1
|
Hammond HR, Chaudhari P, Bunnell A, Nefzi K, Chen C, Zhao P, Eans SO, Masood SR, Dooley CT, Liu-Chen LY, McLaughlin JP, Nefzi A. Peripherally Restricted Fused Heterocyclic Peptidomimetic Multifunctional Opioid Agonists as Novel, Potent Analgesics. ACS Med Chem Lett 2025; 16:388-396. [PMID: 40104791 PMCID: PMC11912268 DOI: 10.1021/acsmedchemlett.4c00333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 03/20/2025] Open
Abstract
Heterocyclic peptidomimetics are constrained compounds that mimic the biological efficacy of peptides while offering increased stability. We have previously generated a diazaheterocyclic peripherally selective, mixed-opioid agonist peptidomimetic that produced synergistic antinociception with decreased side effects. Working from two earlier templates, we report here the synthesis of 15 new diazaheterocyclic analogues. In vitro screening with radioligand competition binding assays and [35S]GTPγS assays demonstrated variable affinity for and activity at μ (MOR), δ (DOR), and κ (KOR) opioid receptors across the series, with three (2663-48, 2638-28 and 2638-33) displaying good affinity for DOR and/or KOR. All three compounds produced dose-dependent, opioid-receptor mediated antinociception in the mouse 55 °C warm-water tail-withdrawal and acetic-acid writhing assay, although a ratio of ED50 values in these assays suggested poor BBB penetration by 2638-33; results confirmed by testing with naloxone-methiodide. The data suggest these diazaheterocyclic mixed-activity, peripherally restricted opioid receptor agonists may hold potential as new, safer analgesics.
Collapse
Affiliation(s)
- Haylee R Hammond
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Prakash Chaudhari
- Department of Cellular Biology & Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Center for Translational Science, Port Saint Lucie, Florida 34987, United States
| | - Ashley Bunnell
- Department of Cellular Biology & Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Center for Translational Science, Port Saint Lucie, Florida 34987, United States
| | - Khadija Nefzi
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Chongguang Chen
- Center for Substance Abuse Research and Biochemical Pharmacology Core, Lewis Katz School of Medicine, Temple University, 3500 N. Broad Street, Eighth Floor, Philadelphia, Pennsylvania 19140, United States
| | - Pingwei Zhao
- Center for Substance Abuse Research and Biochemical Pharmacology Core, Lewis Katz School of Medicine, Temple University, 3500 N. Broad Street, Eighth Floor, Philadelphia, Pennsylvania 19140, United States
| | - Shainnel O Eans
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Sabrina R Masood
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Colette T Dooley
- Technical Writer-Editor-Reviewer, Vero Beach, Florida 32960United States
| | - Lee-Yuan Liu-Chen
- Center for Substance Abuse Research and Biochemical Pharmacology Core, Lewis Katz School of Medicine, Temple University, 3500 N. Broad Street, Eighth Floor, Philadelphia, Pennsylvania 19140, United States
| | - Jay P McLaughlin
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Adel Nefzi
- Department of Cellular Biology & Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Center for Translational Science, Port Saint Lucie, Florida 34987, United States
- Department of Chemistry and Biochemistry, the School of Integrated Science and Humanity, College of Arts, Sciences & Education. Florida International University, 11200 S.W. Eighth Street, Miami, Florida 33199, United States
| |
Collapse
|
2
|
Gao Z, Ding P, Xu R. IUPHAR review - Data-driven computational drug repurposing approaches for opioid use disorder. Pharmacol Res 2024; 199:106960. [PMID: 37832859 DOI: 10.1016/j.phrs.2023.106960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/15/2023]
Abstract
Opioid Use Disorder (OUD) is a chronic and relapsing condition characterized by the misuse of opioid drugs, causing significant morbidity and mortality in the United States. Existing medications for OUD are limited, and there is an immediate need to discover treatments with enhanced safety and efficacy. Drug repurposing aims to find new indications for existing medications, offering a time-saving and cost-efficient alternative strategy to traditional drug discovery. Computational approaches have been developed to further facilitate the drug repurposing process. In this paper, we reviewed state-of-the-art data-driven computational drug repurposing approaches for OUD and discussed their advantages and potential challenges. We also highlighted promising repurposed candidate drugs for OUD that were identified by computational drug repurposing techniques and reviewed studies supporting their potential mechanisms of action in treating OUD.
Collapse
Affiliation(s)
- Zhenxiang Gao
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Pingjian Ding
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Rong Xu
- Center for Artificial Intelligence in Drug Discovery, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
3
|
Cippitelli A, Zribi G, Toll L. PPL-103: A mixed opioid partial agonist with desirable anti-cocaine properties. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110599. [PMID: 35798174 DOI: 10.1016/j.pnpbp.2022.110599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/19/2022]
Abstract
Cocaine use disorder (CUD) is a persistent public health problem for which no effective medications are available. PPL-103 is an opioid receptor ligand with partial agonist activity at mu, kappa and delta opioid receptors, with a greater efficacy for kappa and low efficacy at mu receptors. Because chronic cocaine use induces changes in the kappa opioid receptor/dynorphin system, we hypothesized that a kappa partial agonist, such as PPL-103, would attenuate the aversive properties of the upregulated kappa system, resulting in effective treatment approach for CUD. We tested the effects of PPL-103 on cocaine self-administration models that recapitulate core aspects of CUD in humans. We found that PPL-103 reduced both long and short access cocaine self-administration, motivation to respond for cocaine, and binge-like cocaine taking, in rats. Operant responding for food, fentanyl and locomotor behavior were not altered at doses that decreased cocaine infusions. Repeated PPL-103 treatment did not lead to tolerance development. PPL-103 also reduced both priming- and cue-induced reinstatement of cocaine seeking, being more effective in the former. Surprisingly, PPL-103 reduced self-administration parameters and reinstatement in rats previously treated with the long-acting kappa receptor antagonist JDTic more potently than in non-JDTic treated animals, whereas naltrexone injected to rats subsequent to JDTic administration increased self-administration, suggesting that the partial mu agonist activity, rather than kappa agonism is important for reduction in cocaine taking and seeking. However, partial kappa activation seems to increase safety by limiting dysphoria, tolerance and addiction development. PPL-103 displays a desirable profile as a possible CUD pharmacotherapy.
Collapse
Affiliation(s)
- Andrea Cippitelli
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States.
| | - Gilles Zribi
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| | - Lawrence Toll
- Biomedical Science Department, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, United States
| |
Collapse
|
4
|
El Hayek SA, Shatila MA, Adnan JA, Geagea LE, Kobeissy F, Talih FR. Is there a therapeutic potential in combining bupropion and naltrexone in schizophrenia? Expert Rev Neurother 2022; 22:737-749. [PMID: 36093756 DOI: 10.1080/14737175.2022.2124369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION A sustained-release tablet composed of a combination of the dopamine and norepinephrine reuptake inhibitor bupropion (BUP) and the µ-opioid receptor antagonist naltrexone (NAT) is marketed under the brand name Contrave by Orexigen Therapeutics for appetite control. Minimal literature is available regarding the use of combination bupropion and naltrexone (BUPNAT) in individuals with schizophrenia. AREAS COVERED In this review, we propose a theoretical model where BUPNAT may have a therapeutic effect in the treatment of schizophrenia. We explore the pathways targeted by the constituent drugs BUP and NAT and summarize the literature on their efficacy and possible adverse effects. We then look at the potential use of BUPNAT in schizophrenia. EXPERT OPINION Research has hinted that BUP's dopaminergic properties affect the same striatal pathways involved in schizophrenia. NAT, via opioid receptor antagonism, indirectly increases striatal dopamine release by disinhibiting nicotinic acetylcholine receptors. As such, we hypothesize that BUPNAT can have a therapeutic effect in schizophrenia, particularly on negative symptoms. We also suggest that it may ameliorate comorbidities frequently seen in this group of patients, including obesity, smoking, and substance use. Further research and clinical data are needed to elucidate the potential clinical benefits of BUPNAT in the treatment of schizophrenia.
Collapse
Affiliation(s)
- Samer A. El Hayek
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Malek A. Shatila
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jana A. Adnan
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Luna E. Geagea
- Department of Psychiatry, American University of Beirut, Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Farid R. Talih
- Department of Psychiatry, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
5
|
Sasaki-Tanaka R, Ray R, Moriyama M, Ray RB, Kanda T. Molecular Changes in Relation to Alcohol Consumption and Hepatocellular Carcinoma. Int J Mol Sci 2022; 23:ijms23179679. [PMID: 36077080 PMCID: PMC9456124 DOI: 10.3390/ijms23179679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 12/12/2022] Open
Abstract
Alcohol is the one of the major causes of liver diseases and promotes liver cirrhosis and hepatocellular carcinoma (HCC). In hepatocytes, alcohol is converted to acetaldehyde, which causes hepatic steatosis, cellular apoptosis, endoplasmic reticulum stress, peroxidation, production of cytokines and reduces immune surveillance. Endotoxin and lipopolysaccharide produced from intestinal bacteria also enhance the production of cytokines. The development of hepatic fibrosis and the occurrence of HCC are induced by these alcohol metabolites. Several host genetic factors have recently been identified in this process. Here, we reviewed the molecular mechanism associated with HCC in alcoholic liver disease.
Collapse
Affiliation(s)
- Reina Sasaki-Tanaka
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
- Correspondence: (R.S.-T.); (T.K.); Tel.: +81-3-3972-8111 (R.S.-T. & T.K.)
| | - Ranjit Ray
- Departments of Internal Medicine, and Molecular Microbiology and Immunology, Saint Louis University, Saint Louis, MO 63104, USA
| | - Mitsuhiko Moriyama
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
| | - Ratna B. Ray
- Department of Pathology, Saint Louis University, Saint Louis, MO 63104, USA
| | - Tatsuo Kanda
- Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
- Correspondence: (R.S.-T.); (T.K.); Tel.: +81-3-3972-8111 (R.S.-T. & T.K.)
| |
Collapse
|
6
|
The Potential of Methocinnamox as a Future Treatment for Opioid Use Disorder: A Narrative Review. PHARMACY 2022; 10:pharmacy10030048. [PMID: 35645327 PMCID: PMC9149874 DOI: 10.3390/pharmacy10030048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 02/01/2023] Open
Abstract
The opioid epidemic is an ongoing public health crisis, and the United States health system is overwhelmed with increasing numbers of opioid-related overdoses. Methocinnamox (MCAM) is a novel mu opioid receptor antagonist with an extended duration of action. MCAM has potential to reduce the burden of the opioid epidemic by being used as an overdose rescue treatment and a long-term treatment for opioid use disorder (OUD). The currently available treatments for OUD include naloxone, naltrexone, and methadone. These treatments have certain limitations, which include short duration of action, patient non-compliance, and diversion. A narrative review was conducted using PubMed and Google Scholar databases covering the history of the opioid epidemic, pain receptors, current OUD treatments and the novel drug MCAM. MCAM could potentially be used as both a rescue and long-term treatment for opioid misuse. This is due to its pseudo-irreversible antagonism of the mu opioid receptor, abnormally long duration of action of nearly two weeks, and the possibility of using kappa or delta opioid receptor agonists for pain management during OUD treatment. MCAM’s novel pharmacokinetic and pharmacodynamic properties open a new avenue for treating opioid misuse.
Collapse
|
7
|
Akins N, Mishra N, Harris H, Dudhipala N, Kim SJ, Keasling A, Majumdar S, Zjawiony J, Paris J, Ashpole N, Le H. 6,5‐Fused Ring, C2‐Salvinorin Ester, Dual Kappa and Mu Opioid Receptor Agonists as Analgesics Devoid of Anxiogenic Effects. ChemMedChem 2022; 17:e202100684. [PMID: 35043597 PMCID: PMC9015904 DOI: 10.1002/cmdc.202100684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 01/18/2022] [Indexed: 11/10/2022]
Abstract
Current common analgesics are mediated through the mu or kappa opioid receptor agonism. Unfortunately, selective mu or kappa receptor agonists often cause harmful side effects. However, ligands exhibiting dual agonism to the opioid receptors, such as to mu and kappa, or to mu and delta, have been suggested to temper undesirable adverse effects while retaining analgesic activity. Herein we report an introduction of various 6,5-fused rings to C2 of the salvinorin scaffold via an ester linker. In vitro studies showed that many of these compounds have dual agonism on kappa and mu opioid receptors. In vivo studies on the lead dual kappa and mu opioid receptor agonist demonstrated supraspinal thermal analgesic activity while avoiding anxiogenic effects in male mice, thus providing further strong evidence in support of the therapeutic advantages of dual opioid receptor agonists over selective opioid receptor agonists.
Collapse
Affiliation(s)
- Nicholas Akins
- University of Mississippi School of Pharmacy BioMolecular Sciences UNITED STATES
| | - Nisha Mishra
- University of Mississippi School of Pharmacy BioMolecular Sciences UNITED STATES
| | - Hannah Harris
- University of Mississippi School of Pharmacy BioMolecular Sciences UNITED STATES
| | - Narendar Dudhipala
- University of Mississippi School of Pharmacy Research Institutes of Pharmaceutical Sciences UNITED STATES
| | - Seong Jong Kim
- United States Department of Agriculture Natural Products Utilization Research Unit UNITED STATES
| | - Adam Keasling
- University of Mississippi School of Pharmacy BioMolecular Sciences UNITED STATES
| | - Soumyajit Majumdar
- University of Mississippi School of Pharmacy Pharmaceutics and Drug Delivery UNITED STATES
| | - Jordan Zjawiony
- University of Mississippi School of Pharmacy BioMolecular Sciences UNITED STATES
| | - Jason Paris
- University of Mississippi School of Pharmacy BioMolecular Sciences UNITED STATES
| | - Nicole Ashpole
- University of Mississippi School of Pharmacy BioMolecular Sciences UNITED STATES
| | - Hoang Le
- University of Mississippi Department of BioMolecular Sciences, School of Pharmacy 419 Faser Hall 38677 University UNITED STATES
| |
Collapse
|
8
|
Spetea M, Schmidhammer H. Recent Chemical and Pharmacological Developments on 14-Oxygenated- N-methylmorphinan-6-ones. Molecules 2021; 26:5677. [PMID: 34577147 PMCID: PMC8464912 DOI: 10.3390/molecules26185677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 09/09/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022] Open
Abstract
Adequate pain management, particularly chronic pain, remains a major challenge associated with modern-day medicine. Current pharmacotherapy offers unsatisfactory long-term solutions due to serious side effects related to the chronic administration of analgesic drugs. Morphine and structurally related derivatives (e.g., oxycodone, oxymorphone, buprenorphine) are highly effective opioid analgesics, mediating their effects via the activation of opioid receptors, with the mu-opioid receptor subtype as the primary molecular target. However, they also cause addiction and overdose deaths, which has led to a global opioid crisis in the last decades. Therefore, research efforts are needed to overcome the limitations of present pain therapies with the aim to improve treatment efficacy and to reduce complications. This review presents recent chemical and pharmacological advances on 14-oxygenated-N-methylmorphinan-6-ones, in the search of safer pain therapeutics. We focus on drug design strategies and structure-activity relationships on specific modifications in positions 5, 6, 14 and 17 on the morphinan skeleton, with the goal of aiding the discovery of opioid analgesics with more favorable pharmacological properties, potent analgesia and fewer undesirable effects. Targeted molecular modifications on the morphinan scaffold can afford novel opioids as bi- or multifunctional ligands targeting multiple opioid receptors, as attractive alternatives to mu-opioid receptor selective analgesics.
Collapse
Affiliation(s)
- Mariana Spetea
- Department of Pharmaceutical Chemistry, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria;
| | | |
Collapse
|
9
|
Butelman ER, Fry RS, Kimani R, Reed B, Kreek MJ. Neuroendocrine effects of naltrexone versus nalmefene in humans. Hum Psychopharmacol 2020; 35:e2726. [PMID: 32050055 PMCID: PMC11372649 DOI: 10.1002/hup.2726] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Naltrexone and nalmefene are approved for the treatment of alcohol use disorders, in different countries. Naltrexone is also approved for the treatment for opioid use disorders, most recently in a depot formulation. These compounds target primarily μ(mu)- and κ(kappa)-opioid receptor systems, which are involved in the downstream neurobiological effects of alcohol and in the modulation of neuroendocrine stress systems. The study objective was to compare the neuroendocrine effects of naltrexone and nalmefene on adrenocorticotropic hormone (ACTH), cortisol, and prolactin, in normal volunteers. METHOD Adult normal volunteers (n = 11 male and n = 9 female) were studied in a stress-minimized inpatient setting on three consecutive days, after intravenous saline, naltrexone HCl (10 mg), or nalmefene HCl (10 mg), in fixed order. ACTH, cortisol, and prolactin were analyzed pre-injection and up to 180 min post-injection. RESULTS Naltrexone and nalmefene caused elevations in ACTH and cortisol compared with saline. Nalmefene had a greater effect on ACTH and cortisol, compared with naltrexone. Both compounds also caused elevations in prolactin in males (females were not examined, due to the influence of menstrual cycle on prolactin). CONCLUSIONS This study suggests that both nalmefene and naltrexone have effects potentially due to κ-partial agonism in humans, as well as antagonist effects at μ-receptors.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Rebecca S Fry
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Rachel Kimani
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Brian Reed
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, New York
| |
Collapse
|
10
|
Montgomery D, Anand JP, Griggs NW, Fernandez TJ, Hartman JG, Sánchez-Santiago AA, Pogozheva ID, Traynor JR, Mosberg HI. Novel Dimethyltyrosine-Tetrahydroisoquinoline Peptidomimetics with Aromatic Tetrahydroisoquinoline Substitutions Show in Vitro Kappa and Mu Opioid Receptor Agonism. ACS Chem Neurosci 2019; 10:3682-3689. [PMID: 31199621 DOI: 10.1021/acschemneuro.9b00250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
The dimethyltyrosine-tetrahydroisoquinoline (Dmt-Tiq) scaffold was originally developed in the production of selective delta opioid receptor (DOR) antagonists. Installation of a 7-benzyl pendant on the tetrahydroisoquinoline core of this classic opioid scaffold introduced kappa opioid receptor (KOR) agonism. Further modification of this pendant resulted in retention of KOR agonism and the addition of mu opioid receptor (MOR) partial agonism, a bifunctional profile with potential to be used in the treatment of cocaine addiction.
Collapse
Affiliation(s)
- Deanna Montgomery
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jessica P. Anand
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
- Edward F. Domino Research Center, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Nicholas W. Griggs
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Thomas J. Fernandez
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Joshua G. Hartman
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ashley A. Sánchez-Santiago
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Irina D. Pogozheva
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - John R. Traynor
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
- Edward F. Domino Research Center, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Henry I. Mosberg
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
- Edward F. Domino Research Center, Medical School, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
11
|
Atigari DV, Uprety R, Pasternak GW, Majumdar S, Kivell BM. MP1104, a mixed kappa-delta opioid receptor agonist has anti-cocaine properties with reduced side-effects in rats. Neuropharmacology 2019; 150:217-228. [PMID: 30768946 DOI: 10.1016/j.neuropharm.2019.02.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/23/2019] [Accepted: 02/09/2019] [Indexed: 01/02/2023]
Abstract
Kappa opioid receptor (KOPr) agonists have preclinical anti-cocaine and antinociceptive effects. However, adverse effects including dysphoria, aversion, sedation, anxiety and depression limit their clinical development. MP1104, an analogue of 3-iodobenzoyl naltrexamine, is a potent dual agonist at KOPr and delta opioid receptor (DOPr), with full agonist efficacy at both these receptors. In this study, we evaluate the ability of MP1104 to modulate cocaine-induced behaviors and side-effects preclinically. In male Sprague-Dawley rats trained to self-administer cocaine, MP1104 (0.3 and 1 mg/kg) reduced cocaine-primed reinstatement of drug-seeking behavior and caused significant downward shift of the dose-response curve in cocaine self-administration tests (0.3 and 0.6 mg/kg). The anti-cocaine effects exerted by MP1104 are in part due to increased dopamine (DA) uptake by the dopamine transporter (DAT) in the dorsal striatum (dStr) and nucleus accumbens (NAc). MP1104 (0.3 and 0.6 mg/kg) showed no significant anxiogenic effects in the elevated plus maze, pro-depressive effects in the forced swim test, or conditioned place aversion. Furthermore, pre-treatment with a DOPr antagonist, led to MP1104 producing aversive effects. This data suggests that the DOPr agonist actions of MP1104 attenuate the KOPr-mediated aversive effects of MP1104. The overall results from this study show that MP1104, modulates DA uptake in the dStr and NAc, and exerts potent anti-cocaine properties in self-administration tests with reduced side-effects compared to pure KOPr agonists. This data supports the therapeutic development of dual KOPr/DOPr agonists to reduce the side-effects of selective KOPr agonists. This article is part of the Special Issue entitled 'Opioid Neuropharmacology: Advances in treating pain and opioid addiction'.
Collapse
Affiliation(s)
- Diana V Atigari
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Rajendra Uprety
- Molecular Pharmacology Program and Department of Neurology, Memorial Sloan Kettering Cancer Centre, New York, USA
| | - Gavril W Pasternak
- Molecular Pharmacology Program and Department of Neurology, Memorial Sloan Kettering Cancer Centre, New York, USA
| | - Susruta Majumdar
- Molecular Pharmacology Program and Department of Neurology, Memorial Sloan Kettering Cancer Centre, New York, USA; Center for Clinical Pharmacology, St Louis College of Pharmacy and Washington University School of Medicine, St Louis, MO, USA
| | - Bronwyn M Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand.
| |
Collapse
|
12
|
Fujii K, Koshidaka Y, Adachi M, Takao K. Effects of chronic fentanyl administration on behavioral characteristics of mice. Neuropsychopharmacol Rep 2018; 39:17-35. [PMID: 30506634 PMCID: PMC7292323 DOI: 10.1002/npr2.12040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/10/2018] [Accepted: 10/19/2018] [Indexed: 01/16/2023] Open
Abstract
Background Fentanyl, a synthetic opioid categorized as a narcotic analgesic, has a 100‐ to 200‐fold stronger effect than most opioids, such as morphine. Fatal accidents due to chronic use and abuse of fentanyl are a worldwide social problem. One reason for the abuse of fentanyl is its psychostimulant effects that could induce behavioral changes. The effects of chronic fentanyl administration on behavior, however, are unclear. Methods Adult male C57BL/6J mice were chronically administered fentanyl (0.03 or 0.3 mg/kg/d i.p.), and various behaviors were assessed using a behavioral test battery. Results Mice chronically administered a high dose of fentanyl (0.3 mg/kg/d) exhibited decreased anxiety‐like behavior as assessed by the open field and elevated plus maze tests. On the other hand, interruption of fentanyl administration led to increased anxiety‐like behavior as observed in the light and dark transition test. The hot plate test revealed that chronic administration of fentanyl reduced pain sensitivity. High‐dose chronic fentanyl administration reduced the locomotor stimulatory effects of cocaine. The results, however, failed to reach the threshold for study‐wide statistical significance. Conclusion Chronic fentanyl administration induces some behavioral changes in mice. Although further studies are needed to clarify the underlying mechanisms of the behavioral effects of chronic fentanyl administration, our findings suggest that fentanyl is safe under properly controlled conditions. To investigate the effects of long‐term fentanyl use on brain function, adult male C57BL/6J mice were chronically administered fentanyl (0.03 or 0.3 mg/kg/d ip) and analyzed in a behavioral test battery. Chronic fentanyl administration reduced anxiety‐like behavior, pain sensitivity, and the locomotor stimulatory effects of cocaine in mice. The results, however, failed to reach the threshold for study‐wide statistical significance.![]()
Collapse
Affiliation(s)
- Kazuki Fujii
- Department of Behavioral Physiology, Graduate School of Innovative Life Science, University of Toyama, Toyama, Japan.,Life Science Research Center, University of Toyama, Toyama, Japan
| | - Yumie Koshidaka
- Life Science Research Center, University of Toyama, Toyama, Japan
| | - Mayumi Adachi
- Life Science Research Center, University of Toyama, Toyama, Japan
| | - Keizo Takao
- Department of Behavioral Physiology, Graduate School of Innovative Life Science, University of Toyama, Toyama, Japan.,Life Science Research Center, University of Toyama, Toyama, Japan
| |
Collapse
|
13
|
Kallupi M, Shen Q, de Guglielmo G, Yasuda D, Journigan VB, Zaveri NT, Ciccocioppo R. Buprenorphine requires concomitant activation of NOP and MOP receptors to reduce cocaine consumption. Addict Biol 2018. [PMID: 28635181 DOI: 10.1111/adb.12513] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Buprenorphine's clinical use is approved for the treatment of heroin addiction; however, evidence supporting its efficacy in cocaine abuse also exists. While for heroin it has been demonstrated that the effect of buprenorphine is mediated by its ability to activate μ-opioid peptide receptor (MOP) receptors, the mechanism through which it attenuates cocaine intake remains elusive. We explored this mechanism using operant models where rodents were trained to chronically self-administer cocaine for 2 hours daily. Buprenorphine (0.3, 1.0 and 3.0 mg/kg) given intraperitoneally 90 minutes before access to cocaine significantly and dose dependently reduced its intake. Pre-treatment with naltrexone or with the selective nociceptin/orphanin FQ peptide (NOP) antagonist SB-612111 did not prevent buprenorphine-induced reduction of cocaine intake. However, when naltrexone and SB-612111 were combined, the effect of buprenorphine on cocaine was completely prevented. To confirm that co-activation of MOP and NOP receptors is the underlying mechanism through which buprenorphine reduces cocaine intake, three compounds, namely, AT-034, AT-201 and AT-202, with a range of affinity and intrinsic activity profiles for MOP and NOP receptors, but weak ability for kappa-opioid peptide receptor (KOP) transmission, were tested. Consistent with our hypothesis based on buprenorphine's effects, results demonstrated that AT-034 and AT-201, which co-activate MOP and NOP receptors, reduced cocaine self-administration like buprenorphine. AT-202, which selectively stimulates NOP receptors, was not effective. Together, these data demonstrate that for buprenorphine, co-activation of MOP and NOP receptors is essential to reduce cocaine consumption. These results open new vistas on the treatment of cocaine addiction by developing compounds with mixed MOP/NOP agonist properties.
Collapse
Affiliation(s)
- Marsida Kallupi
- School of Pharmacy, Pharmacology Unit; University of Camerino; Italy
- Committee on the Neurobiology of Addictive Disorders; The Scripps Research Institute; La Jolla CA USA
| | - Qianwei Shen
- School of Pharmacy, Pharmacology Unit; University of Camerino; Italy
| | - Giordano de Guglielmo
- School of Pharmacy, Pharmacology Unit; University of Camerino; Italy
- Committee on the Neurobiology of Addictive Disorders; The Scripps Research Institute; La Jolla CA USA
| | | | | | | | | |
Collapse
|
14
|
Abstract
INTRODUCTION Substance use disorders are a group of chronic relapsing disorders of the brain, which have massive public health and societal impact. In some disorders (e.g., heroin/prescription opioid addictions) approved medications have a major long-term benefit. For other substances (e.g., cocaine, amphetamines and cannabis) there are no approved medications, and for alcohol there are approved treatments, which are not in wide usage. Approved treatments for tobacco use disorders are available, and novel medications are also under study. Areas covered: Medication-based approaches which are in advanced preclinical stages, or which have reached proof-of concept clinical laboratory studies, as well as clinical trials. Expert opinion: Current challenges involve optimizing translation between preclinical and clinical development, and between clinical laboratory studies to therapeutic clinical trials. Comorbidities including depression or anxiety are challenges for study design and analysis. Improved pharmacogenomics, biomarker and phenotyping approaches are areas of interest. Pharmacological mechanisms currently under investigation include modulation of glutamatergic, GABA, vasopressin and κ-receptor function, as well as inhibition of monoamine re-uptake. Other factors that affect potential market size for emerging medications include stigma, availability of treatment settings, adoption by clinicians, and the prevalence of persons with SUD who are not actively treatment-seeking.
Collapse
Affiliation(s)
- Eduardo R Butelman
- a Laboratory in the Biology of Addictive Diseases , The Rockefeller University , New York , NY , USA
| | - Mary Jeanne Kreek
- a Laboratory in the Biology of Addictive Diseases , The Rockefeller University , New York , NY , USA
| |
Collapse
|
15
|
Molecular switches of the κ opioid receptor triggered by 6'-GNTI and 5'-GNTI. Sci Rep 2016; 6:18913. [PMID: 26742690 PMCID: PMC4705513 DOI: 10.1038/srep18913] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 11/30/2015] [Indexed: 12/29/2022] Open
Abstract
The κ opioid receptor (κOR) is a member of G-protein-coupled receptors, and is considered as a promising drug target for treating neurological diseases. κOR selective 6'-GNTI was proved to be a G-protein biased agonist, whereas 5'-GNTI acts as an antagonist. To investigate the molecular mechanism of how these two ligands induce different behaviors of the receptor, we built two systems containing the 5'-GNTI-κOR complex and the 6'-GNTI-κOR complex, respectively, and performed molecular dynamics simulations of the two systems. We observe that transmembrane (TM) helix 6 of the κOR rotates about 4.6(o) on average in the κOR-6'-GNTI complex. Detailed analyses of the simulation results indicate that E297(6.58) and I294(6.55) play crucial roles in the rotation of TM6. In the simulation of the κOR-5'-GNTI system, it is revealed that 5'-GNTI can stabilize TM6 in the inactive state form. In addition, the kink of TM7 is stabilized by a hydrogen bond between S324(7.47) and the residue V69(1.42) on TM1.
Collapse
|
16
|
Váradi A, Marrone GF, Eans SO, Ganno ML, Subrath JJ, Le Rouzic V, Hunkele A, Pasternak GW, McLaughlin JP, Majumdar S. Synthesis and characterization of a dual kappa-delta opioid receptor agonist analgesic blocking cocaine reward behavior. ACS Chem Neurosci 2015; 6:1813-24. [PMID: 26325040 DOI: 10.1021/acschemneuro.5b00153] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
3-Iodobenzoyl naltrexamine (IBNtxA) is a potent analgesic belonging to the pharmacologically diverse 6β-amidoepoxymorphinan group of opioids. We present the synthesis and pharmacological evaluation of five analogs of IBNtxA. The scaffold of IBNtxA was modified by removing the 14-hydroxy group, incorporating a 7,8 double bond and various N-17 alkyl substituents. The structural modifications resulted in analogs with picomolar affinities for opioid receptors. The lead compound (MP1104) was found to exhibit approximately 15-fold greater antinociceptive potency (ED50 = 0.33 mg/kg) compared with morphine, mediated through the activation of kappa- and delta-opioid receptors. Despite its kappa agonism, this lead derivative did not cause place aversion or preference in mice in a place-conditioning assay, even at doses 3 times the analgesic ED50. However, pretreatment with the lead compound prevented the reward behavior associated with cocaine in a conditioned place preference assay. Together, these results suggest the promise of dual acting kappa- and delta-opioid receptor agonists as analgesics and treatments for cocaine addiction.
Collapse
Affiliation(s)
- András Váradi
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Gina F. Marrone
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Shainnel O. Eans
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Michelle L. Ganno
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Joan J. Subrath
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Valerie Le Rouzic
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Amanda Hunkele
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Gavril W. Pasternak
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Jay P. McLaughlin
- Torrey Pines Institute for Molecular Studies, 11350 SW Village Parkway, Port St. Lucie, Florida 34987, United States
| | - Susruta Majumdar
- Molecular
Pharmacology and Chemistry Program, Department of Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| |
Collapse
|
17
|
Crowley NA, Kash TL. Kappa opioid receptor signaling in the brain: Circuitry and implications for treatment. Prog Neuropsychopharmacol Biol Psychiatry 2015; 62:51-60. [PMID: 25592680 PMCID: PMC4465498 DOI: 10.1016/j.pnpbp.2015.01.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/20/2014] [Accepted: 01/04/2015] [Indexed: 12/15/2022]
Abstract
Kappa opioid receptors (KORs) in the central nervous system have been known to be important regulators of a variety of psychiatry illnesses, including anxiety and addiction, but their precise involvement in these disorders is complex and has yet to be fully elucidated. Here, we briefly review the pharmacology of KORs in the brain, including KOR's involvement in anxiety, depression, and drug addiction. We also review the known neuronal circuitry impacted by KOR signaling, and interactions with corticotrophin-releasing factor (CRF), another key peptide in anxiety-related illnesses, as well as the role of glucocorticoids. We suggest that KORs are a promising therapeutic target for a host of neuropsychiatric conditions.
Collapse
Affiliation(s)
- Nicole A. Crowley
- Neurobiology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas L. Kash
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
18
|
Butelman ER, Kreek MJ. Salvinorin A, a kappa-opioid receptor agonist hallucinogen: pharmacology and potential template for novel pharmacotherapeutic agents in neuropsychiatric disorders. Front Pharmacol 2015; 6:190. [PMID: 26441647 PMCID: PMC4561799 DOI: 10.3389/fphar.2015.00190] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/24/2015] [Indexed: 12/19/2022] Open
Abstract
Salvinorin A is a potent hallucinogen, isolated from the ethnomedical plant Salvia divinorum. Salvinorin A is a selective high efficacy kappa-opioid receptor (KOPr) agonist, and thus implicates the KOPr system and its endogenous agonist ligands (the dynorphins) in higher functions, including cognition and perceptual effects. Salvinorin A is the only selective KOPr ligand to be widely available outside research or medical settings, and salvinorin A-containing products have undergone frequent non-medical use. KOPr/dynorphin systems in the brain are known to be powerful counter-modulatory mechanisms to dopaminergic function, which is important in mood and reward engendered by natural and chemical reinforcers (including drugs of abuse). KOPr activation (including by salvinorin A) can thus cause aversion and anhedonia in preclinical models. Salvinorin A is also a completely new scaffold for medicinal chemistry approaches, since it is a non-nitrogenous neoclerodane, unlike other known opioid ligands. Ongoing efforts have the goal of discovering novel semi-synthetic salvinorin analogs with potential KOPr-mediated pharmacotherapeutic effects (including partial agonist or biased agonist effects), with a reduced burden of undesirable effects associated with salvinorin A.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University , New York, NY, USA
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University , New York, NY, USA
| |
Collapse
|
19
|
Bender AM, Griggs NW, Anand JP, Traynor JR, Jutkiewicz EM, Mosberg HI. Asymmetric synthesis and in vitro and in vivo activity of tetrahydroquinolines featuring a diverse set of polar substitutions at the 6 position as mixed-efficacy μ opioid receptor/δ opioid receptor ligands. ACS Chem Neurosci 2015; 6:1428-35. [PMID: 25938166 DOI: 10.1021/acschemneuro.5b00100] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
We previously reported a small series of mixed-efficacy μ opioid receptor (MOR) agonist/δ opioid receptor (DOR) antagonist peptidomimetics featuring a tetrahydroquinoline scaffold and showed the promise of this series as effective analgesics after intraperitoneal administration in mice. We report here an expanded structure-activity relationship study of the pendant region of these compounds and focus in particular on the incorporation of heteroatoms into this side chain. These analogues provide new insight into the binding requirements for this scaffold at MOR, DOR, and the κ opioid receptor (KOR), and several of them (10j, 10k, 10m, and 10n) significantly improve upon the overall MOR agonist/DOR antagonist profile of our previous compounds. In vivo data for 10j, 10k, 10m, and 10n are also reported and show the antinociceptive potency and duration of action of compounds 10j and 10m to be comparable to those of morphine.
Collapse
Affiliation(s)
- Aaron M. Bender
- Interdepartmental Program in Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Nicholas W. Griggs
- Department of Pharmacology, School of Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jessica P. Anand
- Department of Pharmacology, School of Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - John R. Traynor
- Department of Pharmacology, School of Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Emily M. Jutkiewicz
- Department of Pharmacology, School of Medicine, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Henry I. Mosberg
- Interdepartmental Program in Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|