1
|
Bahji A, Bastien G, Bach P, Choi J, Le Foll B, Lim R, Jutras-Aswad D, Socias ME. The Association Between Self-Reported Anxiety and Retention in Opioid Agonist Therapy: Findings From a Canadian Pragmatic Trial. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2024; 69:172-182. [PMID: 37697811 PMCID: PMC10874605 DOI: 10.1177/07067437231194385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
BACKGROUND Prescription-type opioid use disorder (POUD) is often accompanied by comorbid anxiety, yet the impact of anxiety on retention in opioid agonist therapy (OAT) is unclear. Therefore, this study investigated whether baseline anxiety severity affects retention in OAT and whether this effect differs by OAT type (methadone maintenance therapy (MMT) vs. buprenorphine/naloxone (BNX)). METHODS This secondary analysis used data from a pan-Canadian randomized trial comparing flexible take-home dosing BNX and standard supervised MMT for 24 weeks. The study included 268 adults with POUD. Baseline anxiety was assessed using the Beck Anxiety Inventory (BAI), with BAI ≥ 16 indicating moderate-to-severe anxiety. The primary outcomes were retention in assigned and any OAT at week 24. In addition, the impact of anxiety severity on retention was examined, and assigned OAT was considered an effect modifier. RESULTS Of the participants, 176 (65%) reported moderate-to-severe baseline anxiety. In adjusted analyses, there was no significant difference in retention between those with BAI ≥ 16 and those with BAI < 16 assigned (29% vs. 28%; odds ratio (OR) = 2.03, 95% confidence interval (CI) = 0.94-4.40; P = 0.07) or any OAT (35% vs. 34%; OR = 1.57, 95% CI = 0.77-3.21; P = 0.21). In addition, there was no significant effect modification by OAT type for retention in assigned (P = 0.41) or any OAT (P = 0.71). In adjusted analyses, greater retention in treatment was associated with BNX (vs. MMT), male gender identity (vs. female, transgender, or other), enrolment in the Quebec study site (vs. other sites), and absence of a positive urine drug screen for stimulants at baseline. CONCLUSIONS Baseline anxiety severity did not significantly impact retention in OAT for adults with POUD, and there was no significant effect modification by OAT type. However, the overall retention rates were low, highlighting the need to develop new strategies to minimize the risk of attrition from treatment. CLINICAL TRIAL REGISTRATION This study was registered in ClinicalTrials.gov (NCT03033732).
Collapse
Affiliation(s)
- Anees Bahji
- British Columbia Centre on Substance Use, Vancouver, BC, Canada
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Gabriel Bastien
- Department of Psychiatry and Addictology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Research Centre, Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Paxton Bach
- British Columbia Centre on Substance Use, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - JinCheol Choi
- British Columbia Centre on Substance Use, Vancouver, BC, Canada
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Campbell Family Mental Health Research Institute, Center for Addiction and Mental Health, Toronto, ON, Canada
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical Sciences Building, University of Toronto, Toronto, ON, Canada
- Department of Family and Community Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - Ron Lim
- Department of Psychiatry, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Didier Jutras-Aswad
- Department of Psychiatry and Addictology, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
- Research Centre, Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - M. Eugenia Socias
- British Columbia Centre on Substance Use, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
2
|
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
|
3
|
Kong L, Shu X, Tang S, Ye R, Sun H, Jiang S, Li Z, Chai J, Fang Y, Lan Y, Yu L, Xie Q, Fu W, Wang Y, Li W, Qiu Z, Liu J, Shao L. SLL-627 Is a Highly Selective and Potent κ Opioid Receptor (KOR) Agonist with an Unexpected Nonreduction in Locomotor Activity. J Med Chem 2022; 65:10377-10392. [PMID: 35900351 DOI: 10.1021/acs.jmedchem.2c00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Undue central nervous system (CNS) side effects including dysphoria and sedation remain to be a challenge for the development of κ opioid receptor (KOR) agonists as effective and safe analgesics. On the basis of our previous work on morphinan-based KOR agonists, a series of 7α-methyl-7β-substituted northebaine derivatives were designed, synthesized, and biologically assayed. Among others, compound 4a (SLL-627) has been identified as a highly selective and potent KOR agonist both in vitro and in vivo, and its molecular basis was also examined and discussed. Besides low liability to conditioned place aversion (CPA) test, treatment of SLL-627 was associated with a nonreduction in locomotor activity, compared to most of the other arylacetamide- or morphinan-based KOR agonists which generally exhibited apparently sedative effects. This unexpected finding provides new insights to dissociate analgesia from sedation for future discovery of innovative KOR agonists.
Collapse
Affiliation(s)
- Linghui Kong
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Xuelian Shu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, No. 555 Zuchongzhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Siyuan Tang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Rongrong Ye
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, No. 100 Haiquan Road, Shanghai 201418, China
| | - Huijiao Sun
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Shuang Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, No. 138, Xianlin Road, Nanjing 210023, China
| | - Zixiang Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Jingrui Chai
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yun Fang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Yinjie Lan
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Linqian Yu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Qiong Xie
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Wei Fu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Yujun Wang
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Zhuibai Qiu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China
| | - Jinggen Liu
- CAS Key Laboratory of Receptor Research and State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences and Collaborative Innovation Center for Brain Science, No. 555 Zuchongzhi Road, Shanghai 201203, China
| | - Liming Shao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, China.,State Key Laboratory of Medical Neurobiology, Fudan University, No. 138 Yixueyuan Road, Shanghai 200032, China
| |
Collapse
|
4
|
Turan Yücel N, Evren AE, Kandemir Ü, Can ÖD. Antidepressant-like effect of tofisopam in mice: A behavioural, molecular docking and MD simulation study. J Psychopharmacol 2022; 36:819-835. [PMID: 35638175 DOI: 10.1177/02698811221095528] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Depression is a disease that affects millions of people worldwide, and the discovery and development of effective and safe antidepressant drugs is one of the important topics of psychopharmacology. OBJECTIVES In this study, it was aimed to investigate the antidepressant-like activity potential of tofisopam, an anxiolytic drug with 2,3-benzodiazepine structure, and to elucidate the pharmacological mechanisms mediating this effect. METHODS The antidepressant-like activity of tofisopam was investigated using tail suspension and modified forced swimming tests. Possible interactions of tofisopam with µ- and δ-opioid receptor subtypes were clarified by pharmacological antagonism, molecular docking and molecular dynamics simulation studies. RESULTS Tofisopam (50 and 100 mg/kg) significantly shortened the immobility time of mice in both the tail suspension and the modified forced swimming tests. The drug, at the same doses, prolonged the duration of swimming and climbing behaviours measured in modified forced swimming tests. A dosage of 25 mg/kg was ineffective. Mechanistic studies showed that the pretreatment with p-chlorophenylalanine methyl ester (serotonin synthesis inhibitor; 4 consecutive days, 100 mg/kg), α-methyl-para-tyrosine methyl ester (catecholamine synthesis inhibitor; 100 mg/kg), naloxonazine (selective µ-opioid receptor blocker, 7 mg/kg) and naltrindole (a selective δ-opioid receptor blocker, 0.99 mg/kg) abolished the anti-immobility effect induced by the 50 mg/kg dose of tofisopam in the tail suspension tests. Our in silico studies supported the behavioural findings that the antidepressant-like effect of tofisopam is mediated by μ- and δ-opioid receptors. CONCLUSION This study is the first to show that tofisopam has antidepressant-like activity mediated by the serotonergic, catecholaminergic and opioidergic systems.
Collapse
Affiliation(s)
- Nazlı Turan Yücel
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Asaf Evrim Evren
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey.,Pharmacy Services, Vocational School of Health Services, Bilecik Şeyh Edebali University, Bilecik, Turkey
| | - Ümmühan Kandemir
- Department of Pharmacology, Institute of Health Sciences, Anadolu University, Eskişehir, Turkey
| | - Özgür Devrim Can
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
5
|
Marton J, Fekete A, Cumming P, Hosztafi S, Mikecz P, Henriksen G. Diels-Alder Adducts of Morphinan-6,8-Dienes and Their Transformations. Molecules 2022; 27:2863. [PMID: 35566212 PMCID: PMC9102320 DOI: 10.3390/molecules27092863] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023] Open
Abstract
6,14-ethenomorphinans are semisynthetic opiate derivatives containing an ethylene bridge between positions 6 and 14 in ring-C of the morphine skeleton that imparts a rigid molecular structure. These compounds represent an important family of opioid receptor ligands in which the 6,14-etheno bridged structural motif originates from a [4 + 2] cycloaddition of morphinan-6,8-dienes with dienophiles. Certain 6,14-ethenomorphinans having extremely high affinity for opioid receptors are often non-selective for opioid receptor subtypes, but this view is now undergoing some revision. The agonist 20R-etorphine and 20R-dihydroetorphine are several thousand times more potent analgesics than morphine, whereas diprenorphine is a high-affinity non-selective antagonist. The partial agonist buprenorphine is used as an analgesic in the management of post-operative pain or in substitution therapy for opiate addiction, sometimes in combination with the non-selective antagonist naloxone. In the context of the current opioid crisis, we communicated a summary of several decades of work toward generating opioid analgesics with lesser side effects or abuse potential. Our summary placed a focus on Diels-Alder reactions of morphinan-6,8-dienes and subsequent transformations of the cycloadducts. We also summarized the pharmacological aspects of radiolabeled 6,14-ethenomorphinans used in molecular imaging of opioid receptors.
Collapse
Affiliation(s)
- János Marton
- ABX Advanced Biochemical Compounds Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Strasse 10-14, D-01454 Radeberg, Germany
| | - Anikó Fekete
- Department of Medical Imaging, Division of Nuclear Medicine and Translational Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (A.F.); (P.M.)
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Freiburgstraße 18, 3010 Bern, Switzerland;
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Sándor Hosztafi
- Institute of Pharmaceutical Chemistry, Semmelweis Medical University, Högyes Endre utca 9, H-1092 Budapest, Hungary;
| | - Pál Mikecz
- Department of Medical Imaging, Division of Nuclear Medicine and Translational Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary; (A.F.); (P.M.)
| | - Gjermund Henriksen
- Norwegian Medical Cyclotron Centre Ltd., Sognsvannsveien 20, N-0372 Oslo, Norway
- Institute of Basic Medical Sciences, University of Oslo, N-0317 Oslo, Norway
- Institute of Physics, University of Oslo, Sem Sælands vei 24, N-0371 Oslo, Norway
| |
Collapse
|
6
|
Namchuk AB, Lucki I, Browne CA. Buprenorphine as a Treatment for Major Depression and Opioid Use Disorder. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2022; 2:10254. [PMID: 36177442 PMCID: PMC9518754 DOI: 10.3389/adar.2022.10254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Rates of major depressive disorder (MDD) are disproportionally high in subjects with opioid use disorder (OUD) relative to the general population. MDD is often more severe in OUD patients, leading to compliance issues with maintenance therapies and poor outcomes. A growing body of literature suggests that endogenous opioid system dysregulation may play a role in the emergence of MDD. Buprenorphine, a mixed opioid receptor agonist/antagonist approved for the treatment of OUD and chronic pain, may have potential as a novel therapeutic for MDD, especially for patients with a dual diagnosis of MDD and OUD. This paper presents a comprehensive review of papers relevant to the assessment of buprenorphine as a treatment for MDD, OUD, and/or suicide compiled using electronic databases per Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. The principal goal of this literature review was to compile the clinical studies that have interrogated the antidepressant activity of buprenorphine in opioid naïve MDD patients and OUD patients with comorbid MDD. Evidence supporting buprenorphine's superiority over methadone for treating comorbid OUD and MDD was also considered. Finally, recent evidence for the ability of buprenorphine to alleviate suicidal ideation in both opioid-naïve patients and opioid-experienced patients was evaluated. Synthesizing all of this information, buprenorphine emerges as a potentially effective therapeutic for the dual purposes of treating MDD and OUD.
Collapse
Affiliation(s)
- Amanda B. Namchuk
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, Maryland, 20814, USA
| | - Irwin Lucki
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, Maryland, 20814, USA
- Department of Psychiatry, Uniformed Services University, Bethesda, Maryland, 20814, USA
| | - Caroline A. Browne
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University, Bethesda, Maryland, 20814, USA
| |
Collapse
|
7
|
Hillhouse TM, Olson KM, Hallahan JE, Rysztak LG, Sears BF, Meurice C, Ostovar M, Koppenhaver PO, West JL, Jutkiewicz EM, Husbands SM, Traynor JR. The Buprenorphine Analogue BU10119 Attenuates Drug-Primed and Stress-Induced Cocaine Reinstatement in Mice. J Pharmacol Exp Ther 2021; 378:287-299. [PMID: 34183434 PMCID: PMC11047085 DOI: 10.1124/jpet.121.000524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/22/2021] [Indexed: 01/01/2023] Open
Abstract
There are no Food and Drug Administration-approved medications for cocaine use disorder, including relapse. The μ-opioid receptor (MOPr) partial agonist buprenorphine alone or in combination with naltrexone has been shown to reduce cocaine-positive urine tests and cocaine seeking in rodents. However, there are concerns over the abuse liability of buprenorphine. Buprenorphine's partial agonist and antagonist activity at the nociception receptor (NOPr) and κ-opioid receptor (KOPr), respectively, may contribute to its ability to inhibit cocaine seeking. Thus, we hypothesized that a buprenorphine derivative that exhibits antagonist activity at MOPr and KOPr with enhanced agonist activity at the NOPr could provide a more effective treatment. Here we compare the pharmacology of buprenorphine and two analogs, BU10119 and BU12004, in assays for antinociception and for cocaine- and stress-primed reinstatement in the conditioned place preference paradigm. In vitro and in vivo assays showed that BU10119 acts as an antagonist at MOPr, KOPr, and δ-opioid receptor (DOPr) and a partial agonist at NOPr, whereas BU12004 showed MOPr partial agonist activity and DOPr, KOPr, and NOPr antagonism. BU10119 and buprenorphine but not BU12004 lessened cocaine-primed reinstatement. In contrast, BU10119, BU12004, and buprenorphine blocked stress-primed reinstatement. The selective NOPr agonist SCH221510 but not naloxone decreased cocaine-primed reinstatement. Together, these findings are consistent with the concept that NOPr agonism contributes to the ability of BU10119 and buprenorphine to attenuate reinstatement of cocaine-conditioned place preference in mice. The findings support the development of buprenorphine analogs lacking MOPr agonism with increased NOPr agonism for relapse prevention to cocaine addiction. SIGNIFICANCE STATEMENT: There are no Food and Drug Administration-approved medications for cocaine use disorder. Buprenorphine has shown promise as a treatment for cocaine relapse prevention; however, there are concerns over the abuse liability of buprenorphine. Here we show a buprenorphine analogue, BU10119, which lacks μ-opioid receptor agonism and inhibits cocaine-primed and stress-primed reinstatement in a conditioned place-preference paradigm. The results suggest the development of BU10119 for the management of relapse to cocaine seeking.
Collapse
MESH Headings
- Animals
- Buprenorphine/pharmacology
- Buprenorphine/analogs & derivatives
- Mice
- Male
- Cocaine/pharmacology
- Stress, Psychological/drug therapy
- Stress, Psychological/metabolism
- Cocaine-Related Disorders/drug therapy
- Mice, Inbred C57BL
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/metabolism
- Drug-Seeking Behavior/drug effects
- Humans
- Receptors, Opioid/metabolism
- Receptors, Opioid/agonists
- Narcotic Antagonists/pharmacology
- Receptors, Opioid, kappa/agonists
- Receptors, Opioid, kappa/metabolism
Collapse
Affiliation(s)
- Todd M Hillhouse
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Keith M Olson
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - James E Hallahan
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Lauren G Rysztak
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Bryan F Sears
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Claire Meurice
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Mehrnoosh Ostovar
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Peyton O Koppenhaver
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Joshua L West
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Emily M Jutkiewicz
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - Stephen M Husbands
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| | - John R Traynor
- Department of Psychology, University of Wisconsin Green Bay, Green Bay, Wisconsin (T.M.H., P.O.K.); Department of Pharmacology and Edward F. Domino Research Center, University of Michigan Medical School, Ann Arbor, Michigan (K.M.O., J.E.H., L.G.R., B.F.S., C.M., J.W., E.M.J., J.R.T.); Department of Pharmacy and Pharmacology, and Center for Therapeutic Innovation, University of Bath, Bath, United Kingdom (M.O., S.M.H.); and Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.R.T.)
| |
Collapse
|
8
|
Translational value of non-human primates in opioid research. Exp Neurol 2021; 338:113602. [PMID: 33453211 DOI: 10.1016/j.expneurol.2021.113602] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/06/2021] [Accepted: 01/10/2021] [Indexed: 01/02/2023]
Abstract
Preclinical opioid research using animal models not only provides mechanistic insights into the modulation of opioid analgesia and its associated side effects, but also validates drug candidates for improved treatment options for opioid use disorder. Non-human primates (NHPs) have served as a surrogate species for humans in opioid research for more than five decades. The translational value of NHP models is supported by the documented species differences between rodents and primates regarding their behavioral and physiological responses to opioid-related ligands and that NHP studies have provided more concordant results with human studies. This review highlights the utilization of NHP models in five aspects of opioid research, i.e., analgesia, abuse liability, respiratory depression, physical dependence, and pruritus. Recent NHP studies have found that (1) mixed mu opioid and nociceptin/orphanin FQ peptide receptor partial agonists appear to be safe, non-addictive analgesics and (2) mu opioid receptor- and mixed opioid receptor subtype-based medications remain the only two classes of drugs that are effective in alleviating opioid-induced adverse effects. Given the recent advances in pharmaceutical sciences and discoveries of novel targets, NHP studies are posed to identify the translational gap and validate therapeutic targets for the treatment of opioid use disorder. Pharmacological studies using NHPs along with multiple outcome measures (e.g., behavior, physiologic function, and neuroimaging) will continue to facilitate the research and development of improved medications to curb the opioid epidemic.
Collapse
|
9
|
Abstract
This paper is the forty-first consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2018 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (2), the roles of these opioid peptides and receptors in pain and analgesia in animals (3) and humans (4), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (5), opioid peptide and receptor involvement in tolerance and dependence (6), stress and social status (7), learning and memory (8), eating and drinking (9), drug abuse and alcohol (10), sexual activity and hormones, pregnancy, development and endocrinology (11), mental illness and mood (12), seizures and neurologic disorders (13), electrical-related activity and neurophysiology (14), general activity and locomotion (15), gastrointestinal, renal and hepatic functions (16), cardiovascular responses (17), respiration and thermoregulation (18), and immunological responses (19).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY, 11367, United States.
| |
Collapse
|
10
|
Abstract
Buprenorphine has not only had an interdisciplinary impact on our understanding of key neuroscience topics like opioid pharmacology, pain signaling, and reward processing but has also been a key influence in changing the way that substance use disorders are approached in modern medical systems. From its leading role in expanding outpatient treatment of opioid use disorders to its continued influence on research into next-generation analgesics, buprenorphine has been a continuous player in the ever-evolving societal perception of opioids and substance use disorder. To provide a multifaceted account on the enormous diversity of areas where this molecule has made an impact, this article discusses buprenorphine's chemical properties, synthesis and development, pharmacology, adverse effects, manufacturing information, and historical place in the field of chemical neuroscience.
Collapse
Affiliation(s)
- Jillian L. Kyzer
- University of Wisconsin-Madison, School of Pharmacy, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Cody J. Wenthur
- University of Wisconsin-Madison, School of Pharmacy, 777 Highland Avenue, Madison, Wisconsin 53705, United States
| |
Collapse
|
11
|
Piekielna-Ciesielska J, Wtorek K, Janecka A. Biased Agonism as an Emerging Strategy in the Search for Better Opioid Analgesics. Curr Med Chem 2020; 27:1562-1575. [PMID: 31057099 DOI: 10.2174/0929867326666190506103124] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 12/12/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022]
Abstract
Morphine and related drugs that act through activating opioid receptors are the most effective analgesics for the relief of severe pain. They have been used for decades, despite the range of unwanted side effects that they produce, as no alternative has been found so far. The major goal of opioid research is to understand the mechanism of action of opioid receptor agonists and to improve the therapeutic utility of opioid drugs. In the search for safer and more potent analgesics, analogs with mixed opioid receptor profile gained a lot of interest. However, recently the concept of biased agonism, that highlights the fact that some ligands are able to differentially activate receptor downstream pathways, became a new approach in the design of novel drug candidates for clinical application. In this review, we summarize current knowledge on the development of opioid ligands of peptide and nonpeptide structure, showing how much opioid pharmacology evolved in recent years.
Collapse
Affiliation(s)
| | - Karol Wtorek
- Department of Biomolecular Chemistry, Medical University, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University, Mazowiecka 6/8, 92-215 Lodz, Poland
| |
Collapse
|
12
|
Jacobson ML, Browne CA, Lucki I. Kappa Opioid Receptor Antagonists as Potential Therapeutics for Stress-Related Disorders. Annu Rev Pharmacol Toxicol 2020; 60:615-636. [DOI: 10.1146/annurev-pharmtox-010919-023317] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Exposure to stressful stimuli activates kappa opioid receptor (KOR) signaling, a process known to produce aversion and dysphoria in humans and other species. This endogenous opioid system is dysregulated in stress-related disorders, specifically in major depressive disorder (MDD). These findings serve as the foundation for a growing interest in the therapeutic potential of KOR antagonists as novel antidepressants. In this review, data supporting the hypothesis of dysregulated KOR function in MDD are considered. The clinical data demonstrating the therapeutic efficacy and safety of selective and mixed opioid antagonists are then presented. Finally, the preclinical evidence illustrating the induction of behaviors relevant to the endophenotypes of MDD and KOR antagonist activity in stress-naïve and stress-exposed animals is evaluated. Overall, this review highlights the emergent literature supporting the pursuit of KOR antagonists as novel therapeutics for MDD and other stress-related disorders.
Collapse
Affiliation(s)
- Moriah L. Jacobson
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Caroline A. Browne
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Irwin Lucki
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| |
Collapse
|
13
|
Charles SJ, Farias M, Dunbar RI. The aetiology of social deficits within mental health disorders: The role of the immune system and endogenous opioids. Brain Behav Immun Health 2020; 1:100003. [PMID: 38377411 PMCID: PMC8474498 DOI: 10.1016/j.bbih.2019.100003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 09/30/2019] [Indexed: 10/25/2022] Open
Abstract
The American National Institute for Mental Health (NIMH) has put out a set of research goals that include a long-term plan to identify more reliable endogenous explanations for a wide variety of mental health disorders (Insel, 2013). In response to this, we have identified a major symptom that underlies multiple mental health disorders - social bonding dysfunction. We suggest that endogenous opioid abnormalities can lead to altered social bonding, which is a symptom of various mental health disorders, including depression, schizophrenia and ASD. This article first outlines how endogenous opioids play a role in social bonding. Then we show their association with the body's inflammation immune function, and review recent literature linking inflammation to mental health 'immunophenotypes'. We finish by explaining how these immunophenotypes may be caused by alterations in the endogenous opioid system. This is the first overview of the role of inflammation across multiple disorders where we provide a biochemical explanation for why immunophenotypes might exist across diagnoses. We propose a novel mechanism of how the immune system may be causing 'sickness-type' behaviours (fatigue, appetite change, social withdrawal and inhibited motivation) in those who have these immunophenotypes. We hope that this novel aetiology can be used as a basis for future research in mental health.
Collapse
Affiliation(s)
- Sarah J. Charles
- Brain, Belief and Behaviour Research Lab, Centre for Trust Peace and Social Relations, Coventry University, United Kingdom
| | - Miguel Farias
- Brain, Belief and Behaviour Research Lab, Centre for Trust Peace and Social Relations, Coventry University, United Kingdom
| | - Robin I.M. Dunbar
- Department of Experimental Psychology, University of Oxford, United Kingdom
| |
Collapse
|
14
|
Maguire DR, Gerak LR, Cami-Kobeci G, Husbands SM, France CP, Belli B, Flynn P. OREX-1019: A Novel Treatment of Opioid Use Disorder and Relapse Prevention. J Pharmacol Exp Ther 2019; 372:205-215. [PMID: 31748404 DOI: 10.1124/jpet.119.261511] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/13/2019] [Indexed: 12/16/2022] Open
Abstract
There is an urgent need for new pharmacological treatments for substance use disorders, including opioid use disorder, particularly for use in relapse prevention. A combination of buprenorphine with naltrexone has shown particular promise, with clinical studies indicating a substantial improvement over treatment with naltrexone alone. OREX-1019 (formerly BU10119) is a compound that mimics the pharmacology of the buprenorphine/naltrexone combination. This study evaluated, in rhesus monkeys, the therapeutic potential of OREX-1019 for treating opioid use disorder. Pretreatment with OREX-1019 (0.01-0.3 mg/kg s.c.) dose-dependently decreased responding for the μ opioid receptor agonist remifentanil in rhesus monkeys but did not maintain levels of responding above vehicle when it was available for self-administration. OREX-1019 (0.01-1.0 mg/kg s.c.) also decreased cue- plus heroin-primed reinstatement of extinguished responding in monkeys that self-administered remifentanil but did not alter cue- plus cocaine-primed reinstatement of responding in monkeys that self-administered cocaine. OREX-1019 (0.3 mg/kg s.c.), like naltrexone (0.1 mg/kg s.c.), increased heart rate and blood pressure, produced overt observable signs, and eliminated food-maintained responding in monkeys treated chronically with morphine. These results confirm that OREX-1019 has little or no efficacy at μ opioid receptorsand has low abuse potential, and, combined with promising safety (clean profile vs. other off-target proteins including the hERG (human ether-a-go-go-related gene) K+ channel) and pharmacokinetic data (supporting administration by subcutaneous or sublingual routes, but with low oral bioavailability), suggest it could be a safe and effective alternative to current treatments for opioid use disorders particularly as applied to relapse prevention. SIGNIFICANCE STATEMENT: The novel opioid OREX-1019 potentially provides an improved relapse prevention agent for use in opioid use disorder. The current study demonstrates that in monkeys OREX-1019 is able to inhibit the self-administration of, and cue- plus heroin-primed reinstatement of, responding previously maintained by remifentanil.
Collapse
Affiliation(s)
- David R Maguire
- Departments of Pharmacology (D.R.M., L.R.G., C.P.F.) and Psychiatry (C.P.F.), and Addiction Research, Treatment & Training Center of Excellence (D.R.M., L.R.G., C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Pharmacy and Pharmacology (G.C.-K., S.M.H.) and Centre for Therapeutic Innovation (S.M.H.), University of Bath, Bath, United Kingdom; and Orexigen Therapeutics, La Jolla, California (B.B., P.F.)
| | - Lisa R Gerak
- Departments of Pharmacology (D.R.M., L.R.G., C.P.F.) and Psychiatry (C.P.F.), and Addiction Research, Treatment & Training Center of Excellence (D.R.M., L.R.G., C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Pharmacy and Pharmacology (G.C.-K., S.M.H.) and Centre for Therapeutic Innovation (S.M.H.), University of Bath, Bath, United Kingdom; and Orexigen Therapeutics, La Jolla, California (B.B., P.F.)
| | - Gerta Cami-Kobeci
- Departments of Pharmacology (D.R.M., L.R.G., C.P.F.) and Psychiatry (C.P.F.), and Addiction Research, Treatment & Training Center of Excellence (D.R.M., L.R.G., C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Pharmacy and Pharmacology (G.C.-K., S.M.H.) and Centre for Therapeutic Innovation (S.M.H.), University of Bath, Bath, United Kingdom; and Orexigen Therapeutics, La Jolla, California (B.B., P.F.)
| | - Stephen M Husbands
- Departments of Pharmacology (D.R.M., L.R.G., C.P.F.) and Psychiatry (C.P.F.), and Addiction Research, Treatment & Training Center of Excellence (D.R.M., L.R.G., C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Pharmacy and Pharmacology (G.C.-K., S.M.H.) and Centre for Therapeutic Innovation (S.M.H.), University of Bath, Bath, United Kingdom; and Orexigen Therapeutics, La Jolla, California (B.B., P.F.)
| | - Charles P France
- Departments of Pharmacology (D.R.M., L.R.G., C.P.F.) and Psychiatry (C.P.F.), and Addiction Research, Treatment & Training Center of Excellence (D.R.M., L.R.G., C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Pharmacy and Pharmacology (G.C.-K., S.M.H.) and Centre for Therapeutic Innovation (S.M.H.), University of Bath, Bath, United Kingdom; and Orexigen Therapeutics, La Jolla, California (B.B., P.F.)
| | - Barbara Belli
- Departments of Pharmacology (D.R.M., L.R.G., C.P.F.) and Psychiatry (C.P.F.), and Addiction Research, Treatment & Training Center of Excellence (D.R.M., L.R.G., C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Pharmacy and Pharmacology (G.C.-K., S.M.H.) and Centre for Therapeutic Innovation (S.M.H.), University of Bath, Bath, United Kingdom; and Orexigen Therapeutics, La Jolla, California (B.B., P.F.)
| | - Peter Flynn
- Departments of Pharmacology (D.R.M., L.R.G., C.P.F.) and Psychiatry (C.P.F.), and Addiction Research, Treatment & Training Center of Excellence (D.R.M., L.R.G., C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas; Department of Pharmacy and Pharmacology (G.C.-K., S.M.H.) and Centre for Therapeutic Innovation (S.M.H.), University of Bath, Bath, United Kingdom; and Orexigen Therapeutics, La Jolla, California (B.B., P.F.)
| |
Collapse
|
15
|
Insights into the Neurobiology of Anxiety and a Potential Target for Pharmacotherapy. J Neurosci 2019; 38:8919-8921. [PMID: 30333184 DOI: 10.1523/jneurosci.1461-18.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/27/2018] [Accepted: 09/02/2018] [Indexed: 11/21/2022] Open
|
16
|
Wu J, Jiang Q, Zhu H, Zhou Y, Lu D, Liu X, Chen X, Chen J, Wang Y, Liu J, Song R, Huang R, Zhou H. Uncovering kappa-opioid receptor agonist-induced PAK1/2 phosphorylation by quantitative phosphoproteomics. Biochem Biophys Res Commun 2019; 516:320-326. [DOI: 10.1016/j.bbrc.2019.06.111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/20/2019] [Indexed: 12/15/2022]
|
17
|
Browne CA, Lucki I. Targeting opioid dysregulation in depression for the development of novel therapeutics. Pharmacol Ther 2019; 201:51-76. [PMID: 31051197 DOI: 10.1016/j.pharmthera.2019.04.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 04/23/2019] [Indexed: 02/07/2023]
Abstract
Since the serendipitous discovery of the first class of modern antidepressants in the 1950's, all pharmacotherapies approved by the Food and Drug Administration for major depressive disorder (MDD) have shared a common mechanism of action, increased monoaminergic neurotransmission. Despite the widespread availability of antidepressants, as many as 50% of depressed patients are resistant to these conventional therapies. The significant length of time required to produce meaningful symptom relief with these medications, 4-6 weeks, indicates that other mechanisms are likely involved in the pathophysiology of depression which may yield more viable targets for drug development. For decades, no viable candidate target with a different mechanism of action to that of conventional therapies proved successful in clinical studies. Now several exciting avenues for drug development are under intense investigation. One of these emerging targets is modulation of endogenous opioid tone. This review will evaluate preclinical and clinical evidence pertaining to opioid dysregulation in depression, focusing on the role of the endogenous ligands endorphin, enkephalin, dynorphin, and nociceptin/orphanin FQ (N/OFQ) and their respective receptors, mu (MOR), delta (DOR), kappa (KOR), and the N/OFQ receptor (NOP) in mediating behaviors relevant to depression and anxiety. Finally, putative opioid based antidepressants that are under investigation in clinical trials, ALKS5461, JNJ-67953964 (formerly LY2456302 and CERC-501) and BTRX-246040 (formerly LY-2940094) will be discussed. This review will illustrate the potential therapeutic value of targeting opioid dysregulation in developing novel therapies for MDD.
Collapse
Affiliation(s)
- Caroline A Browne
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America
| | - Irwin Lucki
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America.
| |
Collapse
|
18
|
Cunningham CW, Elballa WM, Vold SU. Bifunctional opioid receptor ligands as novel analgesics. Neuropharmacology 2019; 151:195-207. [PMID: 30858102 DOI: 10.1016/j.neuropharm.2019.03.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/30/2019] [Accepted: 03/02/2019] [Indexed: 12/12/2022]
Abstract
Prolonged treatment of chronic severe pain with opioid analgesics is frought with problematic adverse effects including tolerance, dependence, and life-threatening respiratory depression. Though these effects are mediated predominately through preferential activation of μ opioid peptide (μOP) receptors, there is an emerging appreciation that actions at κOP and δOP receptors contribute to the observed pharmacologic and behavioral profile of μOP receptor agonists and may be targeted simultaneously to afford improved analgesic effects. Recent developments have also identified the related nociceptin opioid peptide (NOP) receptor as a key modulator of the effects of μOP receptor signaling. We review here the available literature describing OP neurotransmitter systems and highlight recent drug and probe design strategies.
Collapse
Affiliation(s)
| | - Waleed M Elballa
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, Mequon, WI, USA.
| | - Stephanie U Vold
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, Mequon, WI, USA.
| |
Collapse
|
19
|
Bidlack JM, Knapp BI, Deaver DR, Plotnikava M, Arnelle D, Wonsey AM, Fern Toh M, Pin SS, Namchuk MN. In Vitro Pharmacological Characterization of Buprenorphine, Samidorphan, and Combinations Being Developed as an Adjunctive Treatment of Major Depressive Disorder. J Pharmacol Exp Ther 2018; 367:267-281. [PMID: 30108159 DOI: 10.1124/jpet.118.249839] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022] Open
Abstract
A combination of buprenorphine (BUP) and samidorphan (SAM) at a 1:1 (mg/mg) fixed-ratio dose is being investigated as an adjunctive treatment of major depressive disorder (BUP/SAM, ALKS 5461). Both [3H]BUP and [3H]SAM bound to the μ-, κ-, and δ-opioid receptors (MOR, KOR, and DOR, respectively) with Kd values of 3 nM or less. [3H]BUP dissociated from the MOR more slowly than [3H]SAM did. In the [35S]GTPγS assay, BUP was a partial agonist at the MOR, KOR, and DOR. SAM was an antagonist at the MOR and a partial agonist at the KOR and DOR. The pharmacology of the combination of SAM and BUP was characterized at ratios like the molar ratios of both compounds at steady state in humans. In all assessments, SAM reduced the efficacy of BUP at the MOR without altering its potency. At the KOR, SAM had no significant effect on the activity of BUP. In bioluminescent resonance energy transfer assays, SAM, naltrexone, and naloxone were partial agonists when the MOR was coupled to the Gα oB and Gα z, and were antagonists when coupled to Gα i At the KOR, SAM was a partial agonist activating Gα oA and Gα oB and a full agonist in stimulating Gα z SAM inhibited BUP's recruitment of β-arrestin to the MOR, suggesting an attenuation of BUP's efficacy in activating G proteins correlated with an inhibition of β-arrestin recruitment. The collective data suggest that SAM attenuates the efficacy of BUP under all conditions tested at the MOR and DOR but had little effect on BUP activity at the KOR.
Collapse
Affiliation(s)
- Jean M Bidlack
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Brian I Knapp
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Daniel R Deaver
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Margarita Plotnikava
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Derrick Arnelle
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Angela M Wonsey
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - May Fern Toh
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Sokhom S Pin
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| | - Mark N Namchuk
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, New York (J.M.B., B.I.K.) and Alkermes, Inc., Waltham, Massachusetts (D.R.D., M.P., D.A., A.M.W., M.F.T., S.S.P., M.N.N.)
| |
Collapse
|
20
|
Abstract
This themed section of the British Journal of Pharmacology stems from an International Narcotics Research Conference (INRC) meeting held in July 2016 at The Assembly Rooms in Bath, UK. LINKED ARTICLES This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.
Collapse
Affiliation(s)
- Eamonn Kelly
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Graeme Henderson
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Chris P Bailey
- Department of Pharmacy and PharmacologyUniversity of BathBathUK
| |
Collapse
|
21
|
Gach-Janczak K, Piekielna-Ciesielska J, Adamska-Bartłomiejczyk A, Wtorek K, Ferrari F, Calo' G, Szymaszkiewicz A, Piasecka-Zelga J, Janecka A. In vitro and in vivo activity of cyclopeptide Dmt-c[d-Lys-Phe-Asp]NH 2, a mu opioid receptor agonist biased toward β-arrestin. Peptides 2018; 105:51-57. [PMID: 29684591 DOI: 10.1016/j.peptides.2018.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022]
Abstract
Morphine and related drugs, which are the most effective analgesics for the relief of severe pain, act through activating opioid receptors. The endogenous ligands of these receptors are opioid peptides which cannot be used as antinociceptive agents due to their low bioactivity and stability in biological fluids. The major goal of opioid research is to understand the mechanism of action of opioid receptor agonists in order to improve therapeutic utility of opioids. Analgesic effects of morphine are mediated mostly through activation of the mu opioid receptor. However, in the search for safer and more effective drug candidates, analogs with mixed opioid receptor profile gained a lot of interest. Recently, the concept of biased agonists able to differentially activate GPCR downstream pathways, became a new approach in the design of novel drug candidates. It is hypothesized that compounds promoting G-protein signaling may produce analgesia while β-arrestin recruitment may be responsible for opioid side effects. In this report we showed that replacement of the tyrosine residue in the mu-selective ligand Tyr-c[d-Lys-Phe-Asp]NH2 with 2',6'-dimethyltyrosine (Dmt) produced a cyclopeptide Dmt-c[d-Lys-Phe-Asp]NH2 with mu/delta opioid receptor agonist profile. This analog showed improved antinociception in the hot-plate test, probably due to the simultaneous activation of mu and delta receptors but also significantly inhibited the gastrointestinal transit. Using the bioluminescence resonance energy transfer (BRET) assay it was shown that this analog was a mu receptor agonist biased toward β-arrestin. β-Arrestin-dependent signaling is most likely responsible for the observed inhibition of gastrointestinal motility exerted by the novel cyclopeptide.
Collapse
Affiliation(s)
| | | | | | - Karol Wtorek
- Department of Biomolecular Chemistry, Medical University, Lodz, Poland
| | - Federica Ferrari
- Department of Medical Sciences, Section of Pharmacology and Italian Institute of Neuroscience, University of Ferrara, 44121 Ferrara, Italy
| | - Girolamo Calo'
- Department of Medical Sciences, Section of Pharmacology and Italian Institute of Neuroscience, University of Ferrara, 44121 Ferrara, Italy
| | - Agata Szymaszkiewicz
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Poland
| | - Joanna Piasecka-Zelga
- Institute of Occupational Medicine, Research Laboratory for Medicine and Veterinary Products in the GMP Head of Research Laboratory for Medicine and Veterinary Products, Lodz, Poland
| | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University, Lodz, Poland.
| |
Collapse
|
22
|
Bailey S, Husbands S. Targeting opioid receptor signaling in depression: do we need selective κ opioid receptor antagonists? Neuronal Signal 2018; 2:NS20170145. [PMID: 32714584 PMCID: PMC7373229 DOI: 10.1042/ns20170145] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 12/15/2022] Open
Abstract
The opioid receptors are a family of G-protein coupled receptors (GPCRs) with close structural homology. The opioid receptors are activated by a variety of endogenous opioid neuropeptides, principally β-endorphin, dynorphins, leu- and met-enkephalins. The clinical potential of targeting opioid receptors has largely focused on the development of analgesics. However, more recent attention has turned to the role of central opioid receptors in the regulation of stress responses, anhedonia and mood. Activation of the κ opioid receptor (KOP) subtype has been shown in both human and rodent studies to produce dysphoric and pro-depressive like effects. This has led to the idea that selective KOP antagonists might have therapeutic potential as antidepressants. Here we review data showing that mixed μ opioid (MOP) and KOP antagonists have antidepressant-like effects in rodent behavioural paradigms and highlight comparable studies in treatment-resistant depressed patients. We propose that developing multifunctional ligands which target multiple opioid receptors open up the potential for fine-tuning hedonic responses mediated by opioids. This alternative approach towards targeting multiple opioid receptors may lead to more effective treatments for depression.
Collapse
Affiliation(s)
- Sarah J. Bailey
- Drug and Target Discovery, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, U.K
| | - Stephen M. Husbands
- Drug and Target Discovery, Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, U.K
| |
Collapse
|
23
|
McHugh KL, Kelly JP. Modulation of the central opioid system as an antidepressant target in rodent models. PROGRESS IN BRAIN RESEARCH 2018; 239:49-87. [DOI: 10.1016/bs.pbr.2018.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
24
|
Almatroudi A, Ostovar M, Bailey CP, Husbands SM, Bailey SJ. Antidepressant-like effects of BU10119, a novel buprenorphine analogue with mixed κ/μ receptor antagonist properties, in mice. Br J Pharmacol 2017; 175:2869-2880. [PMID: 28967123 DOI: 10.1111/bph.14060] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/28/2017] [Accepted: 08/07/2017] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND AND PURPOSE The κ receptor antagonists have potential for treating neuropsychiatric disorders. We have investigated the in vivo pharmacology of a novel buprenorphine analogue, BU10119, for the first time. EXPERIMENTAL APPROACH To determine the opioid pharmacology of BU10119 (0.3-3 mg·kg-1 , i.p.) in vivo, the warm-water tail-withdrawal assay was applied in adult male CD1 mice. A range of behavioural paradigms was used to investigate the locomotor effects, rewarding properties and antidepressant or anxiolytic potential of BU10119. Additional groups of mice were exposed to a single (1 × 2 h) or repeated restraint stress (3× daily 2 h) to determine the ability of BU10119 to block stress-induced analgesia. KEY RESULTS BU10119 alone was without any antinociceptive activity. BU10119 (1 mg·kg-1 ) was able to block U50,488, buprenorphine and morphine-induced antinociception. The κ antagonist effects of BU10119 in the tail-withdrawal assay reversed between 24 and 48 h. BU10119 was without significant locomotor or rewarding effects. BU10119 (1 mg·kg-1 ) significantly reduced the latency to feed in the novelty-induced hypophagia task and reduced immobility time in the forced swim test, compared to saline-treated animals. There were no significant effects of BU10119 in either the elevated plus maze or the light-dark box. Both acute and repeated restraint stress-induced analgesia were blocked by pretreatment with BU10119 (1 mg·kg-1 ). Parallel stress-induced increases in plasma corticosterone were not affected. CONCLUSIONS AND IMPLICATIONS BU10119 is a mixed κ/μ receptor antagonist with relatively short-duration κ antagonist activity. Based on these preclinical data, BU10119 has therapeutic potential for the treatment of depression and other stress-induced conditions. LINKED ARTICLES This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.
Collapse
Affiliation(s)
| | - Mehrnoosh Ostovar
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | | | | | - Sarah J Bailey
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| |
Collapse
|