Chemiphar Co., Ltd., 1-22 Hikokawado, Misato, Saitama, 341-0005, Japan
| | - Tomio Yamakawa
- Discovery Research Laboratories, Nippon Chemiphar Co., Ltd., 1-22 Hikokawado, Misato, Saitama, 341-0005, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo, 156-8506, Japan
| |
Collapse
|
24
|
van de Wetering R, Ewald A, Welsh S, Kornberger L, Williamson SE, McElroy BD, Butelman ER, Prisinzano TE, Kivell BM. The Kappa Opioid Receptor Agonist 16-Bromo Salvinorin A Has Anti-Cocaine Effects without Significant Effects on Locomotion, Food Reward, Learning and Memory, or Anxiety and Depressive-like Behaviors. Molecules 2023; 28:4848. [PMID: 37375403 PMCID: PMC10304272 DOI: 10.3390/molecules28124848] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Kappa opioid receptor (KOR) agonists have preclinical antipsychostimulant effects; however, adverse side effects have limited their therapeutic development. In this preclinical study, conducted in Sprague Dawley rats, B6-SJL mice, and non-human primates (NHPs), we evaluated the G-protein-biased analogue of salvinorin A (SalA), 16-bromo salvinorin A (16-BrSalA), for its anticocaine effects, side effects, and activation of cellular signaling pathways. 16-BrSalA dose-dependently decreased the cocaine-primed reinstatement of drug-seeking behavior in a KOR-dependent manner. It also decreased cocaine-induced hyperactivity, but had no effect on responding for cocaine on a progressive ratio schedule. Compared to SalA, 16-BrSalA had an improved side effect profile, with no significant effects in the elevated plus maze, light-dark test, forced swim test, sucrose self-administration, or novel object recognition; however, it did exhibit conditioned aversive effects. 16-BrSalA increased dopamine transporter (DAT) activity in HEK-293 cells coexpressing DAT and KOR, as well as in rat nucleus accumbens and dorsal striatal tissue. 16-BrSalA also increased the early phase activation of extracellular-signal-regulated kinases 1 and 2, as well as p38 in a KOR-dependent manner. In NHPs, 16-BrSalA caused dose-dependent increases in the neuroendocrine biomarker prolactin, similar to other KOR agonists, at doses without robust sedative effects. These findings highlight that G-protein-biased structural analogues of SalA can have improved pharmacokinetic profiles and fewer side effects while maintaining their anticocaine effects.
Collapse
Affiliation(s)
- Ross van de Wetering
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Amy Ewald
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Susan Welsh
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Lindsay Kornberger
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA (T.E.P.)
| | - Samuel E. Williamson
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Bryan D. McElroy
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Eduardo R. Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - Thomas E. Prisinzano
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40506, USA (T.E.P.)
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS 66045, USA
| | - Bronwyn M. Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| |
Collapse
|
25
|
Margolis EB, Moulton MG, Lambeth PS, O'Meara MJ. The life and times of endogenous opioid peptides: Updated understanding of synthesis, spatiotemporal dynamics, and the clinical impact in alcohol use disorder. Neuropharmacology 2023; 225:109376. [PMID: 36516892 PMCID: PMC10548835 DOI: 10.1016/j.neuropharm.2022.109376] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The opioid G-protein coupled receptors (GPCRs) strongly modulate many of the central nervous system structures that contribute to neurological and psychiatric disorders including pain, major depressive disorder, and substance use disorders. To better treat these and related diseases, it is essential to understand the signaling of their endogenous ligands. In this review, we focus on what is known and unknown about the regulation of the over two dozen endogenous peptides with high affinity for one or more of the opioid receptors. We briefly describe which peptides are produced, with a particular focus on the recently proposed possible synthesis pathways for the endomorphins. Next, we describe examples of endogenous opioid peptide expression organization in several neural circuits and how they appear to be released from specific neural compartments that vary across brain regions. We discuss current knowledge regarding the strength of neural activity required to drive endogenous opioid peptide release, clues about how far peptides diffuse from release sites, and their extracellular lifetime after release. Finally, as a translational example, we discuss the mechanisms of action of naltrexone (NTX), which is used clinically to treat alcohol use disorder. NTX is a synthetic morphine analog that non-specifically antagonizes the action of most endogenous opioid peptides developed in the 1960s and FDA approved in the 1980s. We review recent studies clarifying the precise endogenous activity that NTX prevents. Together, the works described here highlight the challenges and opportunities the complex opioid system presents as a therapeutic target.
Collapse
Affiliation(s)
- Elyssa B Margolis
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, CA, USA.
| | - Madelyn G Moulton
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Philip S Lambeth
- UCSF Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew J O'Meara
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|