1
|
Hashemizadeh S, Alaee E, Aghajani N, Azizi H, Semnanian S. Atorvastatin facilitates extinction and prevents reinstatement of morphine-induced conditioned place preference in rats. Biomed Pharmacother 2024; 181:117639. [PMID: 39520913 DOI: 10.1016/j.biopha.2024.117639] [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: 08/10/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Opioid addiction is known as a chronic relapsing disorder associated with long-lasting molecular and cellular neuroadaptations that lead to compulsive behavior. Current pharmacotherapies target the modulation of mu-opioid receptors (MOR); however, the relapse rate remains high. In this study, we evaluated the potential effect of atorvastatin, a blood-brain barrier-permeable statin, on preventing morphine relapse through both extinction-reinstatement and abstinence-reinstatement models using conditioned place preference (CPP). Adult male Wistar rats were used to establish morphine-induced CPP (5 mg/kg), followed by extinction training and subsequent priming injection of morphine (2 mg/kg, i.p.) to induce relapse-like behavior. Extinguished rats significantly reinstated their morphine-seeking behavior. In contrast, rats that received different doses of atorvastatin (0.1, 0.5, 1 mg/kg) 1 hour before each extinction training session did not show a preference for the morphine-paired chamber. Moreover, acute atorvastatin injection (1 mg/kg, i.p.) 1 h before the reinstatement test significantly prevented reinstated morphine-seeking behavior. We found that atorvastatin 1 mg/kg attenuated morphine-seeking behaviors, and this attenuation of reinstatement was partly mediated by the upregulation of brain-derived neurotrophic factor (BDNF) in the prefrontal cortex (PFC) and hippocampus (Hipp). Furthermore, atorvastatin reversed Oprm1 upregulation (mu-opioid receptor gene) induced by relapse in the nucleus accumbens and Hipp. Moreover, treatment with atorvastatin during the extinction period alters the electrophysiological properties of the mPFC neurons following morphine priming and enhances neuronal excitability. We conclude that atorvastatin was effective in decreasing reinstatement.
Collapse
Affiliation(s)
- Shiva Hashemizadeh
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Elham Alaee
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Niloofar Aghajani
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Azizi
- Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran; Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Saeed Semnanian
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences, IPM, Tehran, Iran; Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
2
|
Yu J, Kumar A, Zhang X, Martin C, Van Holsbeeck K, Raia P, Koehl A, Laeremans T, Steyaert J, Manglik A, Ballet S, Boland A, Stoeber M. Structural basis of μ-opioid receptor targeting by a nanobody antagonist. Nat Commun 2024; 15:8687. [PMID: 39384768 PMCID: PMC11464722 DOI: 10.1038/s41467-024-52947-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 09/24/2024] [Indexed: 10/11/2024] Open
Abstract
The μ-opioid receptor (μOR), a prototypical G protein-coupled receptor (GPCR), is the target of opioid analgesics such as morphine and fentanyl. Due to the severe side effects of current opioid drugs, there is considerable interest in developing novel modulators of μOR function. Most GPCR ligands today are small molecules, however biologics, including antibodies and nanobodies, represent alternative therapeutics with clear advantages such as affinity and target selectivity. Here, we describe the nanobody NbE, which selectively binds to the μOR and acts as an antagonist. We functionally characterize NbE as an extracellular and genetically encoded μOR ligand and uncover the molecular basis for μOR antagonism by determining the cryo-EM structure of the NbE-μOR complex. NbE displays a unique ligand binding mode and achieves μOR selectivity by interactions with the orthosteric pocket and extracellular receptor loops. Based on a β-hairpin loop formed by NbE that deeply protrudes into the μOR, we design linear and cyclic peptide analogs that recapitulate NbE's antagonism. The work illustrates the potential of nanobodies to uniquely engage with GPCRs and describes lower molecular weight μOR ligands that can serve as a basis for therapeutic developments.
Collapse
MESH Headings
- Receptors, Opioid, mu/metabolism
- Receptors, Opioid, mu/chemistry
- Receptors, Opioid, mu/antagonists & inhibitors
- Single-Domain Antibodies/chemistry
- Single-Domain Antibodies/metabolism
- Single-Domain Antibodies/pharmacology
- Humans
- Cryoelectron Microscopy
- Ligands
- HEK293 Cells
- Animals
- Protein Binding
- Binding Sites
- Models, Molecular
- Analgesics, Opioid/pharmacology
- Analgesics, Opioid/chemistry
- Analgesics, Opioid/metabolism
- Peptides, Cyclic/chemistry
- Peptides, Cyclic/metabolism
- Peptides, Cyclic/pharmacology
Collapse
Affiliation(s)
- Jun Yu
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Amit Kumar
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Xuefeng Zhang
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Charlotte Martin
- Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kevin Van Holsbeeck
- Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Pierre Raia
- Department of Plant Sciences, University of Geneva, Geneva, Switzerland
| | - Antoine Koehl
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, USA
| | | | - Jan Steyaert
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
- VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
| | - Aashish Manglik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
| | - Steven Ballet
- Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Andreas Boland
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland.
| | - Miriam Stoeber
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
3
|
Maguire DR. Evaluation of potential punishing effects of 2,5-dimethoxy-4-methylamphetamine (DOM) in rhesus monkeys responding under a choice procedure. Behav Pharmacol 2024; 35:378-385. [PMID: 39052019 PMCID: PMC11398979 DOI: 10.1097/fbp.0000000000000787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
OBJECTIVES There has been substantial and growing interest in the therapeutic utility of drugs acting at serotonin 2A subtype (5-HT 2A ) receptors, increasing the need for characterization of potential beneficial and adverse effects of such compounds. Although numerous studies have evaluated the possible rewarding and reinforcing effects of 5-HT 2A receptor agonists, there have been relatively few studies on potential aversive effects. METHODS The current study investigated punishing effects of 2,5-dimethoxy-4-methylamphetamine (DOM) in four rhesus monkeys responding under a choice procedure in which responding on one lever delivered a sucrose pellet alone and responding on the other lever delivered a sucrose pellet plus an intravenous infusion of a range of doses of fentanyl (0.1-3.2 µg/kg/infusion), histamine (3.2-100 µg/kg/infusion), or DOM (3.2-100 µg/kg/infusion). RESULTS When fentanyl was available, responding for a pellet plus an infusion increased dose dependently in all subjects, indicating a positive reinforcing effect of fentanyl. When histamine was available, responding for a pellet plus an infusion decreased in three of four subjects, indicating a punishing effect of histamine. Whether available before or after histamine, DOM did not systematically alter choice across the range of doses tested. CONCLUSION These results suggest that the 5-HT 2A receptor agonist DOM has neither positive reinforcing nor punishing effects under a choice procedure that is sensitive to both processes.
Collapse
Affiliation(s)
- David R Maguire
- Department of Pharmacology and Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
4
|
Ghodrati S, Carey LM, France CP. Antinociceptive effects of fentanyl and nonopioid drugs in methocinnamox-treated rats. Drug Alcohol Depend 2024; 260:111320. [PMID: 38723374 PMCID: PMC11619060 DOI: 10.1016/j.drugalcdep.2024.111320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 06/16/2024]
Abstract
BACKGROUND A single administration of the opioid receptor antagonist methocinnamox (MCAM) antagonizes the antinociceptive effects of µ-opioid receptor agonists for 2 weeks or longer. Such a long duration of antagonism could necessitate the use of nonopioid drugs for treating pain in patients receiving MCAM for opioid use disorder (OUD). METHODS The antinociceptive effects of fentanyl and nonopioid drugs were assessed in 24 male Sprague Dawley rats using a complete Freund's adjuvant (CFA) model of inflammatory pain. Twelve rats received 10mg/kg MCAM and 12 received vehicle; half (n=6) of the animals from each treatment group were treated (intraplantar) with CFA or saline. Hypersensitivity to mechanical stimulation was measured using a von Frey anesthesiometer. Fentanyl (0.01-0.1mg/kg), ketamine (17.8-56mg/kg), gabapentin (32-100mg/kg), meloxicam (3.2-10mg/kg), and ∆9-tetrahydrocannabinol (THC, 1-10mg/kg) were administered intraperitoneally and tested every 3 days in a pseudorandom order. Next, the same drugs were studied for effects on motor performance using a rotarod apparatus. RESULTS CFA-induced hypersensitivity was attenuated by fentanyl in vehicle- but not MCAM-treated rats. THC, ketamine, and gabapentin attenuated (up to 82, 66, and 46 %, respectively) CFA-evoked mechanical hypersensitivity in both MCAM- and vehicle-treated rats. Meloxicam failed to alter CFA-evoked mechanical hypersensitivity in either group. Fentanyl, THC, gabapentin, and meloxicam did not affect motor performance in either group whereas ketamine impaired motor performance in both groups (up to 71 % reduction in latency to fall). CONCLUSIONS These data suggest that ketamine, gabapentin, and THC could be effective for treating inflammatory pain under conditions of long term µ-opioid receptor antagonism.
Collapse
Affiliation(s)
- Saba Ghodrati
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX 78229, USA; Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Lawrence M Carey
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX 78229, USA; Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Charles P France
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX 78229, USA; Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center, San Antonio, TX 78229, USA; Department of Psychiatry, University of Texas Health Science Center, San Antonio, TX 78229, USA.
| |
Collapse
|
5
|
Maguire DR, France CP. Daily methocinnamox treatment dose-dependently attenuates fentanyl self-administration in rhesus monkeys. Neuropharmacology 2024; 243:109777. [PMID: 37944894 DOI: 10.1016/j.neuropharm.2023.109777] [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: 08/11/2023] [Revised: 09/26/2023] [Accepted: 10/22/2023] [Indexed: 11/12/2023]
Abstract
Opioid use disorder and opioid overdose continue to be significant public health challenges despite the availability of effective treatments. Methocinnamox (MCAM) is a novel, long-acting opioid receptor antagonist that might be an effective treatment for opioid use disorder (i.e., preventing relapse and overdose). In nonhuman primates, MCAM selectively blocks the positive reinforcing effects of mu opioid receptor agonists, including heroin, fentanyl, and its ultra-potent analogs (e.g., carfentanil) with a single administration of MCAM being effective for up to two weeks. Because treatment of opioid use disorder would involve repeated administration of a medication, MCAM was studied in rhesus monkeys (3 males and 2 females) responding under a fixed-ratio self-administration procedure for a range of doses of fentanyl (0.000032-0.1 mg/kg/infusion). The fentanyl self-administration dose-effect curve was determined before and during treatment with progressively increasing daily doses of MCAM (0.001-0.1 mg/kg) given subcutaneously 1 h before the session. MCAM dose-dependently shifted the fentanyl dose-effect curve rightward and then, at larger doses, downward. The largest treatment dose of MCAM (0.1 mg/kg/day) shifted the curve more than 120-fold rightward with monkeys receiving doses much larger than the likely lethal dose of fentanyl with no adverse effect or observable change in behavior. This study demonstrates that MCAM reliably and dose-dependently decreases fentanyl self-administration and prevents opioid overdose, with no evidence of adverse effects over a broad dose range, further supporting the potential therapeutic utility of this novel antagonist.
Collapse
Affiliation(s)
- David R Maguire
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA; Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
| | - Charles P France
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA; Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA; Department of Psychiatry, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA.
| |
Collapse
|
6
|
Tabanelli R, Brogi S, Calderone V. Targeting Opioid Receptors in Addiction and Drug Withdrawal: Where Are We Going? Int J Mol Sci 2023; 24:10888. [PMID: 37446064 PMCID: PMC10341731 DOI: 10.3390/ijms241310888] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/14/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
This review article offers an outlook on the use of opioids as therapeutics for treating several diseases, including cancer and non-cancer pain, and focuses the analysis on the opportunity to target opioid receptors for treating opioid use disorder (OUD), drug withdrawal, and addiction. Unfortunately, as has been well established, the use of opioids presents a plethora of side effects, such as tolerance and physical and physiological dependence. Accordingly, considering the great pharmacological potential in targeting opioid receptors, the identification of opioid receptor ligands devoid of most of the adverse effects exhibited by current therapeutic agents is highly necessary. To this end, herein, we analyze some interesting molecules that could potentially be useful for treating OUD, with an in-depth analysis regarding in vivo studies and clinical trials.
Collapse
Affiliation(s)
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (R.T.); (V.C.)
| | | |
Collapse
|
7
|
Gerak LR, France CP. Attenuation of the Positive-Reinforcing Effects of Ultra-Potent Fentanyl Analogs, Along with Those of Fentanyl and Heroin, During Daily Treatment with Methocinnamox in Rhesus Monkeys. J Pharmacol Exp Ther 2023; 384:363-371. [PMID: 36575032 PMCID: PMC9976789 DOI: 10.1124/jpet.122.001267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 11/22/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
Without substantial intervention, the opioid crisis is projected to continue, underscoring the need to develop new treatments for opioid use disorder (OUD). One drug under development is the µ opioid receptor antagonist methocinnamox (MCAM), which appears to offer advantages over currently available medications; however, some questions remain about its potential utility, including its ability to block the effects of ultra-potent fentanyl analogs. The goal of this study was to examine its effectiveness in attenuating the abuse-related effects of the fentanyl analogs carfentanil and 3-methylfentanyl in monkeys responding for food or intravenous infusions under a choice procedure. These drugs were compared with fentanyl, heroin, methamphetamine, and cocaine. Food was preferred over saline, and there was a dose-dependent increase in responding for drug over food with no marked decrease in response rates or number of choice trials completed for any of the six drugs studied. Naltrexone (0.032 mg/kg) antagonized choice of µ opioid receptor agonists, producing rightward shifts in dose-effect curves ranging from 27-fold (carfentanil) to 71-fold (heroin). In contrast, naltrexone was less effective in attenuating choice of methamphetamine or cocaine with curves obtained in the presence of naltrexone shifted <3-fold. Daily treatment with 0.032 mg/kg MCAM also antagonized the effects of opioids, shifting curves 20-fold (fentanyl) to 72-fold (heroin) rightward; MCAM did not significantly change dose-effect curves for methamphetamine or cocaine. Thus, antagonism by MCAM is similar across a variety of µ opioid receptor agonists, including ultra-potent fentanyl analogs, further supporting its potential utility as a treatment for OUD. SIGNIFICANCE STATEMENT: Treatments for opioid use disorder (OUD) should attenuate the effects of a variety of opioids, including emerging threats like the ultra-potent fentanyl analogs. The novel µ opioid receptor antagonist MCAM is being developed to treat OUD because it provides long-lasting blockade of the reinforcing effects of heroin and fentanyl. The current study shows that MCAM attenuates the abuse-related effects of the fentanyl analogs carfentanil and 3-methylfentanyl, further supporting the utility of MCAM as a treatment for OUD.
Collapse
Affiliation(s)
- Lisa R Gerak
- Department of Pharmacology (L.R.G., C.P.F.) and Department of Psychiatry (C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas; and Addiction Research, Treatment, & Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, Texas (L.R.G., C.P.F.)
| | - Charles P France
- Department of Pharmacology (L.R.G., C.P.F.) and Department of Psychiatry (C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas; and Addiction Research, Treatment, & Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, Texas (L.R.G., C.P.F.)
| |
Collapse
|
8
|
Maguire DR, France CP. Behavioral pharmacology of methocinnamox: A potential new treatment for opioid overdose and opioid use disorder. J Exp Anal Behav 2023; 119:392-406. [PMID: 36759567 PMCID: PMC10281830 DOI: 10.1002/jeab.831] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/28/2022] [Indexed: 02/11/2023]
Abstract
Opioid overdose and opioid use disorder continue to be significant public health challenges despite the availability of effective medications and significant efforts at all levels of society. The emergence of highly potent and efficacious opioids such as fentanyl and its derivatives over the last decade has only exacerbated what was already a substantial problem. Behavioral pharmacology research has proven invaluable for understanding the effects of drugs as well as developing and evaluating pharmacotherapies for disorders involving the central nervous system, including substance abuse disorders. This paper describes a program of research characterizing a potent, selective, and long-lasting mu opioid receptor antagonist, methocinnamox, and evaluating its potential for treating opioid overdose and opioid use disorder. Studies in rodents and nonhuman primates demonstrate that methocinnamox prevents and reverses opioid-induced ventilatory depression and selectively blocks opioid self-administration. This work, taken together with rigorous in vitro and ex vivo studies investigating methocinnamox neuropharmacology, lays a solid foundation for the therapeutic utility of this potentially life-saving medication. Moreover, these studies demonstrate how rigorous behavioral pharmacological studies can be integrated in a broader drug discovery and development research program.
Collapse
Affiliation(s)
- David R. Maguire
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, TX, United States
- Addiction Research, Treatment, and Training, Center of Excellence, University of Texas, Health Science Center at San Antonio, TX, United States
| | - Charles P. France
- Department of Pharmacology, University of Texas Health Science Center at San Antonio, TX, United States
- Addiction Research, Treatment, and Training, Center of Excellence, University of Texas, Health Science Center at San Antonio, TX, United States
- Department of Psychiatry, University of Texas, Health Science Center at San Antonio, TX, United States
| |
Collapse
|
9
|
Maguire DR. Effects of 2,5-Dimethoxy-4-Methylamphetamine (DOM) and 2-Piperazin-1-yl-Quinoline (Quipazine) on Fentanyl Versus Food Choice in Rhesus Monkeys. J Pharmacol Exp Ther 2023; 384:155-162. [PMID: 36272734 PMCID: PMC9827501 DOI: 10.1124/jpet.122.001318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/18/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
Abstract
There has been increasing interest in the potential therapeutic effects of drugs with agonist properties at serotonin 2A subtype (5-HT2A) receptors (e.g., psychedelics), including treatment of substance use disorders. Studying interactions between 5-HT2A receptor agonists and other drugs is important for understanding potential therapeutic effects as well as adverse interactions. Direct-acting 5-HT2A receptor agonists such as 2,5-dimethoxy-4-methylamphetamine (DOM) and 2-piperazin-1-yl-quinoline (quipazine) enhance some (e.g., antinociceptive) effects of opioids; however, it is unclear whether they alter the abuse-related effects of opioids. This study examined whether DOM and quipazine alter the reinforcing effects of fentanyl in rhesus monkeys (n = 6) responding under a food versus drug choice procedure. Responding on one lever delivered sucrose pellets and responding on the other lever delivered intravenous (i.v.) infusions. In one set of experiments, fentanyl (0.1-3.2 µg/kg/infusion) versus food choice sessions were preceded by noncontingent i.v. pretreatments with DOM (0032-0.32 mg/kg), quipazine (0.32-1.0 mg/kg), naltrexone (0.032 mg/kg), or heroin (0.1 mg/kg). In another set of experiments, fentanyl was available during choice sessions in combination with DOM (0.32-100 µg/kg/infusion) or quipazine (3.2-320 µg/kg/infusion) in varying dose ratios. Naltrexone decreased and heroin increased fentanyl choice, demonstrating sensitivity of responding to pharmacological manipulation. However, whether given as a pretreatment or made available in combination with fentanyl as a mixture, neither DOM nor quipazine significantly altered fentanyl choice. These results suggest that 5-HT2A receptor agonists do not enhance the reinforcing effects of opioids and, thus, will not likely enhance abuse potential. SIGNIFICANCE STATEMENT: Serotonin 2A subtype receptor agonists enhance some (e.g., antinociceptive) effects of opioids, suggesting they could be combined with opioids in some therapeutic contexts such as treating pain. However, it is unclear whether they also enhance adverse effects of opioids, including abuse. Results of this study indicate that serotonin 2A subtype receptor agonists do not reliably enhance opioid self-administration and, thus, are unlikely to enhance the abuse potential of opioids.
Collapse
Affiliation(s)
- David R Maguire
- Department of Pharmacology and Addiction Research, Treatment & Training Center of Excellence, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
10
|
Berro LF, Zamarripa CA, Talley JT, Freeman KB, Rowlett JK. Effects of methadone, buprenorphine, and naltrexone on actigraphy-based sleep-like parameters in male rhesus monkeys. Addict Behav 2022; 135:107433. [PMID: 35901553 PMCID: PMC9495253 DOI: 10.1016/j.addbeh.2022.107433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 06/28/2022] [Accepted: 07/15/2022] [Indexed: 11/01/2022]
Abstract
Opioid use disorder (OUD) has been associated with the emergence of sleep disturbances. Although effective treatments for OUD exist, evidence suggests that these treatments also may be associated with sleep impairment. The extent to which these effects are an effect of OUD treatment or a result of chronic opioid use remains unknown. We investigated the acute effects of methadone, buprenorphine, and naltrexone on actigraphy-based sleep-like parameters in non-opioid-dependent male rhesus monkeys (Macaca mulatta, n = 5). Subjects were fitted with actigraphy monitors attached to primate collars to measure sleep-like parameters. Actigraphy recordings were conducted under baseline conditions, or following acute injections of vehicle, methadone (0.03-1.0 mg/kg, i.m.), buprenorphine (0.01-1.0 mg/kg, i.m.), or naltrexone (0.03-1.0 mg/kg, i.m.) in the morning (4 h after "lights on") or in the evening (1.5 h before "lights off"). Morning and evening treatments with methadone or buprenorphine significantly increased sleep latency and decreased sleep efficiency. The effects of buprenorphine on sleep-like measures resulted in a biphasic dose-response function, with the highest doses not disrupting actigraphy-based sleep. Buprenorphine induced a much more robust increase in sleep latency and decrease in sleep efficiency compared to methadone, particularly with evening administration, and detrimental effects of buprenorphine on sleep-like measures were observed up to 25.5 h after drug injection. Treatment with naltrexone, on the other hand, significantly improved sleep-like measures, with evening treatments improving both sleep latency and sleep efficiency. The currently available pharmacotherapies for OUD significantly alter sleep-like parameters in non-opioid-dependent monkeys, and opioid-dependent mechanisms may play a significant role in sleep-wake regulation.
Collapse
Affiliation(s)
- Lais F Berro
- Department of Psychiatry & Human Behavior, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA.
| | - C Austin Zamarripa
- Department of Psychiatry & Human Behavior, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA
| | - Joseph T Talley
- Department of Psychiatry & Human Behavior, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA
| | - Kevin B Freeman
- Department of Psychiatry & Human Behavior, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA
| | - James K Rowlett
- Department of Psychiatry & Human Behavior, University of Mississippi Medical Center, 2500 N State Street, Jackson, MS 39216, USA
| |
Collapse
|
11
|
Maguire DR, France CP. Effects of Daily Methocinnamox Treatment on Fentanyl Self-Administration in Rhesus Monkeys. J Pharmacol Exp Ther 2022; 382:181-187. [PMID: 35643857 PMCID: PMC9341267 DOI: 10.1124/jpet.122.001233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/04/2022] [Indexed: 08/03/2023] Open
Abstract
Methocinnamox (MCAM), a long-acting μ-opioid receptor antagonist, attenuates the positive reinforcing effects of opioids, such as heroin and fentanyl, suggesting it could be an effective treatment of opioid use disorder (OUD). Because treatment of OUD often involves repeated administration of a medication, this study evaluated effects of daily injections of a relatively small dose of MCAM on fentanyl self-administration and characterized the shift in the fentanyl dose-effect curve. Rhesus monkeys (3 males and 2 females) lever-pressed for intravenous infusions of fentanyl (0.032-10 μg/kg infusion) or cocaine (32-100 μg/kg infusion) under a fixed-ratio 30 schedule. MCAM (0.032 mg/kg) or naltrexone (0.0032-0.032 mg/kg) was administered subcutaneously 60 or 15 minutes, respectively, before sessions. When administered acutely, naltrexone and MCAM decreased fentanyl self-administration, with effects of naltrexone lasting less than 24 hours and effects of MCAM lasting for up to 3 days. Daily MCAM treatment attenuated responding for fentanyl, but not cocaine; effects were maintained for the duration of treatment with responding recovering quickly (within 2 days) following discontinuation of treatment. MCAM treatment shifted the fentanyl dose-effect curve in a parallel manner approximately 20-fold to the right. Naltrexone pretreatment decreased fentanyl intake with equal potency before and after MCAM treatment, confirming sensitivity of responding to antagonism by an opioid receptor antagonist. Although antagonist effects of treatment with a relatively small dose were surmountable, MCAM produced sustained and selective attenuation of opioid self-administration, supporting the view that it could be an effective treatment of OUD. SIGNIFICANCE STATEMENT: Opioid use disorder and opioid overdose continue to be significant public health challenges despite the availability of effective treatments. Methocinnamox (MCAM) is a long-acting μ-opioid receptor antagonist that blocks the reinforcing and ventilatory depressant effects of opioids in nonhuman subjects. This study demonstrates that daily treatment with MCAM reliably and selectively decreases fentanyl self-administration, further supporting the potential therapeutic utility of this novel antagonist.
Collapse
Affiliation(s)
- David R Maguire
- Department of Pharmacology, (D.R.M., C.P.F.), Addiction Research, Treatment & Training Center of Excellence (D.R.M., C.P.F.), and Department of Psychiatry (C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Charles P France
- Department of Pharmacology, (D.R.M., C.P.F.), Addiction Research, Treatment & Training Center of Excellence (D.R.M., C.P.F.), and Department of Psychiatry (C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
12
|
Han Y, Cao L, Yuan K, Shi J, Yan W, Lu L. Unique Pharmacology, Brain Dysfunction, and Therapeutic Advancements for Fentanyl Misuse and Abuse. Neurosci Bull 2022; 38:1365-1382. [PMID: 35570233 PMCID: PMC9107910 DOI: 10.1007/s12264-022-00872-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/13/2022] [Indexed: 11/20/2022] Open
Abstract
Fentanyl is a fully synthetic opioid with analgesic and anesthetic properties. It has become a primary driver of the deadliest opioid crisis in the United States and elsewhere, consequently imposing devastating social, economic, and health burdens worldwide. However, the neural mechanisms that underlie the behavioral effects of fentanyl and its analogs are largely unknown, and approaches to prevent fentanyl abuse and fentanyl-related overdose deaths are scarce. This review presents the abuse potential and unique pharmacology of fentanyl and elucidates its potential mechanisms of action, including neural circuit dysfunction and neuroinflammation. We discuss recent progress in the development of pharmacological interventions, anti-fentanyl vaccines, anti-fentanyl/heroin conjugate vaccines, and monoclonal antibodies to attenuate fentanyl-seeking and prevent fentanyl-induced respiratory depression. However, translational studies and clinical trials are still lacking. Considering the present opioid crisis, the development of effective pharmacological and immunological strategies to prevent fentanyl abuse and overdose are urgently needed.
Collapse
|
13
|
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
|
14
|
Zamora JC, Smith HR, Jennings EM, Chavera TS, Kotipalli V, Jay A, Husbands SM, Disney A, Berg KA, Clarke WP. Long-term antagonism and allosteric regulation of mu opioid receptors by the novel ligand, methocinnamox. Pharmacol Res Perspect 2021; 9:e00887. [PMID: 34713624 PMCID: PMC8554411 DOI: 10.1002/prp2.887] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 09/30/2021] [Indexed: 01/05/2023] Open
Abstract
Opioid overdose is a leading cause of death in the United States. The only treatment available currently is the competitive antagonist, naloxone (Narcan® ). Although naloxone is very effective and has saved many lives, as a competitive antagonist it has limitations. Due to the short half-life of naloxone, renarcotization can occur if the ingested opioid agonist remains in the body longer. Moreover, because antagonism by naloxone is surmountable, renarcotization can also occur in the presence of naloxone if a relatively larger dose of opioid agonist is taken. In such circumstances, a long-lasting, non-surmountable antagonist would offer an improvement in overdose treatment. Methocinnamox (MCAM) has been reported to have a long duration of antagonist action at mu opioid receptors in vivo. In HEK cells expressing the human mu opioid receptor, MCAM antagonism of mu agonist-inhibition of cAMP production was time-dependent, non-surmountable and non-reversible, consistent with (pseudo)-irreversible binding. In vivo, MCAM injected locally into the rat hindpaw antagonized mu agonist-mediated inhibition of thermal allodynia for up to 96 h. By contrast, antagonism by MCAM of delta or kappa agonists in HEK cells and in vivo was consistent with simple competitive antagonism. Surprisingly, MCAM also shifted the concentration-response curves of mu agonists in HEK cells in the absence of receptor reserve in a ligand-dependent manner. The shift in the [D-Ala2 ,N-MePhe4 ,Gly-ol5 ]-enkephalin (DAMGO) concentration-response curve by MCAM was insensitive to naloxone, suggesting that in addition to (pseudo)-irreversible orthosteric antagonism, MCAM acts allosterically to alter the affinity and/or intrinsic efficacy of mu agonists.
Collapse
Affiliation(s)
- Joshua C. Zamora
- Department of PharmacologyUT Health San AntonioSan AntonioTexasUSA
| | - Hudson R. Smith
- Department of PharmacologyUT Health San AntonioSan AntonioTexasUSA
| | | | | | - Varun Kotipalli
- Department of PharmacologyUT Health San AntonioSan AntonioTexasUSA
| | - Aleasha Jay
- Department of PharmacologyUT Health San AntonioSan AntonioTexasUSA
| | | | - Alex Disney
- Department of Pharmacy and PharmacologyUniversity of BathBathUnited Kingdom
| | - Kelly A. Berg
- Department of PharmacologyUT Health San AntonioSan AntonioTexasUSA
- Addiction ResearchTreatment & Training Center of ExcellenceUT Health San AntonioSan AntonioTexasUSA
| | - William P. Clarke
- Department of PharmacologyUT Health San AntonioSan AntonioTexasUSA
- Addiction ResearchTreatment & Training Center of ExcellenceUT Health San AntonioSan AntonioTexasUSA
| |
Collapse
|
15
|
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: 1.8] [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
|
16
|
Jimenez VM, Castaneda G, France CP. Methocinnamox Reverses and Prevents Fentanyl-Induced Ventilatory Depression in Rats. J Pharmacol Exp Ther 2021; 377:29-38. [PMID: 33431611 DOI: 10.1124/jpet.120.000387] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/06/2021] [Indexed: 11/22/2022] Open
Abstract
Opioid use disorder affects over 2 million Americans with an increasing number of deaths due to overdose from the synthetic opioid fentanyl and its analogs. The Food and Drug Administration-approved opioid receptor antagonist naloxone (e.g., Narcan) is used currently to treat overdose; however, a short duration of action limits its clinical utility. Methocinnamox (MCAM) is a long-lasting opioid receptor antagonist that may reverse and prevent the ventilatory-depressant effects of fentanyl. This study compared the ability of naloxone (0.0001-10 mg/kg) and MCAM (0.0001-10 mg/kg) to reverse and prevent ventilatory depression by fentanyl and compared the duration of action of MCAM intravenously and subcutaneously in two procedures: ventilation and warm-water tail withdrawal. In male Sprague-Dawley rats (N = 8), fentanyl (0.0032-0.178 mg/kg, i.v.) decreased minute volume in a dose- and time-dependent manner with a dose of 0.178 mg/kg decreasing VE to less than 40% of control. MCAM and naloxone reversed the ventilatory-depressant effects of 0.178 mg/kg fentanyl in a dose-related manner. The day after antagonist administration, MCAM but not naloxone attenuated the ventilatory-depressant effects of fentanyl. The duration of action of MCAM lasted up to 3 days and at least 2 weeks after intravenous and subcutaneous administration, respectively. MCAM attenuated the antinociceptive effects of fentanyl, with antagonism lasting up to 5 days and more than 2 weeks after intravenous and subcutaneous administration, respectively. Reversal and prolonged antagonism by MCAM might provide an effective treatment option for the opioid crisis, particularly toxicity from fentanyl and related highly potent analogs. SIGNIFICANCE STATEMENT: This study demonstrates that like naloxone, methocinnamox (MCAM) reverses the ventilatory-depressant effects of fentanyl in a time- and dose-related manner. However, unlike naloxone, the duration of action of MCAM was greater than 2 weeks when administered subcutaneously and up to 5 days when administered intravenously. These data suggest that MCAM might be particularly useful for rescuing individuals from opioid overdose, including fentanyl overdose, as well as protecting against the reemergence of ventilatory depression (renarconization).
Collapse
Affiliation(s)
- Victor M Jimenez
- Departments of Pharmacology (V.M.J., G.C., C.P.F.), Psychiatry (C.P.F.), and Addiction Research, Treatment and Training Center of Excellence (V.M.J., G.C., C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Gabriel Castaneda
- Departments of Pharmacology (V.M.J., G.C., C.P.F.), Psychiatry (C.P.F.), and Addiction Research, Treatment and Training Center of Excellence (V.M.J., G.C., C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Charles P France
- Departments of Pharmacology (V.M.J., G.C., C.P.F.), Psychiatry (C.P.F.), and Addiction Research, Treatment and Training Center of Excellence (V.M.J., G.C., C.P.F.), University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
17
|
France CP, Ahern GP, Averick S, Disney A, Enright HA, Esmaeli-Azad B, Federico A, Gerak LR, Husbands SM, Kolber B, Lau EY, Lao V, Maguire DR, Malfatti MA, Martinez G, Mayer BP, Pravetoni M, Sahibzada N, Skolnick P, Snyder EY, Tomycz N, Valdez CA, Zapf J. Countermeasures for Preventing and Treating Opioid Overdose. Clin Pharmacol Ther 2020; 109:578-590. [PMID: 33113208 DOI: 10.1002/cpt.2098] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
The only medication available currently to prevent and treat opioid overdose (naloxone) was approved by the US Food and Drug Administration (FDA) nearly 50 years ago. Because of its pharmacokinetic and pharmacodynamic properties, naloxone has limited utility under some conditions and would not be effective to counteract mass casualties involving large-scale deployment of weaponized synthetic opioids. To address shortcomings of current medical countermeasures for opioid toxicity, a trans-agency scientific meeting was convened by the US National Institute of Allergy and Infectious Diseases/National Institutes of Health (NIAID/NIH) on August 6 and 7, 2019, to explore emerging alternative approaches for treating opioid overdose in the event of weaponization of synthetic opioids. The meeting was initiated by the Chemical Countermeasures Research Program (CCRP), was organized by NIAID, and was a collaboration with the National Institute on Drug Abuse/NIH (NIDA/NIH), the FDA, the Defense Threat Reduction Agency (DTRA), and the Biomedical Advanced Research and Development Authority (BARDA). This paper provides an overview of several presentations at that meeting that discussed emerging new approaches for treating opioid overdose, including the following: (1) intranasal nalmefene, a competitive, reversible opioid receptor antagonist with a longer duration of action than naloxone; (2) methocinnamox, a novel opioid receptor antagonist; (3) covalent naloxone nanoparticles; (4) serotonin (5-HT)1A receptor agonists; (5) fentanyl-binding cyclodextrin scaffolds; (6) detoxifying biomimetic "nanosponge" decoy receptors; and (7) antibody-based strategies. These approaches could also be applied to treat opioid use disorder.
Collapse
Affiliation(s)
- Charles P France
- Department of Pharmacology, Addiction Research, Treatment and Training Center of Excellence, University of Texas Health Science Center, San Antonio, Texas, USA
| | | | - Saadyah Averick
- Neuroscience Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Alex Disney
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | | | - Babak Esmaeli-Azad
- CellCure (Stem Cell Division of CiBots, Inc.), San Diego, California, USA
| | - Arianna Federico
- CellCure (Stem Cell Division of CiBots, Inc.), San Diego, California, USA
| | - Lisa R Gerak
- Department of Pharmacology, Addiction Research, Treatment and Training Center of Excellence, University of Texas Health Science Center, San Antonio, Texas, USA
| | | | | | - Edmond Y Lau
- Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Victoria Lao
- Lawrence Livermore National Laboratory, Livermore, California, USA
| | - David R Maguire
- Department of Pharmacology, Addiction Research, Treatment and Training Center of Excellence, University of Texas Health Science Center, San Antonio, Texas, USA
| | | | - Girardo Martinez
- CellCure (Stem Cell Division of CiBots, Inc.), San Diego, California, USA
| | - Brian P Mayer
- Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Marco Pravetoni
- Department of Pharmacology, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | | | - Phil Skolnick
- Opiant Pharmaceuticals, Inc., Santa Monica, California, USA
| | - Evan Y Snyder
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Nestor Tomycz
- Neuroscience Institute, Allegheny Health Network, Pittsburgh, Pennsylvania, USA
| | - Carlos A Valdez
- Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Jim Zapf
- CellCure (Stem Cell Division of CiBots, Inc.), San Diego, California, USA
| |
Collapse
|
18
|
Townsend EA. The lasting impact of methocinnamox on opioid self-administration. Neuropsychopharmacology 2020; 45:1963-1964. [PMID: 32444781 PMCID: PMC7547696 DOI: 10.1038/s41386-020-0717-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/12/2020] [Indexed: 11/09/2022]
Affiliation(s)
- E Andrew Townsend
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA, 23298, USA.
| |
Collapse
|