Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 388(2): 244-256, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-37739803

RESUMEN

The number of drug overdoses and deaths has increased significantly over the past decade and co-use of opioids and stimulants is associated with greater likelihood of overdose and decreased likelihood of accessing treatment, compared with use of opioids alone. Potential adverse effects of opioid/stimulant mixtures, particularly methamphetamine, are not well characterized. Two structurally different drugs with agonist properties at µ-opioid receptors (MOR), fentanyl and heroin, and d-methamphetamine, alone and in mixtures, were assessed for their effects on ventilation in rats breathing normal air. Whole-body phethysmography chambers were equipped with a tower and swivel allowing infusions to indwelling intravenous catheters. After a 45-minute habituation period, saline, fentanyl, heroin, or d-methamphetamine, alone and in mixtures, was administered. Five minutes later, the opioid receptor antagonist naloxone or vehicle was injected. Fentanyl (0.0032-0.1 mg/kg) and heroin (0.32-3.2 mg/kg) decreased ventilation [frequency (f) and tidal volume (VT)] in a dose-related manner whereas d-methamphetamine (0.1-3.2 mg/kg) increased f to >400% of control and decreased VT to <60% of control, overall increasing minute volume (product of f and VT) to >240% of control. When combined, d-methamphetamine (0.1-3.2 mg/kg) attenuated the ventilatory depressant effects of fentanyl (0.1 mg/kg) and heroin (3.2 mg/kg). d-Methamphetamine did not alter the potency of naloxone to reverse the ventilatory depressant effects of fentanyl or heroin. These studies demonstrate that d-methamphetamine can attenuate the ventilatory depressant effects of moderate doses of opioid receptor agonists while not altering the potency of naloxone to reverse opioid hypoventilation. SIGNIFICANCE STATEMENT: Co-use of opioids and stimulants is associated with greater likelihood of overdose and decreased likelihood of accessing treatment, compared with use of opioids alone. Potential adverse effects of opioid/stimulant mixtures are not well characterized. This study reports that 1) d-methamphetamine attenuates the ventilatory depressant effects of moderate doses of two structurally different opioid receptor agonists, fentanyl and heroin, and 2) d-methamphetamine does not alter potency or effectiveness of naloxone to reverse the ventilatory depressant effects of these opioid receptor agonists.


Asunto(s)
Sobredosis de Droga , Metanfetamina , Masculino , Animales , Ratas , Heroína/farmacología , Fentanilo/efectos adversos , Analgésicos Opioides/efectos adversos , Metanfetamina/farmacología , Naloxona , Sobredosis de Droga/tratamiento farmacológico , Receptores Opioides
2.
J Pharmacol Exp Ther ; 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38409115

RESUMEN

The number of opioid overdose deaths has increased significantly over the past decade. The life-threatening effect of opioids is hypoventilation that can be reversed by the µ-opioid receptor (MOR) antagonist naloxone; however, because of the very short duration of action of naloxone, re-emergence of MOR agonist-induced hypoventilation can occur, requiring additional doses of naloxone. The MOR antagonist methocinnamox (MCAM) antagonizes hypoventilation by the non-morphinan fentanyl and the morphinan heroin in laboratory animals with an unusually long duration of action. Whole-body plethysmography was used to compare the potency and effectiveness of MCAM and naloxone for preventing and reversing hypoventilation by fentanyl, heroin, and the ultra-potent and longer-acting fentanyl analogs carfentanil and 3-methylfentanyl in male rats breathing normal air. Sessions comprised a 45-minute habituation period followed by intravenous (i.v.) administration of saline or an acute dose of MOR agonist. The rank order of potency to decrease ventilation was 3-methylfentanyl > carfentanil > fentanyl > heroin. MCAM (0.0001-0.1 mg/kg) and naloxone (0.0001-0.01 mg/kg) dose-dependently reversed hypoventilation by 3-methylfentanyl (0.01 mg/kg), carfentanil (0.01 mg/kg), fentanyl (0.1 mg/kg), or heroin (3.2 mg/kg). For prevention studies, MCAM, naloxone, or vehicle was administered i.v. 22, 46, or 70 hours prior to a MOR agonist. When administered 22 hours earlier, MCAM (0.1-1.0 mg/kg) but not naloxone (1.0 mg/kg) prevented hypoventilation by each MOR agonist. This study demonstrates the effectiveness of MCAM to reverse and prevent hypoventilation by MOR agonists including ultra-potent fentanyl analogs that have a long duration of action. Significance Statement The number of opioid overdose deaths increased over the past decade despite the availability of antagonists that can prevent and reverse the effects of opioids. This study demonstrates the effectiveness and long duration of action of the µ-opioid receptor (MOR) antagonist methocinnamox (MCAM) for reversing and preventing hypoventilation by MOR agonists including ultra-potent fentanyl analogs. These results provide support for the notion that MCAM has the potential to positively impact the ongoing opioid crisis by reversing and preventing opioid overdose.

3.
Behav Pharmacol ; 35(4): 161-171, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38660819

RESUMEN

Cannabis is a pharmacologically complex plant consisting of hundreds of potentially active compounds. One class of compounds present in cannabis that has received little attention are terpenes. Traditionally thought to impart aroma and flavor to cannabis, it has become increasingly recognized that terpenes might exert therapeutic effects themselves. Several recent reports have also indicated terpenes might behave as cannabinoid type 1 (CB1) receptor agonists. This study aimed to investigate whether several terpenes present in cannabis produce discriminative stimulus effects similar to or enhance the effects of Δ 9 -tetrahydrocannabinol (THC). Subsequent experiments explored other potential cannabimimetic effects of these terpenes. Rats were trained to discriminate THC from vehicle while responding under a fixed-ratio 10 schedule of food presentation. Substitution testing was performed with the CB receptor agonist JWH-018 and the terpenes linalool, limonene, γ-terpinene and α-humulene alone. Terpenes were also studied in combination with THC. Finally, THC and terpenes were tested in the tetrad assay to screen for CB1-receptor agonist-like effects. THC and JWH-018 dose-dependently produced responding on the THC-paired lever. When administered alone, none of the terpenes produced responding predominantly on the THC-paired lever. When administered in combination with THC, none of the terpenes enhanced the potency of THC, and in the case of α-humulene, decreased the potency of THC to produce responding on the THC-paired lever. While THC produced effects in all four tetrad components, none of the terpenes produced effects in all four components. Therefore, the terpenes examined in this report do not have effects consistent with CB1 receptor agonist properties in the brain.


Asunto(s)
Cannabis , Dronabinol , Terpenos , Animales , Terpenos/farmacología , Ratas , Dronabinol/farmacología , Masculino , Cannabinoides/farmacología , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Indoles/farmacología , Naftalenos/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Ratas Sprague-Dawley , Relación Dosis-Respuesta a Droga , Aprendizaje Discriminativo/efectos de los fármacos , Discriminación en Psicología/efectos de los fármacos
4.
J Pharmacol Exp Ther ; 384(3): 363-371, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36575032

RESUMEN

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.


Asunto(s)
Cocaína , Metanfetamina , Trastornos Relacionados con Opioides , Animales , Heroína , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Macaca mulatta , Receptores Opioides mu , Analgésicos Opioides/farmacología , Fentanilo/farmacología , Trastornos Relacionados con Opioides/tratamiento farmacológico , Cocaína/farmacología , Relación Dosis-Respuesta a Droga
5.
J Pharmacol Exp Ther ; 382(2): 181-187, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35643857

RESUMEN

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.


Asunto(s)
Cinamatos , Derivados de la Morfina , Trastornos Relacionados con Opioides , Analgésicos Opioides , Animales , Cinamatos/uso terapéutico , Cocaína/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Fentanilo/farmacología , Macaca mulatta , Masculino , Derivados de la Morfina/uso terapéutico , Naltrexona/farmacología , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/farmacología , Antagonistas de Narcóticos/uso terapéutico , Trastornos Relacionados con Opioides/tratamiento farmacológico , Receptores Opioides mu , Autoadministración
6.
J Pharmacol Exp Ther ; 380(3): 171-179, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34903563

RESUMEN

Drug overdose deaths involving synthetic opioids, primarily fentanyl, have risen dramatically over the past decade and are currently the driving force of the opioid epidemic in the United States. Fentanyl analogs with greater potency than fentanyl (e.g., carfentanil) pose serious risk to public health. Although fentanyl analogs are primarily encountered by humans as constituents of a mixture of drugs, research has primarily evaluated the effects of these drugs alone. The present study characterized interactions between mu opioid receptor agonists in seven male Sprague-Dawley rats trained to discriminate 10 µg/kg fentanyl from saline while responding under a fixed-ratio 10 schedule of food presentation. Dose-effect curves were determined for each drug alone and in binary mixtures (fentanyl:heroin, fentanyl:carfentanil, and heroin:carfentanil) at fixed-dose ratios of 3:1, 1:1, and 1:3 relative to the ED50 for each drug when given alone. Dose addition analyses were used to determine the nature of the drug-drug interaction for each mixture. Additive interactions were observed for all binary mixtures at each fixed dose ratio, except the 1:3 fentanyl:carfentanil mixture, which exhibited supra-additive effects at the 80% effect level. These results suggest a lack of a significant interaction between the discriminative stimulus effects of these mu opioid receptor agonists at the doses tested in this study. Future studies expanding these findings to the respiratory depressant effects of these drugs are of significant importance to rule out possible interactions directly relevant to opioid overdose that occur at doses much larger than those tested in this study. SIGNIFICANCE STATEMENT: In the United States, drug overdose deaths involving synthetic opioids, primarily fentanyls including superpotent fentanyl analogs (e.g., carfentanil), have increased 12-fold over the past decade. Although previous studies have evaluated the effects of carfentanil alone, fentanyl analogs are encountered by humans as a mixture with other drugs; this study determined the effects of mixtures of carfentanil and other opioids (fentanyl and heroin) to characterize interactions between these drugs that might contribute to their apparent increased lethality in humans.


Asunto(s)
Sobredosis de Droga , Receptores Opioides mu , Analgésicos Opioides/farmacología , Animales , Fentanilo/farmacología , Heroína , Masculino , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/agonistas , Estados Unidos
7.
Behav Pharmacol ; 33(6): 377-394, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35947066

RESUMEN

Drugs targeting mu opioid receptors are the mainstay of clinical practice for treating moderate-to-severe pain. While they can offer excellent analgesia, their use can be limited by adverse effects, including constipation, respiratory depression, tolerance, and abuse liability. Multifunctional ligands acting at mu opioid and nociceptin/orphanin FQ peptide receptors might provide antinociception with substantially improved adverse-effect profiles. This study explored one of these ligands, OREX-1038 (BU10038), in several assays in rodents and nonhuman primates. Binding and functional studies confirmed OREX-1038 to be a low-efficacy agonist at mu opioid and nociceptin/orphanin FQ peptide receptors and an antagonist at delta and kappa opioid receptors with selectivity for opioid receptors over other proteins. OREX-1038 had long-acting antinociceptive effects in postsurgical and complete Freund's adjuvant (CFA)-induced thermal hyperalgesia assays in rats and a warm water tail-withdrawal assay in monkeys. OREX-1038 was active for at least 24 h in each antinociception assay, and its effects in monkeys did not diminish over 22 days of daily administration. This activity was coupled with limited effects on physiological signs (arterial pressure, heart rate, and body temperature) and no evidence of withdrawal after administration of naltrexone or discontinuation of treatment in monkeys receiving OREX-1038 daily. Over a range of doses, OREX-1038 was only transiently self-administered, which diminished rapidly to nonsignificant levels; overall, both OREX-1038 and buprenorphine maintained less responding than remifentanil. These results support the concept of dual mu and nociceptin/orphanin FQ peptide receptor partial agonists having improved pharmacological profiles compared with opioids currently used to treat pain.


Asunto(s)
Analgésicos Opioides , Dolor , Analgésicos Opioides/efectos adversos , Animales , Isoquinolinas , Naltrexona/análogos & derivados , Dolor/tratamiento farmacológico , Fenilpropionatos , Ratas , Receptores Opioides/agonistas , Receptores Opioides mu/agonistas
8.
J Pharmacol Exp Ther ; 377(1): 29-38, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33431611

RESUMEN

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).


Asunto(s)
Cinamatos/uso terapéutico , Derivados de la Morfina/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Insuficiencia Respiratoria/tratamiento farmacológico , Animales , Cinamatos/administración & dosificación , Fentanilo/toxicidad , Inyecciones Intravenosas , Masculino , Derivados de la Morfina/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Narcóticos/toxicidad , Ratas , Ratas Sprague-Dawley , Insuficiencia Respiratoria/etiología , Insuficiencia Respiratoria/prevención & control
9.
Behav Pharmacol ; 32(4): 278-285, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33491991

RESUMEN

Intravenous drug self-administration remains the 'gold standard' for assessing abuse liability. Failure of a drug to maintain self-administration might indicate the absence of positive reinforcing effects but might also indicate the presence of aversive effects. Sensitivity to aversive and punishing effects of drugs (as well as nondrug stimuli) might collectively determine the likelihood of use, abuse and relapse. Using a choice procedure, this study compared the effects of remifentanil (mu opioid receptor agonist; 0.001-0.01 mg/kg/infusion) and histamine (H1-4 receptor agonist; 0.32-3.2 mg/kg/infusion), alone and in mixtures, to test the hypothesis that remifentanil/histamine mixtures are less reinforcing compared with remifentanil alone and less punishing compared with histamine alone. Male Sprague-Dawley rats (n = 10) chose between an intravenous infusion + a pellet and a pellet alone. Rats were indifferent to saline, chose remifentanil + a pellet over a pellet alone, and chose a pellet alone over histamine + a pellet. The effects of remifentanil/histamine mixtures generally were different from the constituent doses of histamine alone but not from remifentanil alone. A mixture containing 3.2 mg/kg/infusion histamine and either 0.001 or 0.0032 mg/kg/infusion remifentanil was not different from saline but was different from the effects of the constituent dose, insofar as choice increased compared with 3.2 mg/kg/infusion histamine alone and decreased compared with 0.001 or 0.0032 mg/kg/infusion remifentanil alone. Reinforcing doses of remifentanil combined with punishing doses of histamine can yield mixtures that are neither preferred nor avoided, offering 'proof-of-principle' for using drug mixtures to avoid adverse effects of opioid receptor agonists.


Asunto(s)
Conducta de Elección/efectos de los fármacos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/prevención & control , Histamina/farmacología , Refuerzo en Psicología , Remifentanilo , Autoadministración/psicología , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/farmacología , Animales , Conducta Animal/efectos de los fármacos , Formas de Dosificación , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Composición de Medicamentos/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Agonistas de los Receptores Histamínicos/farmacología , Infusiones Intravenosas/métodos , Ratas , Receptores Opioides mu/agonistas , Remifentanilo/efectos adversos , Remifentanilo/farmacología
10.
J Pharmacol Exp Ther ; 372(2): 205-215, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31748404

RESUMEN

There is an urgent need for new pharmacological treatments for substance use disorders, including opioid use disorder, particularly for use in relapse prevention. A combination of buprenorphine with naltrexone has shown particular promise, with clinical studies indicating a substantial improvement over treatment with naltrexone alone. OREX-1019 (formerly BU10119) is a compound that mimics the pharmacology of the buprenorphine/naltrexone combination. This study evaluated, in rhesus monkeys, the therapeutic potential of OREX-1019 for treating opioid use disorder. Pretreatment with OREX-1019 (0.01-0.3 mg/kg s.c.) dose-dependently decreased responding for the µ opioid receptor agonist remifentanil in rhesus monkeys but did not maintain levels of responding above vehicle when it was available for self-administration. OREX-1019 (0.01-1.0 mg/kg s.c.) also decreased cue- plus heroin-primed reinstatement of extinguished responding in monkeys that self-administered remifentanil but did not alter cue- plus cocaine-primed reinstatement of responding in monkeys that self-administered cocaine. OREX-1019 (0.3 mg/kg s.c.), like naltrexone (0.1 mg/kg s.c.), increased heart rate and blood pressure, produced overt observable signs, and eliminated food-maintained responding in monkeys treated chronically with morphine. These results confirm that OREX-1019 has little or no efficacy at µ opioid receptorsand has low abuse potential, and, combined with promising safety (clean profile vs. other off-target proteins including the hERG (human ether-a-go-go-related gene) K+ channel) and pharmacokinetic data (supporting administration by subcutaneous or sublingual routes, but with low oral bioavailability), suggest it could be a safe and effective alternative to current treatments for opioid use disorders particularly as applied to relapse prevention. SIGNIFICANCE STATEMENT: The novel opioid OREX-1019 potentially provides an improved relapse prevention agent for use in opioid use disorder. The current study demonstrates that in monkeys OREX-1019 is able to inhibit the self-administration of, and cue- plus heroin-primed reinstatement of, responding previously maintained by remifentanil.


Asunto(s)
Buprenorfina/uso terapéutico , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/farmacología , Trastornos Relacionados con Opioides/tratamiento farmacológico , Trastornos Relacionados con Opioides/prevención & control , Analgésicos Opioides/metabolismo , Animales , Técnicas de Observación Conductual , Presión Sanguínea/efectos de los fármacos , Buprenorfina/efectos adversos , Buprenorfina/farmacocinética , Cocaína/farmacología , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Femenino , Interacciones Alimento-Droga , Frecuencia Cardíaca/efectos de los fármacos , Heroína/metabolismo , Humanos , Macaca mulatta , Masculino , Morfina/metabolismo , Naltrexona/efectos adversos , Naltrexona/farmacocinética , Canales de Potasio/metabolismo , Receptores Opioides mu/metabolismo , Remifentanilo/farmacología , Prevención Secundaria , Autoadministración , Resultado del Tratamiento
11.
Behav Pharmacol ; 31(2&3): 233-248, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32101988

RESUMEN

The opioid epidemic underscores the need for safer and more effective treatments for pain. Combining opioid receptor agonists with drugs that relieve pain through nonopioid mechanisms could be a useful strategy for reducing the dose of opioid needed to treat pain, thereby reducing risks associated with opioids alone. Opioid/cannabinoid mixtures might be useful in this context; individually, opioids and cannabinoids have modest effects on cognition, and it is important to determine whether those effects occur with mixtures. Delay discounting and delayed matching-to-sample tasks were used to examine effects of the mu-opioid receptor agonist morphine (0.32-5.6 mg/kg), the cannabinoid CB1/CB2 receptor agonist CP55940 (0.0032-0.1 mg/kg), and morphine/CP55940 mixtures on impulsivity (n = 3) and memory (n = 4) in rhesus monkeys. Alone, each drug decreased rate of responding without modifying choice in the delay-discounting task, and morphine/CP55940 mixtures reduced choice of one pellet in a delay dependent manner, with monkeys instead choosing delayed delivery of the larger number of pellets. With the exception of one dose in one monkey, accuracy in the delayed matching-to-sample task was not altered by either drug alone. Morphine/CP55940 mixtures decreased accuracy in two monkeys, but the doses in the mixture were equal to or greater than doses that decreased accuracy or response rate with either drug alone. Rate-decreasing effects of morphine/CP55940 mixtures were additive. These data support the notion that opioid/cannabinoid mixtures that might be effective for treating pain do not have greater, and might have less, adverse effects compared with larger doses of each drug alone.


Asunto(s)
Analgésicos Opioides/farmacología , Cannabinoides/farmacología , Conducta Impulsiva/efectos de los fármacos , Memoria/efectos de los fármacos , Analgésicos Opioides/metabolismo , Animales , Agonistas de Receptores de Cannabinoides/farmacología , Cannabinoides/metabolismo , Cognición/efectos de los fármacos , Ciclohexanoles/farmacología , Descuento por Demora/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada/métodos , Femenino , Macaca mulatta , Masculino , Morfina/farmacología , Receptores Opioides mu/agonistas
12.
Behav Pharmacol ; 31(2&3): 216-220, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31577559

RESUMEN

The behavioral economics framework has been used extensively to study factors that control operant behavior, including quantification of reinforcing effectiveness of drugs of abuse. Generally, consumption of a commodity decreases with increasing price, and the rate of decrease reflects demand elasticity, which is inversely related to reinforcing effectiveness. Drugs with low elasticity have greater effectiveness than those with greater elasticity. Price is often manipulated by varying the number of responses required to obtain an infusion (e.g. fixed ratio schedule); however, many studies present the fixed ratio in only one order (usually ascending), which could introduce sequence effects that influence estimates of demand. This study examined the impact of the order of fixed ratio presentation on demand for the mu opioid receptor agonist remifentanil (0.0032 mg/kg/infusion) using an ascending and a mixed order of fixed ratio presentation. Seven male rats lever pressed for intravenous infusions. The fixed ratio varied across 3-session blocks, yielding a demand curve. During the first and third phases, the fixed ratio increased, and, during the second phase, fixed ratio values were presented in a mixed order. On average, rats obtained more than 60 infusions per session under baseline (fixed ratio 1) during the each of the three phases, with the number of infusions received decreasing progressively with increasing fixed ratio values. Estimates of elasticity across the three phases were not statistically different indicating that the order of fixed ratio presentation did not markedly alter estimates of demand and further demonstrating the robustness of price as a source of control over operant behavior, including behavior maintained by drug reinforcers.


Asunto(s)
Condicionamiento Operante/efectos de los fármacos , Esquema de Refuerzo , Remifentanilo/farmacología , Analgésicos Opioides/farmacología , Animales , Relación Dosis-Respuesta a Droga , Masculino , Ratas , Ratas Sprague-Dawley , Refuerzo en Psicología , Remifentanilo/metabolismo , Autoadministración/métodos
13.
Handb Exp Pharmacol ; 258: 127-145, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31451969

RESUMEN

Despite the therapeutic utility of opioids for relieving pain, other behavioral effects, including their potential for abuse and overdose, can be quite detrimental to individuals as well as society and have contributed to the ongoing opioid crisis. The dramatic escalation in overdose deaths over the last 15 years was initially driven by abuse of prescription opioids, although abuse of heroin, fentanyl, and fentanyl analogs has been increasing, largely due to increased availability and lower cost compared with prescription opioids. All of these opioids share pharmacological properties, acting as agonists at mu opioid receptors, and produce similar behavioral effects, including abuse-related, pain-relieving, dependence-producing, and respiratory-depressant effects. Despite their similarities, opioids are not pharmacologically identical. In fact, drugs that act at mu opioid receptors, including abused opioids, can vary on a number of dimensions, including pharmacological efficacy, drug-receptor interactions, receptor selectivity, and pharmacokinetics. Overall, these differences impact behavioral effects of drugs acting at mu opioid receptors, and this chapter describes variations in those behavioral effects and how these differences continue to provide new strategies that can be developed to address the ongoing opioid epidemic.


Asunto(s)
Analgésicos Opioides/efectos adversos , Analgésicos Opioides/uso terapéutico , Conducta/efectos de los fármacos , Receptores Opioides mu/agonistas , Humanos , Trastornos Relacionados con Opioides
14.
J Pharmacol Exp Ther ; 368(1): 88-99, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30401680

RESUMEN

Opioid abuse remains a serious public health challenge, despite the availability of medications that are effective in some patients (naltrexone, buprenorphine, and methadone). This study explored the potential of a pseudoirreversible mu-opioid receptor antagonist [methocinnamox (MCAM)] as a treatment for opioid abuse by examining its capacity to attenuate the reinforcing effects of mu-opioid receptor agonists in rhesus monkeys. In one experiment, monkeys responded for heroin (n = 5) or cocaine (n = 4) under a fixed-ratio schedule. Another group (n = 3) worked under a choice procedure with one alternative delivering food and the other alternative delivering the mu-opioid receptor agonist remifentanil. A third group (n = 4) responded for food and physiologic parameters were measured via telemetry. The effects of MCAM were determined in all experiments and, in some cases, were compared with those of naltrexone. When given immediately before sessions, naltrexone dose-dependently decreased responding for heroin and decreased choice of remifentanil while increasing choice of food, with responding returning to baseline levels 1 day after naltrexone injection. MCAM also decreased responding for heroin and decreased choice of remifentanil while increasing choice of food; however, opioid-maintained responding remained decreased for several days after treatment. Doses of MCAM that significantly decreased opioid-maintained responding did not decrease responding for cocaine or food. MCAM did not impact heart rate, blood pressure, body temperature, or activity at doses that decreased opioid self-administration. Because MCAM selectively attenuates opioid self-administration for prolonged periods, this novel drug could be a safe and effective alternative to currently available treatments for opioid abuse.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/antagonistas & inhibidores , Cinamatos/uso terapéutico , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Derivados de la Morfina/uso terapéutico , Receptores Opioides mu/antagonistas & inhibidores , Analgésicos Opioides/metabolismo , Animales , Cinamatos/farmacología , Comportamiento de Búsqueda de Drogas/fisiología , Femenino , Macaca mulatta , Masculino , Derivados de la Morfina/farmacología , Naltrexona/farmacología , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/farmacología , Antagonistas de Narcóticos/uso terapéutico , Receptores Opioides mu/metabolismo , Autoadministración , Factores de Tiempo
15.
J Pharmacol Exp Ther ; 368(2): 229-236, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30463875

RESUMEN

One consequence of the ongoing opioid epidemic is a large number of overdose deaths. Naloxone reverses opioid-induced respiratory depression; however, its short duration of action limits the protection it can provide. Methocinnamox (MCAM) is a novel opioid receptor antagonist with a long duration of action. This study examined the ability of MCAM to prevent and reverse the respiratory-depressant effects (minute volume [VE]) of heroin in five monkeys. MCAM (0.32 mg/kg) was given before heroin to determine whether it prevents respiratory depression; heroin dose-effect curves were generated 1, 2, 4, and 8 days later, and these effects were compared with those of naltrexone (0.032 mg/kg). Heroin dose dependently decreased VE MCAM and naltrexone prevented respiratory depression, shifting the heroin dose-effect curve rightward at least 10-fold. MCAM, but not naltrexone, attenuated these effects of heroin for 4 days. MCAM (0.1-0.32 mg/kg) was given 30 minutes after heroin to determine whether it reverses respiratory depression; heroin dose-effect curves were generated 1, 2, 4, 8, and 16 days later, and these effects were compared with those of naloxone (0.0032-0.1 mg/kg). MCAM and naloxone reversed respiratory depression within 30 minutes, although only MCAM antagonized heroin on subsequent days. Thus, MCAM prevents and reverses respiratory depression, the potentially lethal effect of heroin, longer than opioid receptor antagonists currently in use. Because of its sustained effects, MCAM might provide more effective rescue from and protection against the fatal respiratory-depressant effects of opioids, thereby improving treatment of opioid overdose.


Asunto(s)
Cinamatos/uso terapéutico , Heroína/toxicidad , Derivados de la Morfina/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Receptores Opioides mu/antagonistas & inhibidores , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/tratamiento farmacológico , Analgésicos Opioides/toxicidad , Animales , Cinamatos/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Macaca mulatta , Masculino , Derivados de la Morfina/farmacología , Antagonistas de Narcóticos/farmacología , Receptores Opioides mu/fisiología , Insuficiencia Respiratoria/fisiopatología
16.
J Pharmacol Exp Ther ; 371(2): 507-516, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31439807

RESUMEN

A novel µ-opioid receptor antagonist, methocinnamox (MCAM), attenuates some abuse-related and toxic effects of opioids. This study further characterized the pharmacology of MCAM in separate groups of rats using procedures to examine antinociception, gastrointestinal motility, and withdrawal in morphine-dependent rats. Antinociceptive effects of opioid receptor agonists were measured before and after MCAM (1-10 mg/kg) using warm water tail withdrawal and sensitivity to mechanical stimulation in inflamed paws (complete Freund's adjuvant). Before MCAM, morphine, fentanyl, and the κ-opioid receptor agonist spiradoline dose dependently increased tail-withdrawal latency from 50°C water; MCAM attenuated the antinociceptive effects of morphine and fentanyl, but not spiradoline. Morphine increased sensitivity to mechanical stimulation and decreased gastrointestinal motility, and MCAM blocked both effects. These antagonist effects of 10 mg/kg MCAM were persistent, lasting for 2 weeks or longer. Withdrawal emerged after discontinuation of morphine treatment or administration of 10 mg/kg MCAM or 17.8 mg/kg naloxone; other than the day of antagonist administration when withdrawal signs were greater in rats that received antagonist compared with rats that received vehicle, there was no difference among groups in directly observable withdrawal signs or decreased body weight. These results confirm that MCAM is a selective µ-opioid receptor antagonist with an exceptionally long duration of action, likely due to pseudoirreversible binding. Despite its sustained antagonist effects, the duration of withdrawal precipitated by MCAM is not different from that precipitated by naloxone, suggesting that the long duration of antagonism provided by MCAM could be particularly effective for treating opioid abuse and overdose. SIGNIFICANCE STATEMENT: The opioid receptor antagonist MCAM attenuates some abuse-related and toxic effects of opioids. This study demonstrates that MCAM selectively antagonizes multiple effects mediated by µ-opioid receptor agonists for 2 weeks or longer, and like naloxone, MCAM precipitates withdrawal in morphine-dependent rats. Despite this persistent antagonism, withdrawal signs precipitated by MCAM are not significantly different from signs precipitated by naloxone or occurring after discontinuation of morphine, suggesting that using MCAM for opioid abuse or overdose would not produce sustained withdrawal.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Cinamatos/administración & dosificación , Derivados de la Morfina/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Síndrome de Abstinencia a Sustancias/prevención & control , Analgésicos Opioides/efectos adversos , Animales , Cinamatos/metabolismo , Relación Dosis-Respuesta a Droga , Masculino , Derivados de la Morfina/metabolismo , Antagonistas de Narcóticos/metabolismo , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo
17.
Behav Pharmacol ; 30(4): 335-342, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30320606

RESUMEN

Intravenous (i.v.) drug self-administration remains the 'gold standard' for assessing abuse potential of drugs. Failure of a drug to maintain self-administration might indicate merely the absence of positive-reinforcing effects but might also indicate presence of aversive effects. Sensitivity to aversive effects is thought to affect the initiation and maintenance of drug use as well as relapse. Choice procedures are used to study positive-reinforcing effects of drugs and to a much lesser extent to study punishing effects of drugs. Experiment 1 compared the µ-opioid receptor agonist remifentanil (0.001-0.01 mg/kg/infusion), the κ-opioid receptor agonist spiradoline (0.0056-0.056 mg/kg/infusion), and histamine (1.0 mg/kg/infusion) in rats choosing between a food pellet only and an i.v. infusion+a food pellet. To test whether a history with one punishing drug affects the punishing effects of a second drug, experiment 2 compared sensitivity with spiradoline in rats with and without a history of histamine punishment. All rats predominantly chose a pellet alone when histamine+a pellet was the alternative, and they predominantly chose remifentanil+a pellet over a pellet alone. In experiment 2, spiradoline was punishing in rats with a history of histamine punishment but not drug-naive rats. This food choice procedure is sensitive to reinforcing and punishing effects of different drugs in the same subjects, suggesting that the procedure is well-suited for studying drug mixtures (e.g. µ and κ agonists) and the impact of different physiological conditions (e.g. pain) on reinforcement and punishment.


Asunto(s)
Conducta de Elección/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Analgésicos Opioides/farmacología , Animales , Relación Dosis-Respuesta a Droga , Alimentos , Histamina/metabolismo , Histamina/farmacología , Masculino , Castigo/psicología , Pirrolidinas/metabolismo , Pirrolidinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Refuerzo en Psicología , Remifentanilo/metabolismo , Remifentanilo/farmacología , Recompensa , Autoadministración
18.
Behav Pharmacol ; 29(1): 60-70, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28952976

RESUMEN

µ-Opioid receptor agonists are commonly used to treat pain despite their adverse effects. In preclinical studies, cannabinoid receptor agonists increase the potency of opioids for producing antinociceptive but not reinforcing effects. It is unknown whether other adverse effects of these drugs, such as impairment of complex behavior, are enhanced by their co-administration. This study characterized the effects of morphine (µ-opioid receptor agonist; 0.32-5.6 mg/kg, subcutaneously) and CP55940 (CB1/CB2 cannabinoid receptor agonist; 0.0032-0.32 mg/kg, subcutaneously), alone and in mixtures, in monkeys (n=3) choosing between one pellet delivered immediately and two pellets delivered after a delay. Two consecutive choices of the immediate or delayed reward decreased or increased, respectively, the delay. The median adjusted delay, indicating indifference between the immediate and delayed reinforcers, was increased by morphine (3.2 mg/kg) and CP55940 (0.01-0.032 mg/kg). Performance after administration of morphine (0.32 and 1 mg/kg)/CP55940 (0.0032-0.032 mg/kg) mixtures was not different from performance after CP55940 alone. Neither morphine, CP55940, nor mixtures decreased the median adjusted delay (i.e. increased impulsivity). These findings failed to confirm previous studies showing that morphine increases impulsivity, perhaps because of procedural differences among studies. Treatment of pain often requires repeated drug administration; thus, it remains to be determined whether the present findings predict the effects of chronically administered morphine/CP5540 mixtures on impulsive choice.


Asunto(s)
Ciclohexanoles/farmacología , Conducta Impulsiva/efectos de los fármacos , Morfina/farmacología , Analgésicos Opioides/farmacología , Animales , Agonistas de Receptores de Cannabinoides/farmacología , Conducta de Elección/efectos de los fármacos , Ciclohexanoles/metabolismo , Relación Dosis-Respuesta a Droga , Macaca mulatta , Masculino , Morfina/metabolismo , Dolor , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Refuerzo en Psicología
19.
Behav Pharmacol ; 29(8): 676-687, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29595542

RESUMEN

Opioids can enhance delay discounting and premature responding under attentional tasks that might reflect increased impulsivity; although it is not clear whether repeated opioid administration alters behavioral inhibition. Effects of morphine and amphetamine were determined before, during, and after daily morphine administration in rats responding under a stop-signal reaction time task, measuring behavioral inhibition and motor impulsivity. Rats (n=5) completed a two-response sequence to earn food. Occasionally, a tone (stop signal) was presented signifying that food would only be presented if the second response was withheld. Responding after the stop signal measured inhibition, and responding before the start of the trial (premature) measured motor impulsivity. Before daily treatment, morphine (0.32-17.8 mg/kg, intraperitoneally) decreased premature responding but did not increase responding on stop trials, whereas amphetamine (0.1-3.2 mg/kg, intraperitoneally) increased premature responding. Daily morphine administration (3.2 mg/kg/day) enhanced its effects on premature responding but did not impact other effects. Daily morphine treatment diminished the effects of amphetamine on premature and timeout responding. Repeated morphine treatment increased motor impulsivity but did not enhance behavioral inhibition. These data add to studies elucidating the relationship between impulsivity and opioid treatment and suggest that opioids differentially impact impulsive behaviors.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Impulsiva/efectos de los fármacos , Inhibición Psicológica , Morfina/farmacología , Tiempo de Reacción/efectos de los fármacos , Detección de Señal Psicológica/efectos de los fármacos , Análisis de Varianza , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Condicionamiento Operante/efectos de los fármacos , Dextroanfetamina/farmacología , Masculino , Uso Fuera de lo Indicado , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
20.
J Pharmacol Exp Ther ; 360(2): 280-287, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27903642

RESUMEN

Pain is a significant clinical problem, and there is a need for more effective treatments with reduced adverse effects that currently limit the use of µ opioid receptor agonists. Synthetic κ opioid receptor agonists have no abuse liability and well-documented antinociceptive effects; however, adverse effects (diuresis, dysphoria) preclude their use in the clinic. Combining κ opioids with nonopioid drugs (cannabinoid receptor agonists) allows for smaller doses of each drug to produce antinociception. This study tested whether a potentially useful effect of the κ opioid receptor agonist 2-(3,4-dichlorophenyl)-N-methyl-N-[(5R,7S,8S)-7-pyrrolidin-1-yl-1-oxaspiro[4.5]decan-8-yl] (spiradoline; antinociception) is selectively enhanced by the cannabinoid receptor agonist 2-[(1R,2R,5R)-5-hydroxy-2-(3-hydroxypropyl) cyclohexyl]-5-(2-methyloctan-2-yl)phenol (CP55940). Cumulative dose-response functions were determined in eight male Sprague-Dawley rats for spiradoline (0.032-32.0 mg/kg, i.p.) and CP55940 (0.0032-1.0 mg/kg, i.p.) for antinociception, hypothermia, food-maintained responding, and diuresis. Alone, each drug dose dependently increased tail withdrawal latencies from 50°C water, decreased body temperature by ∼4°C, and eliminated food-maintained responding. Spiradoline, but not CP55940, significantly increased urine output at doses that eliminated responding. Smaller doses of spiradoline and CP55940 in mixtures (3:1, 1:1, and 1:3 spiradoline:CP55940) had effects comparable to those observed with larger doses of either drug administered alone: the interaction was additive for antinociception and additive or greater than additive for hypothermia and food-maintained responding. Collectively, these data fail to provide support for the use of these mixtures for treating acute pain; however, κ opioid/cannabinoid mixtures might be useful for treating pain under other conditions (e.g., chronic pain), but only if the adverse effects of both drugs are not enhanced in mixtures.


Asunto(s)
Conducta Animal/efectos de los fármacos , Agonistas de Receptores de Cannabinoides/farmacología , Ciclohexanoles/farmacología , Pirrolidinas/farmacología , Receptores de Cannabinoides/metabolismo , Receptores Opioides kappa/agonistas , Animales , Sinergismo Farmacológico , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA