1
|
Tan LA, Gajipara N, Sun L, Bacolod M, Zhou Y, Namchuk M, Cunningham JI. In vivo Characterization of the Opioid Receptor-Binding Profiles of Samidorphan and Naltrexone in Rats: Comparisons at Clinically Relevant Concentrations. Neuropsychiatr Dis Treat 2022; 18:2497-2506. [PMID: 36345421 PMCID: PMC9636859 DOI: 10.2147/ndt.s373195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 10/07/2022] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION The atypical antipsychotic olanzapine is approved for the treatment of schizophrenia and bipolar I disorder; however, weight gain and metabolic dysregulation associated with olanzapine therapy have limited its clinical utility. In clinical studies, treatment with the combination of olanzapine and the opioid receptor antagonist samidorphan (OLZ/SAM) mitigated olanzapine-associated weight gain while providing antipsychotic efficacy similar to that of olanzapine. Although samidorphan is structurally similar to the opioid receptor antagonist naltrexone, the two differ in their pharmacokinetics and in vitro binding affinities to mu, delta, and kappa opioid receptors (MOR, DOR, and KOR, respectively). The objective of this series of nonclinical studies was to compare the in vivo binding profiles of samidorphan and naltrexone and their receptor occupancies at MOR, DOR, and KOR in rat brains. METHODS Male rats were injected with samidorphan or naltrexone to obtain total and unbound plasma and brain concentrations representing levels observed in humans at clinically relevant oral doses. Subsequently, samidorphan and naltrexone brain receptor occupancy at MOR, DOR, and KOR was measured using ultra-performance liquid chromatography and high-resolution accurate-mass mass spectrometry. RESULTS A dose-dependent increase in samidorphan occupancy was observed at MOR, DOR, and KOR (EC50: 5.1, 54.7, and 42.9 nM, respectively). Occupancy of naltrexone at MOR (EC50: 15.5 nM) and KOR was dose dependent; minimal DOR occupancy was detected. At the clinically relevant unbound brain concentration of 23.1 nM, samidorphan bound to MOR, DOR, and KOR with 93.2%, 36.1%, and 41.9% occupancy, respectively. At 33.5 nM, naltrexone bound to MOR and KOR with 79.4% and 9.4% occupancy, respectively, with no binding at DOR. DISCUSSION At clinically relevant concentrations, samidorphan occupied MOR, DOR, and KOR, whereas naltrexone occupied only MOR and KOR. The binding profile of samidorphan differs from that of naltrexone, with potential clinical implications.
Collapse
Affiliation(s)
| | | | - Lei Sun
- Alkermes, Inc, Waltham, MA, USA
| | | | | | | | | |
Collapse
|
2
|
Klein ME, Grice AB, Sheth S, Go M, Murrough JW. Pharmacological Treatments for Anhedonia. Curr Top Behav Neurosci 2022; 58:467-489. [PMID: 35507281 DOI: 10.1007/7854_2022_357] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Anhedonia - the reduced ability to experience or respond to pleasure - is an important symptom domain for many psychiatric disorders. It is particularly relevant to depression and other mood disorders and it is a diagnostic criterion of a major depressive episode. Developing safe and effective pharmacological interventions for anhedonia is a critical public health need. The current chapter will review the state of the field with respect to both the efficacy of currently available pharmacotherapies for anhedonia and the recent clinical research focusing on new brain targets, including the kappa-opioid receptor and the KCNQ2/3 receptors. The evidence for anti-anhedonic effects of ketamine and psychedelic agents will be reviewed, as well.
Collapse
Affiliation(s)
- Matthew E Klein
- Depression and Anxiety Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Ariela Buxbaum Grice
- Depression and Anxiety Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sahil Sheth
- Depression and Anxiety Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Megan Go
- Depression and Anxiety Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James W Murrough
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
3
|
Gerum M, Simonin F. Behavioral characterization, potential clinical relevance and mechanisms of latent pain sensitization. Pharmacol Ther 2021; 233:108032. [PMID: 34763010 DOI: 10.1016/j.pharmthera.2021.108032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 10/22/2021] [Accepted: 11/03/2021] [Indexed: 10/19/2022]
Abstract
Chronic pain is a debilitating disorder that can occur as painful episodes that alternates with bouts of remission and occurs despite healing of the primary insult. Those episodes are often triggered by stressful events. In the last decades, a similar situation has been evidenced in a wide variety of rodent models (including inflammatory pain, neuropathy and opioid-induced hyperalgesia) where animals develop a chronic latent hyperalgesia that silently persists after behavioral signs of pain resolution. This state, referred as latent pain sensitization, is due to the compensatory activation of antinociceptive systems, such as the opioid system or NPY and its receptors. A transitory phase of hyperalgesia can then be reinstated by pharmacological or genetic blockade of these antinociceptive systems or by submitting animals to acute stress. Those observations reveal that there is a constant endogenous analgesia responsible for chronic pain inhibition that might paradoxically contribute to maintain this maladaptive state and could then participate to the transition from acute to chronic pain. Thus, demonstration of the existence of this phenomenon in humans and a better understanding of the mechanisms by which latent pain sensitization develops and maintains over long periods of time will be of particular interest to help identifying new therapeutic strategies and targets for chronic pain treatment. The present review aims to recapitulate behavioral expression, potential clinical relevance, cellular mechanisms and intracellular signaling pathways involved so far in latent pain sensitization.
Collapse
Affiliation(s)
- Manon Gerum
- Biotechnologie et Signalisation Cellulaire, UMR7242 CNRS, Université de Strasbourg, Institut du Médicament de Strasbourg, Illkirch-Graffenstaden, France
| | - Frédéric Simonin
- Biotechnologie et Signalisation Cellulaire, UMR7242 CNRS, Université de Strasbourg, Institut du Médicament de Strasbourg, Illkirch-Graffenstaden, France.
| |
Collapse
|
4
|
MEYE: Web App for Translational and Real-Time Pupillometry. eNeuro 2021; 8:ENEURO.0122-21.2021. [PMID: 34518364 PMCID: PMC8489024 DOI: 10.1523/eneuro.0122-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 01/02/2023] Open
Abstract
Pupil dynamics alterations have been found in patients affected by a variety of neuropsychiatric conditions, including autism. Studies in mouse models have used pupillometry for phenotypic assessment and as a proxy for arousal. Both in mice and humans, pupillometry is noninvasive and allows for longitudinal experiments supporting temporal specificity; however, its measure requires dedicated setups. Here, we introduce a convolutional neural network that performs online pupillometry in both mice and humans in a web app format. This solution dramatically simplifies the usage of the tool for the nonspecialist and nontechnical operators. Because a modern web browser is the only software requirement, this choice is of great interest given its easy deployment and setup time reduction. The tested model performances indicate that the tool is sensitive enough to detect both locomotor-induced and stimulus-evoked pupillary changes, and its output is comparable to state-of-the-art commercial devices.
Collapse
|
5
|
Hijma HJ, Groeneveld GJ. Analgesic drug development: proof-of-mechanism and proof-of-concept in early phase clinical studies. MEDICINE IN DRUG DISCOVERY 2021. [DOI: 10.1016/j.medidd.2021.100083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
6
|
Pajser A, Fisher H, Pickens CL. Pre-training naltrexone increases conditioned fear learning independent of adolescent alcohol consumption history. Physiol Behav 2021; 229:113212. [PMID: 33069685 DOI: 10.1016/j.physbeh.2020.113212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/26/2020] [Accepted: 10/14/2020] [Indexed: 11/27/2022]
Abstract
Our previous research has shown a relationship between low voluntary alcohol consumption and high conditioned fear in male Long Evans rats. Here, we examined whether differences in the endogenous opioid systems might be responsible for these differences. Rats received 6 weeks of chronic intermittent to 20% alcohol (v/v) or water-only from PND 26-66. Based on their consumption during the last 2 weeks of alcohol access, the alcohol-access rats were divided into high drinking (>2.5 g/kg/24-h) or low drinking (<2 g/kg/24-h). Rats were then given injections of the preferential mu opioid receptor antagonist naltrexone (1 mg/kg, s.c.) or the selective kappa opioid receptor antagonist LY2456302 (10 mg/kg, s.c.) prior to fear conditioning and were then tested for conditioned fear 2 days later. Pre-training naltrexone increased conditioned suppression of lever-pressing during training and testing, with no differences between high versus low alcohol drinkers or between water-only versus alcohol access groups (averaged across drinking levels). There was no effect of LY2456302 on conditioned fear in any comparison. We also found no differences between high and low alcohol drinkers and no reliable effect of prior alcohol access (averaged across drinking levels) on conditioned fear. Our experiment replicates and extends previous demonstrations that a preferential MOR antagonist can increase fear learning using conditioned suppression of lever-pressing as a fear measure. However, additional research is needed to determine the cause of the differences in conditioned fear that we previously observed (as they were not observed in the current experiments).
Collapse
Affiliation(s)
- Alisa Pajser
- Department of Psychological Sciences, Kansas State University, Manhattan, KS 66506, USA
| | - Hayley Fisher
- Department of Psychological Sciences, Kansas State University, Manhattan, KS 66506, USA
| | - Charles L Pickens
- Department of Psychological Sciences, Kansas State University, Manhattan, KS 66506, USA.
| |
Collapse
|
7
|
The Role of Dynorphin and the Kappa Opioid Receptor in Schizophrenia and Major Depressive Disorder: A Translational Approach. Handb Exp Pharmacol 2021; 271:525-546. [PMID: 33459877 DOI: 10.1007/164_2020_396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The kappa opioid receptor (KOR) and its endogenous ligands dynorphins (DYN) have been implicated in the development or symptomatology of a variety of neuropsychiatric disorders. This review covers a brief history of the development of KOR agonists and antagonists, their effects in healthy volunteers, and the potential role of DYN/KOR dysfunction in schizophrenia and major depressive disorder from a translational perspective. The potential role of DYN/KOR dysfunction in schizophrenia is based on several lines of evidence. Selective KOR agonists induce affective states in healthy volunteers with similarities to the symptoms of schizophrenia. Studies have shown increased DYN in patients with schizophrenia, although the data have been mixed. Finally, meta-analytic data have shown that opioid antagonists are associated with reductions in the symptoms of schizophrenia. The potential role of DYN/KOR dysfunction in major depressive disorder is also based on a combination of preclinical and clinical data. Selective KOR agonists have shown pro-depressive effects in human volunteers, while selective KOR antagonists have shown robust efficacy in several preclinical models of antidepressant activity. Small studies have shown that nonselective KOR antagonists may have efficacy in treatment-resistant depression. Additionally, recent clinical data have shown that the KOR may be an effective target for treating anhedonia, a finding relevant to both schizophrenia and depression. Finally, recommendations are provided for translating preclinical models for schizophrenia and major depressive disorder into the clinic.
Collapse
|
8
|
Margolis EB, Wallace TL, Van Orden LJ, Martin WJ. Differential effects of novel kappa opioid receptor antagonists on dopamine neurons using acute brain slice electrophysiology. PLoS One 2020; 15:e0232864. [PMID: 33373369 PMCID: PMC7771853 DOI: 10.1371/journal.pone.0232864] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 12/01/2020] [Indexed: 12/03/2022] Open
Abstract
Activation of the kappa opioid receptor (KOR) contributes to the aversive properties of stress, and modulates key neuronal circuits underlying many neurobehavioral disorders. KOR agonists directly inhibit ventral tegmental area (VTA) dopaminergic neurons, contributing to aversive responses (Margolis et al. 2003, 2006); therefore, selective KOR antagonists represent a novel therapeutic approach to restore circuit function. We used whole cell electrophysiology in acute rat midbrain slices to evaluate pharmacological properties of four novel KOR antagonists: BTRX-335140, BTRX-395750, PF-04455242, and JNJ-67953964. Each compound concentration-dependently reduced the outward current induced by the KOR selective agonist U-69,593. BTRX-335140 and BTRX-395750 fully blocked U-69,593 currents (IC50 = 1.2 ± 0.9 and 1.2 ± 1.3 nM, respectively). JNJ-67953964 showed an IC50 of 3.0 ± 4.6 nM. PF-04455242 exhibited partial antagonist activity asymptoting at 55% blockade (IC50 = 6.7 ± 15.1 nM). In 3/8 of neurons, 1 μM PF-04455242 generated an outward current independent of KOR activation. BTRX-335140 (10 nM) did not affect responses to saturating concentrations of the mu opioid receptor (MOR) agonist DAMGO or the delta opioid receptor (DOR) agonist DPDPE, while JNJ-67953964 (10 nM) partially blocked DAMGO and DPDPE responses. Importantly, BTRX-335140 (10 nM) rapidly washed out with complete recovery of U-69,593 responses within 10 min. Collectively, we show electrophysiological evidence of key differences amongst KOR antagonists that could impact their therapeutic potential and have not been observed using recombinant systems. The results of this study demonstrate the value of characterizing compounds in native neuronal tissue and within circuits implicated in the neurobehavioral disorders of interest.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Benzamides/pharmacology
- Biphenyl Compounds/pharmacology
- Dopaminergic Neurons/drug effects
- Dopaminergic Neurons/metabolism
- Electrophysiology
- Enkephalin, D-Penicillamine (2,5)-/pharmacology
- Male
- Membrane Potentials/drug effects
- Mesencephalon/metabolism
- Narcotic Antagonists/pharmacology
- Oxadiazoles/pharmacology
- Patch-Clamp Techniques/methods
- Piperidines/pharmacology
- Pyrrolidines/pharmacology
- Quinolines/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Opioid, kappa/antagonists & inhibitors
- Receptors, Opioid, kappa/metabolism
- Receptors, Opioid, mu/metabolism
- Sulfonamides/pharmacology
- Ventral Tegmental Area/drug effects
Collapse
Affiliation(s)
- Elyssa B. Margolis
- Department of Neurology, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States of America
- * E-mail:
| | - Tanya L. Wallace
- BlackThorn Therapeutics, San Francisco, CA, United States of America
| | | | - William J. Martin
- BlackThorn Therapeutics, San Francisco, CA, United States of America
| |
Collapse
|
9
|
The kappa opioid receptor antagonist aticaprant reverses behavioral effects from unpredictable chronic mild stress in male mice. Psychopharmacology (Berl) 2020; 237:3715-3728. [PMID: 32894343 PMCID: PMC7686052 DOI: 10.1007/s00213-020-05649-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/20/2020] [Indexed: 12/20/2022]
Abstract
RATIONALE Major depressive disorder is a leading cause of disability worldwide and is likely precipitated by chronic stress. Although many antidepressants are currently available, these drugs require weeks to months of daily administration before reduction of symptoms occurs and many patients remain treatment-resistant despite several courses of treatment. There is a pressing need for new treatments for stress-related disorders. Kappa opioid receptors (KORs) are a promising new therapeutic target for major depressive disorder and anhedonia because acute KOR blockade prevents many effects of stress in rodents. OBJECTIVES The following study assessed whether repeated treatment with the selective KOR antagonist aticaprant (also known as JNJ-67953964, and previously LY-2456302 and CERC-501) was effective in reversing behaviors in rodents following exposure to unpredictable chronic mild stress (UCMS). METHODS Adult male C57BL/6J mice were exposed to 4 weeks of UCMS. After 3 weeks of stress, aticaprant (10 mg/kg) was administered daily for 11 treatments. Behavioral assessments included the sucrose preference test, nesting, forced swim test, hot plate test, light-dark test, and social interaction test. RESULTS Aticaprant significantly reversed stress-induced deficits produced by UCMS on the SPT, nesting, FST, and hot plate test. The effects of aticaprant persisted through a stress and treatment recovery period. Aticaprant was not effective at reversing behavioral effects caused by stress in the light-dark and social interaction tests. CONCLUSIONS The results support further study of the role of KORs in regulating circuits related to reward, self-care, and cognition when they are disrupted by chronic stress. They are also consistent with the clinical development of aticaprant as a therapeutic for stress-related disorders targeted at anhedonia, such as depression and post-traumatic stress disorder.
Collapse
|
10
|
Reichard KL, Newton KA, Rivera ZMG, Sotero de Menezes PM, Schattauer SS, Land BB, Chavkin C. Regulation of Kappa Opioid Receptor Inactivation Depends on Sex and Cellular Site of Antagonist Action. Mol Pharmacol 2020; 98:548-558. [PMID: 32913138 PMCID: PMC7569314 DOI: 10.1124/molpharm.120.000124] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022] Open
Abstract
The prototypical member of the receptor-inactivating kappa opioid receptor (KOR) antagonists, norbinaltorphimine (norBNI), produces prolonged receptor inactivation by a cJun kinase mechanism. These antagonists have potential therapeutic utility in the treatment of stress disorders; however, additional preclinical characterization is necessary to understand important aspects of their action. In this study, we report that norBNI does not work as effectively in female mice as in males because of estrogen regulation of G protein receptor kinase (GRK); pretreatment of ovary-intact female mice with the selective GRK2/3 inhibitor, Compound 101, made females equally sensitive to norBNI as males. Prior observations suggested that in vivo treatment with norBNI does not produce long-lasting inhibition of KOR regulation of dopamine release in the nucleus accumbens. We assessed the persistence of norBNI receptor inactivation in subcellular compartments. Fast-scan cyclic voltammetry recordings confirmed that presynaptic inhibition of dopamine release by the KOR agonist U69,593 was not blocked by in vivo pretreatment with norBNI under conditions that prevented KOR-mediated aversion and analgesia. We employed a novel in vivo proxy sensor of KOR activation, adenovirus associated double floxed inverted-HyPerRed, and demonstrated that KOR activation stimulates cJun kinase-dependent reactive oxygen species (ROS) production in somatic regions of ventral tegmental area dopamine neurons, but did not activate ROS production in dopamine terminals. The compartment selective action helps explain how dopamine somatic, but not terminally expressed, KORs are inactivated by norBNI. These results further elucidate molecular signaling mechanisms mediating receptor-inactivating KOR antagonist action and advance medication development for this novel class of stress-resilience medications. SIGNIFICANCE STATEMENT: Kappa opioid receptor (KOR) antagonists are being developed as novel proresilience therapeutics for the treatment of mood and substance use disorders. This study showed that the long-acting KOR antagonists are affected by both the sex of the animal and the subcellular compartment in which the receptor is expressed.
Collapse
Affiliation(s)
- Kathryn L Reichard
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Keionna A Newton
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Zeena M G Rivera
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Paulo M Sotero de Menezes
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Selena S Schattauer
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Benjamin B Land
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| | - Charles Chavkin
- Neurobiology of Addiction, Pain, and Emotion (K.L.R., K.A.N., Z.M.G.R., P.M.S. ., S.S.S., B.B.L., C.C.), University of Washington Department of Pharmacology (K.L.R., K.A.N., Z.M.G.R., P.M.S.M. S.S.S., B.B.L., C.C.), University of Washington Graduate Program in Neuroscience (K.L.R., C.C.), Seattle, Washington
| |
Collapse
|
11
|
Jones JD, Babalonis S, Marcus R, Vince B, Kelsh D, Lofwall MR, Fraser H, Paterson B, Martinez S, Martinez DM, Nunes EV, Walsh SL, Comer SD. A randomized, double-blind, placebo-controlled study of the kappa opioid receptor antagonist, CERC-501, in a human laboratory model of smoking behavior. Addict Biol 2020; 25:e12799. [PMID: 31240842 DOI: 10.1111/adb.12799] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/14/2019] [Accepted: 05/27/2019] [Indexed: 12/18/2022]
Abstract
Preclinical data indicate that selective kappa opioid receptor antagonists reduce nicotine self-administration and withdrawal symptoms. The aim of the current study was to determine whether treatment with CERC-501, an orally available, potent, and selective kappa opioid receptor antagonist, could alleviate nicotine withdrawal and craving and mitigate mood alterations associated with nicotine withdrawal in humans. Healthy, adult cigarette smokers were enrolled into this randomized, multisite, double-blind, placebo-controlled, crossover study. Participants completed two 8-day treatment phases during which they received either CERC-501 (15 mg, p.o., once daily) or placebo. On the seventh day of each dosing phase, participants were admitted as inpatients for an 18-hour cigarette abstinence period followed by experimental testing. The primary outcome measures were (a) performance on the McKee Smoking Lapse test (ie, latency to smoke in exchange for money) and (b) number of cigarettes self-administered during a 60-minute ad lib smoking period. Other outcomes included measures of craving, mood, anxiety, nicotine withdrawal, and subjective effects of cigarette smoking. A total of 71 participants who smoked an average of approximately 23 cigarettes per day were enrolled, and 56 subjects completed the study. CERC-501 was well tolerated, but it did not significantly alter the latency to start smoking (CERC-501: 16.5 min vs placebo: 17.7 min) or the number of cigarettes smoked (CERC-501: 3.3 cigarettes vs placebo: 3.1 cigarettes). Compared with placebo, CERC-501 also did not affect cigarette craving, mood, anxiety, nicotine withdrawal, or subjective effects of smoking. These findings do not support a role for CERC-501 in the treatment of nicotine use disorder.
Collapse
Affiliation(s)
- Jermaine D. Jones
- Division on Substance Use Disorders New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons New York City New York USA
| | - Shanna Babalonis
- Department of Behavioral Science, Center on Drug and Alcohol Research University of Kentucky Lexington Kentucky USA
| | | | | | - Debra Kelsh
- Vince and Associates Overland Park Kansas USA
| | - Michelle R. Lofwall
- Department of Behavioral Science, Center on Drug and Alcohol Research University of Kentucky Lexington Kentucky USA
| | | | | | - Suky Martinez
- Division on Substance Use Disorders New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons New York City New York USA
- Translational Research Training Program in Addiction City College of New York New York City New York USA
| | - Diana M. Martinez
- Division on Substance Use Disorders New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons New York City New York USA
| | - Edward V. Nunes
- Division on Substance Use Disorders New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons New York City New York USA
| | - Sharon L. Walsh
- Department of Behavioral Science, Center on Drug and Alcohol Research University of Kentucky Lexington Kentucky USA
| | - Sandra D. Comer
- Division on Substance Use Disorders New York State Psychiatric Institute and Columbia University Vagelos College of Physicians and Surgeons New York City New York USA
| |
Collapse
|
12
|
Design, synthesis and biological evaluation of N-hydroxy-aminobenzyloxyarylamide analogues as novel selective κ opioid receptor antagonists. Bioorg Med Chem Lett 2020; 30:127236. [PMID: 32386980 DOI: 10.1016/j.bmcl.2020.127236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 01/05/2023]
Abstract
Aminobenzyloxyarylamide derivatives 1a-i and 2a-t were designed and synthesized as novel selective κ opioid receptor (KOR) antagonists. The benzoyl amide moiety of LY2456302 was changed into N-hydroxybenzamide and benzisoxazole-3(2H)-one to investigate whether it could increase the binding affinity or selectivity for KOR. All target compounds were evaluated in radioligand binding assays for opioid receptor binding affinity. These efforts led to the identification of compound 1c (κ Ki = 179.9 nM), which exhibited high affinity for KOR. Moreover, the selectivity of KOR over MOR and DOR increased nearly 2-fold and 7-fold, respectively, compared with (±)LY2456302.
Collapse
|
13
|
Reed B, Butelman ER, Kreek MJ. Kappa Opioid Receptor Antagonists as Potential Therapeutics for Mood and Substance Use Disorders. Handb Exp Pharmacol 2020; 271:473-491. [PMID: 33174064 DOI: 10.1007/164_2020_401] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The kappa opioid receptor (KOR) and its primary cognate ligands, the dynorphin peptides, are involved in diverse physiological processes. Disruptions to the KOR/dynorphin system have been found to likely play a role in multiple neuropsychological disorders, and hence KOR has emerged as a potential therapeutic target. Targeting KOR is complicated by close homology to the mu and delta opioid receptors (MOR and DOR), and many KOR ligands have at least moderate affinity to MOR and/or DOR. Animal models utilizing primarily very long-lasting selective KOR antagonists (>3 weeks following a single dose) have demonstrated that KOR antagonism attenuates certain anxiety-like and depression-like behaviors and blocks stress- and cue-induced reinstatement to drug seeking. Recently, relatively selective KOR antagonists with medication-like pharmacokinetic and pharmacodynamic properties and durations of action have been developed. One of these, JNJ-67953964 (also referred to as CERC-501, LY2456302, OpraKappa or Aticaprant) has been studied in humans, and shown to be safe, relatively KOR selective, and able to substantially attenuate binding of a KOR PET tracer to CNS localized KOR for greater than 24 h. While animal studies have indicated that compounds of this structural class are capable of normalizing withdrawal signs in animal models of cocaine and alcohol dependence and reducing cocaine and alcohol intake/seeking, additional studies are needed to determine the value of these second generation KOR antagonists in treating mood disorders and substance use disorders in humans.
Collapse
Affiliation(s)
- Brian Reed
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA.
| | - Eduardo R Butelman
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY, USA
| |
Collapse
|
14
|
Himmel H, Eriksson Faelker T. Pupillary function test in rat: Establishment of imaging setup and pharmacological validation within modified Irwin test. J Pharmacol Toxicol Methods 2019; 99:106588. [DOI: 10.1016/j.vascn.2019.106588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/26/2019] [Accepted: 05/23/2019] [Indexed: 01/23/2023]
|
15
|
Custodio-Patsey L, Donahue RR, Fu W, Lambert J, Smith BN, Taylor BK. Sex differences in kappa opioid receptor inhibition of latent postoperative pain sensitization in dorsal horn. Neuropharmacology 2019; 163:107726. [PMID: 31351975 DOI: 10.1016/j.neuropharm.2019.107726] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/11/2019] [Accepted: 07/25/2019] [Indexed: 12/31/2022]
Abstract
Tissue injury produces a delicate balance between latent pain sensitization (LS) and compensatory endogenous opioid receptor analgesia that continues for months, even after re-establishment of normal pain thresholds. To evaluate the contribution of mu (MOR), delta (DOR), and/or kappa (KOR) opioid receptors to the silencing of chronic postoperative pain, we performed plantar incision at the hindpaw, waited 21 days for the resolution of hyperalgesia, and then intrathecally injected subtype-selective ligands. We found that the MOR-selective inhibitor CTOP (1-1000 ng) dose-dependently reinstated mechanical hyperalgesia. Two DOR-selective inhibitors naltrindole (1-10 μg) and TIPP[Ψ] (1-20 μg) reinstated mechanical hyperalgesia, but only at the highest dose that also produced itching, licking, and tail biting. Both the prototypical KOR-selective inhibitors nor-BNI (0.1-10 μg) and the newer KOR inhibitor with more canonical pharmocodynamic effects, LY2456302 (0.1-10 μg), reinstated mechanical hyperalgesia. Furthermore, LY2456302 (10 μg) increased the expression of phosphorylated signal-regulated kinase (pERK), a marker of central sensitization, in dorsal horn neurons but not glia. Sex studies revealed that LY2456302 (0.3 μg) reinstated hyperalgesia and pERK expression to a greater degree in female as compared to male mice. Our results suggest that spinal MOR and KOR, but not DOR, maintain LS within a state of remission to reduce the intensity and duration of postoperative pain, and that endogenous KOR but not MOR analgesia is greater in female mice.
Collapse
Affiliation(s)
- Lilian Custodio-Patsey
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536-0298, USA
| | - Renée R Donahue
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536-0298, USA
| | - Weisi Fu
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536-0298, USA
| | - Joshua Lambert
- Department of Statistics, College of Arts and Sciences, University of Kentucky, 302 Multidisciplinary Science Building, Lexington, KY, 40536-0082, USA
| | - Bret N Smith
- Department of Physiology, College of Medicine, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536-0298, USA; Department of Neuroscience, College of Medicine, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY, 40536-0298, USA
| | - Bradley K Taylor
- Department of Anesthesiology, Pittsburgh Center for Pain Research, and the Pittsburgh Project to End Opioid Misuse, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
16
|
Butelman ER, McElroy BD, Prisinzano TE, Kreek MJ. Impact of Pharmacological Manipulation of the κ-Opioid Receptor System on Self-grooming and Anhedonic-like Behaviors in Male Mice. J Pharmacol Exp Ther 2019; 370:1-8. [PMID: 30975792 PMCID: PMC6538891 DOI: 10.1124/jpet.119.256354] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/09/2019] [Indexed: 12/11/2022] Open
Abstract
The kappa (κ) opioid receptor/dynorphin system modulates depression-like states and anhedonia, as well adaptations to stress and exposure to drugs of abuse. Several relatively short-acting small molecule κ-receptor antagonists have been synthesized, and their behavioral profile has been examined under some conditions. The hypothesis of this study is that pharmacological manipulations of the κ-receptor system will result in changes in ethologically relevant anhedonic-like behaviors in mice. Adult male C57BL/6j mice (n = 6-8) were examined for self-grooming behavior in the splash test (in which robust self-grooming is elicited by spraying the dorsum of the mouse with a sucrose solution). The κ-agonist salvinorin A (0.56-1.8 mg/kg) produced dose-dependent decreases in self-grooming, a marker of anhedonia. The selectivity, potency, and duration of action of two relatively short-acting κ-antagonists, LY2444296 [(S)-3-fluoro-4-(4-((2-(3-fluorophenyl) pyrrolidin-1-yl)methyl)phenoxy)benzamide] and LY2795050 [3-chloro-4-(4-(((2S)-2-pyridin-3-ylpyrrolidin-1-yl)methyl) phenoxy)benzamide], were studied for their effectiveness in preventing grooming deficits caused by salvinorin A (1.8 mg/kg). κ-selective doses of both LY2444296 (0.032-1 mg/kg) and LY2795050 (0.032-0.32 mg/kg) dose- and time-dependently prevented the grooming deficits caused by salvinorin A (1.8 m/kg). We also found that a κ-selective dose of each of these antagonists decreased immobility in the forced swim test, a common test of anti-anhedonia effects. This study shows that the κ-receptor system is involved in an ethologically relevant measure of anhedonia, and that κ-selective doses of these antagonists can produce effects consistent with rapid anti-anhedonia. SIGNIFICANCE STATEMENT: Activation of the κ-opioid receptor system results in grooming deficits in mice, an ethologically relevant marker of anhedonia. Shorter acting κ-antagonists are able to cause effects consistent with rapid antianhedonia.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Bryan D McElroy
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Thomas E Prisinzano
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| |
Collapse
|
17
|
Mathew SJ, Rivas-Grajales AM. "Does the opioid system block or enhance the antidepressant effects of ketamine?". ACTA ACUST UNITED AC 2019; 3. [PMID: 31276079 PMCID: PMC6604658 DOI: 10.1177/2470547019852073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sanjay J Mathew
- Menninger Department of Psychiatry and Behavioral Sciences, Houston, TX, USA.,Michael E. Debakey VA Medical Center, Houston, TX, USA
| | - Ana Maria Rivas-Grajales
- Menninger Department of Psychiatry and Behavioral Sciences, Houston, TX, USA.,Michael E. Debakey VA Medical Center, Houston, TX, USA
| |
Collapse
|
18
|
Browne CA, Lucki I. Targeting opioid dysregulation in depression for the development of novel therapeutics. Pharmacol Ther 2019; 201:51-76. [PMID: 31051197 DOI: 10.1016/j.pharmthera.2019.04.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 04/23/2019] [Indexed: 02/07/2023]
Abstract
Since the serendipitous discovery of the first class of modern antidepressants in the 1950's, all pharmacotherapies approved by the Food and Drug Administration for major depressive disorder (MDD) have shared a common mechanism of action, increased monoaminergic neurotransmission. Despite the widespread availability of antidepressants, as many as 50% of depressed patients are resistant to these conventional therapies. The significant length of time required to produce meaningful symptom relief with these medications, 4-6 weeks, indicates that other mechanisms are likely involved in the pathophysiology of depression which may yield more viable targets for drug development. For decades, no viable candidate target with a different mechanism of action to that of conventional therapies proved successful in clinical studies. Now several exciting avenues for drug development are under intense investigation. One of these emerging targets is modulation of endogenous opioid tone. This review will evaluate preclinical and clinical evidence pertaining to opioid dysregulation in depression, focusing on the role of the endogenous ligands endorphin, enkephalin, dynorphin, and nociceptin/orphanin FQ (N/OFQ) and their respective receptors, mu (MOR), delta (DOR), kappa (KOR), and the N/OFQ receptor (NOP) in mediating behaviors relevant to depression and anxiety. Finally, putative opioid based antidepressants that are under investigation in clinical trials, ALKS5461, JNJ-67953964 (formerly LY2456302 and CERC-501) and BTRX-246040 (formerly LY-2940094) will be discussed. This review will illustrate the potential therapeutic value of targeting opioid dysregulation in developing novel therapies for MDD.
Collapse
Affiliation(s)
- Caroline A Browne
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America
| | - Irwin Lucki
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America.
| |
Collapse
|
19
|
Guerrero M, Urbano M, Kim EK, Gamo AM, Riley S, Abgaryan L, Leaf N, Van Orden LJ, Brown SJ, Xie JY, Porreca F, Cameron MD, Rosen H, Roberts E. Design and Synthesis of a Novel and Selective Kappa Opioid Receptor (KOR) Antagonist (BTRX-335140). J Med Chem 2019; 62:1761-1780. [PMID: 30707578 PMCID: PMC6395531 DOI: 10.1021/acs.jmedchem.8b01679] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
κ opioid receptor (KOR) antagonists are potential pharmacotherapies for the treatment of migraine and stress-related mood disorders including depression, anxiety, and drug abuse, thus the development of novel KOR antagonists with an improved potency/selectivity profile and medication-like duration of action has attracted the interest of the medicinal chemistry community. In this paper, we describe the discovery of 1-(6-ethyl-8-fluoro-4-methyl-3-(3-methyl-1,2,4-oxadiazol-5-yl)quinolin-2-yl)- N-(tetrahydro-2 H-pyran-4-yl)piperidin-4 amine (CYM-53093, BTRX-335140) as a potent and selective KOR antagonist, endowed with favorable in vitro ADMET and in vivo pharmacokinetic profiles and medication-like duration of action in rat pharmacodynamic experiments. Orally administered CYM-53093 showed robust efficacy in antagonizing KOR agonist-induced prolactin secretion and in tail-flick analgesia in mice. CYM-53093 exhibited a broad selectivity over a panel of off-target proteins. This compound is in phase 1 clinical trials for the treatment of neuropsychiatric disorders wherein dynorphin is thought to contribute to the underlying pathophysiology.
Collapse
Affiliation(s)
- Miguel Guerrero
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Mariangela Urbano
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Eun-Kyong Kim
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Ana M. Gamo
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Sean Riley
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Lusine Abgaryan
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Nora Leaf
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Lori Jean Van Orden
- BlackThorn Therapeutics, Inc. 780 Brannan St. San Francisco CA 94103, United States
| | - Steven J. Brown
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Jennifer Y. Xie
- Current address: Department of Basic Sciences, New York Institute of Technology College of Osteopathic Medicine @ Arkansas State University, Jonesboro, AR 72446, United States
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, United States
| | - Frank Porreca
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, United States
| | - Michael D. Cameron
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida 33458, United States
| | - Hugh Rosen
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| | - Edward Roberts
- Department of Molecular Medicine, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, United States
| |
Collapse
|
20
|
Pathak S, Vince B, Kelsh D, Shram MJ, Setnik B, Lu H, Nangia N, Stanford AD, Ehrich E. Abuse Potential of Buprenorphine/Samidorphan Combination Compared to Buprenorphine and Placebo: A Phase 1 Randomized Controlled Trial. J Clin Pharmacol 2018; 59:206-217. [DOI: 10.1002/jcph.1280] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/05/2018] [Indexed: 11/10/2022]
Affiliation(s)
| | - Bradley Vince
- Vince & Associates Clinical Research
- Altasciences; Overland Park KS USA
| | - Debra Kelsh
- Vince & Associates Clinical Research
- Altasciences; Overland Park KS USA
| | - Megan J. Shram
- University of Toronto; Department of Pharmacology & Toxicology; Toronto ON Canada
- Altreos Research Partners; Toronto ON Canada
| | - Beatrice Setnik
- University of Toronto; Department of Pharmacology & Toxicology; Toronto ON Canada
- Syneos Health; Raleigh NC USA
| | - Hong Lu
- Alkermes, Inc.; Waltham MA USA
| | | | | | | |
Collapse
|
21
|
Reed B, Butelman ER, Fry RS, Kimani R, Kreek MJ. Repeated Administration of Opra Kappa (LY2456302), a Novel, Short-Acting, Selective KOP-r Antagonist, in Persons with and without Cocaine Dependence. Neuropsychopharmacology 2018; 43:739-750. [PMID: 28857070 PMCID: PMC5809790 DOI: 10.1038/npp.2017.205] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/14/2017] [Accepted: 08/27/2017] [Indexed: 12/14/2022]
Abstract
The κ-opioid receptor (KOP-r) system and its endogenous ligands, the dynorphins, are involved in the neurobiological regulation of addictive states, and of mood. There are limited data on the impact of selective KOP-r antagonism in humans on basic biobehavioral functions, or on addictive diseases and mood disorders. Previously studied selective KOP-r antagonists have unusual pharmacodynamic and pharmacokinetic properties (slow development of KOP-r selectivity, extremely long duration of action) that limit translation to human studies. A recently developed selective KOP-r-antagonist, Opra Kappa (LY2456302; CERC-501), has medication-like duration of action, oral bioavailability, and target engagement. The current study is the first investigation of the effects of a KOP-r-antagonist in cocaine-dependent persons in comparison with normal volunteers. In a stress-minimized inpatient setting, we determined the neuroendocrine and neurobehavioral effects of repeated administration of an active dose of Opra Kappa (10 mg p.o. daily, four consecutive days in comparison with an initial baseline day). Healthy volunteers (n=40), persons diagnosed with cocaine dependence in early abstinence (<2 months, EACD) (n=23), and drug-free former cocaine-dependent persons (7-month to 25-year abstinence, DFFCD) (n=7) were studied, with measurements including circulating neuroendocrine hormones, affect, and, in cocaine-dependent persons, cocaine craving. Modest adverse events related to Opra Kappa included pruritus, observed in a subset of individuals. No significant change was observed in serum prolactin levels following Opra Kappa administration, but modest increases in circulating adrenocorticotropic hormone and cortisol were observed. No significant changes were noted in measures of depression or cocaine craving in this stress-minimized setting. Overall, these studies demonstrate that effects of 10 mg Opra Kappa are largely consistent with those predicted for a selective KOP-r antagonist. This medication regimen was tolerable, and is therefore feasible for further studies in cocaine-dependent persons.
Collapse
Affiliation(s)
- Brian Reed
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, NY, USA,Laboratory of the Biology of Addictive Diseases, Rockefeller University, Box 171, New York, NY 10065, USA, Tel: 212 327 8247, Fax: 212 327 8574, E-mail:
| | - Eduardo R Butelman
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, NY, USA
| | - Rebecca S Fry
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, NY, USA
| | - Rachel Kimani
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, NY, USA
| | - Mary Jeanne Kreek
- Laboratory of the Biology of Addictive Diseases, Rockefeller University, New York, NY, USA
| |
Collapse
|
22
|
Browne CA, Falcon E, Robinson SA, Berton O, Lucki I. Reversal of Stress-Induced Social Interaction Deficits by Buprenorphine. Int J Neuropsychopharmacol 2017; 21:164-174. [PMID: 29020387 PMCID: PMC5793841 DOI: 10.1093/ijnp/pyx079] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/22/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Patients with post-traumatic stress disorder frequently report persistent problems with social interactions, emerging after a traumatic experience. Chronic social defeat stress is a widely used rodent model of stress that produces robust and sustained social avoidance behavior. The avoidance of other rodents can be reversed by 28 days of treatment with selective serotonin reuptake inhibitors, the only pharmaceutical class approved by the U.S. Food and Drug Administration for treating post-traumatic stress disorder. In this study, the sensitivity of social interaction deficits evoked by 10 days of chronic social defeat stress to prospective treatments for post-traumatic stress disorder was examined. METHODS The effects of acute and repeated treatment with a low dose of buprenorphine (0.25 mg/kg/d) on social interaction deficits in male C57BL/6 mice by chronic social defeat stress were studied. Another cohort of mice was used to determine the effects of the selective serotonin reuptake inhibitor fluoxetine (10 mg/kg/d), the NMDA antagonist ketamine (10 mg/kg/d), and the selective kappa opioid receptor antagonist CERC-501 (1 mg/kg/d). Changes in mRNA expression of Oprm1 and Oprk1 were assessed in a separate cohort. RESULTS Buprenorphine significantly reversed social interaction deficits produced by chronic social defeat stress following 7 days of administration, but not after acute injection. Treatment with fluoxetine for 7 days, but not 24 hours, also reinstated social interaction behavior in mice that were susceptible to chronic social defeat. In contrast, CERC-501 and ketamine failed to reverse social avoidance. Gene expression analysis found: (1) Oprm1 mRNA expression was reduced in the hippocampus and increased in the frontal cortex of susceptible mice and (2) Oprk1 mRNA expression was reduced in the amygdala and increased in the frontal cortex of susceptible mice compared to non-stressed controls and stress-resilient mice. CONCLUSIONS Short-term treatment with buprenorphine and fluoxetine normalized social interaction after chronic social defeat stress. In concert with the changes in opioid receptor expression produced by chronic social defeat stress, we speculate that buprenorphine's efficacy in this model of post-traumatic stress disorder may be associated with the ability of this compound to engage multiple opioid receptors.
Collapse
Affiliation(s)
| | | | | | | | - Irwin Lucki
- Departments of Psychiatry, Philadelphia, Pennsylvania,Systems Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania,University of Pennsylvania, Philadelphia, Pennsylvania,Correspondence: Irwin Lucki, PhD, Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 ()
| |
Collapse
|
23
|
"Effects of the novel relatively short-acting kappa opioid receptor antagonist LY2444296 in behaviors observed after chronic extended-access cocaine self-administration in rats". Psychopharmacology (Berl) 2017; 234:2219-2231. [PMID: 28550455 PMCID: PMC5591939 DOI: 10.1007/s00213-017-4647-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/09/2017] [Indexed: 12/11/2022]
Abstract
RATIONALE The recruitment of the stress circuitry contributes to a shift from positive to negative reinforcement mechanisms sustaining long-term cocaine addiction. The kappa opioid receptor (KOPr) signaling is upregulated by stress and chronic cocaine exposure. While KOPr agonists induce anhedonia and dysphoria, KOPr antagonists display antidepressant and anxiolytic properties. Most of the knowledge on KOPr antagonism is based on drugs with unusual pharmacokinetic and pharmacodynamic properties, complicating interpretation of results. Here we characterized in vivo behavioral and neuroendocrine effects of the novel relatively short-acting KOPr antagonist LY2444296. To date, no study has investigated whether systemic KOPr blockade reduced anxiety-like and depressive-like behaviors in animals previously exposed to chronic extended access cocaine self-administration. OBJECTIVES We tested the effect of LY2444296 in blocking KOPr-mediated aversive and neuroendocrine effects. Then, we tested acute systemic LY2444296 in reducing anxiety- and depression-like behaviors, as well as releasing the stress hormone corticosterone (CORT), observed after chronic extended access (18 h/day for 14 days) cocaine self-administration. RESULTS LY2444296 blocked U69,593-induced place aversion and -reduced motor activity as well as U69,593-induced release of serum CORT, confirming its major site of action, without exerting an effect per se. Acute systemic administration of LY2444296 reduced anxiety-like and depressive-like behaviors, as well as CORT release, in rats tested after chronic extended access cocaine self-administration, but not in cocaine-naïve rats. CONCLUSIONS Results suggest that acute blockade of KOPr by a relatively short-acting antagonist produces therapeutic-like effects selectively in rats with a history of chronic extended access cocaine self-administration.
Collapse
|
24
|
Effects of the 5-HT1A receptor agonists buspirone and 8-OH-DPAT on pupil size in common marmosets. Behav Pharmacol 2017; 28:313-317. [DOI: 10.1097/fbp.0000000000000275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Naganawa M, Dickinson GL, Zheng MQ, Henry S, Vandenhende F, Witcher J, Bell R, Nabulsi N, Lin SF, Ropchan J, Neumeister A, Ranganathan M, Tauscher J, Huang Y, Carson RE. Receptor Occupancy of the κ-Opioid Antagonist LY2456302 Measured with Positron Emission Tomography and the Novel Radiotracer 11C-LY2795050. J Pharmacol Exp Ther 2016; 356:260-6. [PMID: 26628406 PMCID: PMC4727157 DOI: 10.1124/jpet.115.229278] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/30/2015] [Indexed: 11/22/2022] Open
Abstract
The κ-opioid receptor (KOR) is thought to play an important therapeutic role in a wide range of neuropsychiatric and substance abuse disorders, including alcohol dependence. LY2456302 is a recently developed KOR antagonist with high affinity and selectivity and showed efficacy in the suppression of ethanol consumption in rats. This study investigated brain penetration and KOR target engagement after single oral doses (0.5-25 mg) of LY2456302 in 13 healthy human subjects. Three positron emission tomography scans with the KOR antagonist radiotracer (11)C-LY2795050 were conducted at baseline, 2.5 hours postdose, and 24 hours postdose. LY2456302 was well tolerated in all subjects without serious adverse events. Distribution volume was estimated using the multilinear analysis 1 method for each scan. Receptor occupancy (RO) was derived from a graphical occupancy plot and related to LY2456302 plasma concentration to determine maximum occupancy (rmax) and IC50. LY2456302 dose dependently blocked the binding of (11)C-LY2795050 and nearly saturated the receptors at 10 mg, 2.5 hours postdose. Thus, a dose of 10 mg of LY2456302 appears well suited for further clinical testing. Based on the pharmacokinetic (PK)-RO model, the rmax and IC50 of LY2456302 were estimated as 93% and 0.58 ng/ml to 0.65 ng/ml, respectively. Assuming that rmax is 100%, IC50 was estimated as 0.83 ng/ml.
Collapse
Affiliation(s)
- Mika Naganawa
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Gemma L Dickinson
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Ming-Qiang Zheng
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Shannan Henry
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Francois Vandenhende
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Jennifer Witcher
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Robert Bell
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Nabeel Nabulsi
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Shu-Fei Lin
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Jim Ropchan
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Alexander Neumeister
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Mohini Ranganathan
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Johannes Tauscher
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Richard E Carson
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| |
Collapse
|
26
|
Abstract
This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants). This paper is the thirty-seventh consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2014 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (endogenous opioids and receptors), and the roles of these opioid peptides and receptors in pain and analgesia (pain and analgesia); stress and social status (human studies); tolerance and dependence (opioid mediation of other analgesic responses); learning and memory (stress and social status); eating and drinking (stress-induced analgesia); alcohol and drugs of abuse (emotional responses in opioid-mediated behaviors); sexual activity and hormones, pregnancy, development and endocrinology (opioid involvement in stress response regulation); mental illness and mood (tolerance and dependence); seizures and neurologic disorders (learning and memory); electrical-related activity and neurophysiology (opiates and conditioned place preferences (CPP)); general activity and locomotion (eating and drinking); gastrointestinal, renal and hepatic functions (alcohol and drugs of abuse); cardiovascular responses (opiates and ethanol); respiration and thermoregulation (opiates and THC); and immunological responses (opiates and stimulants).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|