Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38645066

RESUMO

The treatment landscape for opioid use disorder (OUD) faces challenges stemming from the limited efficacy of existing medications, poor adherence to prescribed regimens, and a heightened risk of fatal overdose post-treatment cessation. Therefore, there is a pressing need for innovative therapeutic strategies that enhance the effectiveness of interventions and the overall well-being of individuals with OUD. This study explored the therapeutic potential of nor-Levo-α-acetylmethadol (nor-LAAM) to treat OUD. We developed sustained release nor-LAAM-loaded poly (lactic-co-glycolic acid) (PLGA) microparticles (MP) using a hydrophobic ion pairing (HIP) approach. The nor-LAAM-MP prepared using HIP with pamoic acid had high drug loading and exhibited minimal initial burst release and sustained release. The nor-LAAM-MP was further optimized for desirable particle size, drug loading, and release kinetics. The lead nor-LAAM-MP (F4) had a relatively high drug loading (11 wt.%) and an average diameter (19 µm) and maintained a sustained drug release for 4 weeks. A single subcutaneous injection of nor-LAAM-MP (F4) provided detectable nor-LAAM levels in rabbit plasma for at least 15 days. We further evaluated the therapeutic efficacy of nor-LAAM-MP (F4) in a well-established fentanyl-addiction rat model, and revealed a marked reduction in fentanyl choice and withdrawal symptoms in fentanyl-dependent rats. These findings provide insights into further developing long-acting nor-LAAM-MP for treating OUD. It has the potential to offer a new effective medication to the existing sparse armamentarium of products available to treat OUD.

2.
J Pharmacol Exp Ther ; 2024 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-38409113

RESUMO

While agonists of mu (MOR) and kappa (KOR) opioid receptors have analgesic effects, they produce euphoria and dysphoria, respectively. Other side effects include respiratory depression and addiction for MOR agonists and sedation for KOR agonists. We reported that 17-cyclopropylmethyl-3,14ß-dihydroxy-4,5α-epoxy-6ß-{[4'-(2'-cyanopyridyl)]carboxamido}cmorphinan (NCP) displayed potent KOR full agonist and MOR partial agonist activities (58%) with 6.5x KOR-over-MOR selectivity in vitro Herein, we characterized pharmacological effects of NCP in rodents. In mice, NCP exerted analgesic effects against inflammatory pain in both the formalin test and the acetic acid writhing test, with A50 values of 47.6 and 14.4 microg/kg (s.c.), respectively. The analgesic effects in the acetic acid writhing test were mediated by the KOR. NCP at doses much higher than those effective in reducing inflammatory pain did not produce antinociception in the hot plate and tail flick tests, inhibit compound 48/80-induced scratching, cause conditioned place aversion (CPA) or preference, impair rotarod performance, inhibit locomotor activity, cause respiratory depression, or precipitate morphine withdrawal. However, NCP (10~100 microg/kg) inhibited gastrointestinal transit with a maximum of ~40% inhibition. In MOR knockout mice, NCP caused CPA, demonstrating that its lack of CPA is due to combined actions on the MOR and KOR. Following s.c. injection, NCP penetrated into the mouse brain. In rats trained to self-administer heroin, NCP (1~320 microg/kg/infusion) did not function as a reinforcer. Thus, NCP produces potent analgesic effects via KOR without side effects except constipation. Therefore, dual full KOR/partial MOR agonists with moderate KOR-over-MOR selectivity may be promising as non-addictive analgesics for inflammatory pain. Significance Statement Developing non-addictive analgesics is crucial for reducing opioid overdose deaths, minimizing drug misuse, and promoting safer pain management practices. Herein, pharmacology of a potential non-addictive analgesic, NCP, is reported. NCP has full KOR agonist / partial MOR agonist activities with a 6.5 x selectivity for KOR over MOR. Unlike MOR agonists, analgesic doses of NCP do not lead to self-administration or respiratory depression. Furthermore, NCP does not produce aversion, hypolocomotion, or motor incoordination, side effects typically associated with KOR activation.

3.
Psychopharmacology (Berl) ; 241(2): 305-314, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37870564

RESUMO

RATIONALE: Combinations of mu and kappa-opioid receptor (KOR) agonists have been proposed as analgesic formulations with reduced abuse potential. The feasibility of this approach has been increased by the development of KOR agonists with biased signaling profiles that produce KOR-typical antinociception with fewer KOR-typical side effects. OBJECTIVE: The present study determined if the biased KOR agonists, nalfurafine and triazole 1.1, could reduce choice for oxycodone in rhesus monkeys as effectively as the typical KOR agonist, salvinorin A. METHODS: Adult male rhesus monkeys (N = 5) responded under a concurrent schedule of food delivery and intravenous cocaine injections (0.018 mg/kg/injection). Once trained, cocaine (0.018 mg/kg/injection) or oxycodone (0.0056 mg/kg/injection) was tested alone or in combination with contingent injections of salvinorin A (0.1-3.2 µg/kg/injection), nalfurafine (0.0032-0.1 µg/kg/injection), triazole 1.1 (3.2-100.0 µg/kg/injection), or vehicle. In each condition, the cocaine or oxycodone dose, as well as the food amount, was held constant across choice components, while the dose of the KOR agonist was increased across choice components. RESULTS: Cocaine and oxycodone were chosen over food on more than 80% of trials when administered alone or contingently with vehicle. When KOR agonists were administered contingently with either cocaine or oxycodone, drug choice decreased in a dose-dependent manner. Salvinorin A and triazole 1.1 decreased drug-reinforcer choice without altering total trials completed (i.e., choice allocation shifted to food), while nalfurafine dose dependently decreased total trials completed. CONCLUSIONS: These results demonstrate that salvinorin A and triazole 1.1, but not nalfurafine, selectively reduce cocaine and oxycodone self-administration independent of nonspecific effects on behavior, suggesting that G-protein bias does not appear to be a moderating factor in this outcome. Triazole 1.1 represents an important prototypical compound for developing novel KOR agonists as deterrents for prescription opioid abuse.


Assuntos
Cocaína , Diterpenos Clerodânicos , Morfinanos , Oxicodona , Compostos de Espiro , Animais , Masculino , Oxicodona/farmacologia , Analgésicos Opioides/farmacologia , Macaca mulatta , Preparações Farmacêuticas , Autoadministração , Cocaína/farmacologia , Triazóis , Receptores Opioides kappa/agonistas , Relação Dose-Resposta a Droga
4.
ACS Chem Neurosci ; 13(24): 3608-3628, 2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36449691

RESUMO

Discovery of analgesics void of abuse liability is critical to battle the opioid crisis in the United States. Among many strategies to achieve this goal, targeting more than one opioid receptor seems promising to minimize this unwanted side effect while achieving a reasonable therapeutic profile. In the process of understanding the structure-activity relationship of nalfurafine, we identified a potential analgesic agent, NMF, as a dual kappa opioid receptor/delta opioid receptor agonist with minimum abuse liability. Further characterizations, including primary in vitro ADMET studies (hERG toxicity, plasma protein binding, permeability, and hepatic metabolism), and in vivo pharmacodynamic and toxicity profiling (time course, abuse liability, tolerance, withdrawal, respiratory depression, body weight, and locomotor activity) further confirmed NMF as a promising drug candidate for future development.


Assuntos
Analgésicos Opioides , Morfinanos , Humanos , Analgésicos Opioides/química , Receptores Opioides kappa/agonistas , Morfinanos/farmacologia , Analgésicos/farmacologia , Relação Estrutura-Atividade , Receptores Opioides mu/agonistas
5.
Pharmacol Biochem Behav ; 221: 173473, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36228740

RESUMO

Recent studies have shown that social interaction can serve as an alternative reinforcer to opioid self-administration under a choice context in rats. However, additional parametric studies are needed to evaluate the sensitivity of opioid-vs.-social interaction procedures relative to more established opioid-vs.-food procedures. The current study evaluated the sensitivity of a novel fentanyl-vs.-social interaction choice procedure to environmental and pharmacological manipulations previously shown to affect fentanyl-vs.-food choice. Male and female rats (responder rats; n = 6/sex) were trained to respond in a discrete-trial choice procedure for either 30-s access to a same-sex "partner" rat or an intravenous fentanyl infusion. Once trained, the effects of fentanyl unit dose (0, 0.32-10 µg/kg/inf), partner rat presence, opioid-dependence status, chronic naltrexone administration (0.032, 0.1 mg/kg/h), and response requirement for fentanyl self-administration (fixed ratio 1-320) were determined across weeks. The fentanyl-vs.-social interaction choice procedure was sensitive to the unit dose of fentanyl, chronic naltrexone treatment, and fentanyl response requirement. However, the magnitude of these effects on fentanyl choice was smaller than those reported in published fentanyl-vs.-food choice studies. Furthermore, fentanyl-vs.-social interaction choice was not sensitive to removal of the partner rat or opioid-dependence status. Minimal sex differences were detected. These results suggest that this fentanyl-vs.-social interaction choice procedure is less sensitive to environmental and pharmacological interventions than previously established opioid-vs.-food choice procedures. The observed discrepancy in sensitivity between the procedures suggests that social interaction may have qualitatively different reinforcing properties compared to more commonly assessed alternative reinforcers such as food (preclinical) or money (human laboratory).


Assuntos
Fentanila , Transtornos Relacionados ao Uso de Opioides , Humanos , Feminino , Masculino , Ratos , Animais , Fentanila/farmacologia , Naltrexona/farmacologia , Analgésicos Opioides/farmacologia , Interação Social , Autoadministração , Relação Dose-Resposta a Droga
6.
Neuropsychopharmacology ; 47(12): 2132-2139, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35906489

RESUMO

The high efficacy mu-opioid receptor (MOR) agonist methadone is an effective opioid use disorder (OUD) medication used exclusively in opioid-dependent patients. However, methadone has undesirable effects that limit its clinical efficacy. Intermediate efficacy MOR agonists may treat OUD with fewer undesirable effects. We compared the effects of methadone with the intermediate efficacy MOR agonist TRV130 (oliceridine) on fentanyl-vs.-food choice and somatic withdrawal signs in opioid-dependent and post-opioid-dependent rats. Male rats (n = 20) were trained under a fentanyl-vs.-food choice procedure. Rats were then provided extended fentanyl (3.2 µg/kg/infusion) access (6 p.m.-6 a.m.) for 10 days to produce opioid dependence/withdrawal. Rats were treated with vehicle (n = 7), TRV130 (3.2 mg/kg; n = 8), or methadone (3.2 mg/kg; n = 5) three times per day after each extended-access session (8:30 a.m., 11 a.m., 1:30 p.m.). Withdrawal sign scoring (1:55 p.m.) and choice tests (2-4 p.m.) were conducted daily. Vehicle, TRV130, and methadone effects on fentanyl choice were redetermined in post-opioid-dependent rats. Vehicle-, TRV130-, and methadone-treated rats had similar fentanyl intakes during extended access. Vehicle-treated rats exhibited increased withdrawal signs and decreased bodyweights. Both methadone and TRV130 decreased these withdrawal signs. TRV130 was less effective than methadone to decrease fentanyl choice and increase food choice in opioid-dependent rats. Neither methadone nor TRV130 decreased fentanyl choice in post-opioid-dependent rats. Results suggest that higher MOR activation is required to reduce fentanyl choice than withdrawal signs in fentanyl-dependent rats. Additionally, given that TRV130 did not precipitate withdrawal in opioid-dependent rats, intermediate efficacy MOR agonists like TRV130 may facilitate the transition of patients with OUD from methadone to lower efficacy treatments like buprenorphine.


Assuntos
Buprenorfina , Transtornos Relacionados ao Uso de Opioides , Síndrome de Abstinência a Substâncias , Analgésicos Opioides , Animais , Buprenorfina/farmacologia , Fentanila/farmacologia , Masculino , Metadona/farmacologia , Metadona/uso terapêutico , Entorpecentes , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Ratos , Receptores Opioides , Compostos de Espiro , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Tiofenos
7.
Br J Pharmacol ; 179(2): 227-241, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34505281

RESUMO

BACKGROUND AND PURPOSE: Maintenance treatment with opioid agonists (buprenorphine, methadone) decreases opioid use and relapse. We recently modelled maintenance treatment in rats and found that chronic delivery of buprenorphine or the µ opioid receptor partial agonist TRV130 decreased relapse to oxycodone seeking and taking. Here, we tested the buprenorphine analogue BU08028 on different heroin relapse-related measures and heroin versus food choice. EXPERIMENTAL APPROACH: For relapse assessment, we trained male and female rats to self-administer heroin (6 h·day-1 , 14 days) in Context A and then implanted osmotic minipumps containing BU08028 (0, 0.03 or 0.1 mg·kg-1 ·d-1 ). Effects of chronic BU08028 delivery were tested on (1) incubation of heroin-seeking in a non-drug Context B, (2) extinction responding reinforced by heroin-associated discrete cues in Context B, (3) reinstatement of heroin-seeking induced by re-exposure to Context A and (4) re-acquisition of heroin self-administration in Context A. For choice assessment, we tested the effect of chronic BU08028 delivery on heroin versus food choice. KEY RESULTS: Chronic BU08028 delivery decreased incubation of heroin seeking. Unexpectedly, BU08028 increased re-acquisition of heroin self-administration selectively in females. Chronic BU08028 had minimal effects on context-induced reinstatement and heroin versus food choice in both sexes. Finally, exploratory post hoc analyses suggest that BU08028 decreased extinction responding selectively in males. CONCLUSIONS AND IMPLICATIONS: Chronic BU08028 delivery had both beneficial and detrimental, sex-dependent, effects on different triggers of heroin relapse and minimal effects on heroin choice in both sexes. Results suggest that BU08028 would not be an effective opioid maintenance treatment in humans.


Assuntos
Buprenorfina , Heroína , Analgésicos Opioides/farmacologia , Animais , Buprenorfina/análogos & derivados , Buprenorfina/farmacologia , Extinção Psicológica , Feminino , Masculino , Ratos , Recidiva , Autoadministração , Caracteres Sexuais
8.
Biol Psychiatry Glob Open Sci ; 1(2): 112-122, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34458885

RESUMO

BACKGROUND: Opioid withdrawal is a key driver of opioid addiction and an obstacle to recovery. However, withdrawal effects on opioid reinforcement and mesolimbic neuroadaptation are understudied and the role of sex is largely unknown. METHODS: Male (n=13) and female (n=12) rats responded under a fentanyl-vs.-food "choice" procedure during daily 2h sessions. In addition to the daily choice sessions, rats were provided extended access to fentanyl during 12h self-administration sessions. After two weeks of this self-administration regimen, the nucleus accumbens (NAc) and ventral tegmental area (VTA) of a subset of rats were subjected to RNA sequencing. In the remaining rats, a third week of this self-administration regimen was conducted, during which methadone effects on fentanyl-vs.-food choice were determined. RESULTS: Prior to opioid dependence, male and female rats similarly allocated responding between fentanyl and food. Abstinence from extended fentanyl access elicited similar increases in somatic withdrawal signs in both sexes. Despite similar withdrawal signs and extended access fentanyl intake, opioid withdrawal was accompanied by a maladaptive increase in fentanyl choice in males, but not females. Behavioral sex differences corresponded with a greater number of differentially expressed genes in the NAc and VTA of opioid-withdrawn females relative to males. Methadone blocked withdrawal-associated increases in fentanyl choice in males, but failed to further decrease fentanyl choice in females. CONCLUSIONS: These results provide foundational evidence of sex-specific neuroadaptations to opioid withdrawal, which may be relevant to the female-specific resilience to withdrawal-associated increases in opioid choice and aid in the identification of novel therapeutic targets.

9.
Psychopharmacology (Berl) ; 238(7): 1885-1897, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33765177

RESUMO

RATIONALE AND OBJECTIVE: Pain-related factors increase the risk for opioid addiction, and pain may function as a negative reinforcer to increase opioid taking and seeking. However, experimental pain-related manipulations generally do not increase opioid self-administration in rodents. This discrepancy may reflect insufficient learning of pain-relief contingencies or confounding effects of pain-related behavioral impairments. Here, we determined if pairing noxious stimuli with opioid self-administration would promote pain-related reinstatement of opioid seeking or increase opioid choice over food. METHODS: In Experiment 1, rats self-administered fentanyl in the presence or absence of repeated intraplantar capsaicin injections in distinct contexts to model context-specific exposure to cutaneous nociception. After capsaicin-free extinction in both contexts, we tested if capsaicin would reinstate fentanyl seeking. In Experiment 2, rats self-administered heroin after intraperitoneal (i.p.) lactic acid injections to model acute visceral inflammatory pain. After lactic acid-free extinction, we tested if lactic acid would reinstate heroin seeking. In Experiment 3, we tested if repeated i.p. lactic acid or intraplantar Complete Freund's Adjuvant (CFA; to model sustained inflammatory pain) would increase fentanyl choice over food. RESULTS: In Experiments 1-2, neither capsaicin nor lactic acid reinstated opioid seeking after extinction, and lactic acid did not increase heroin-induced reinstatement. In Experiment 3, lactic acid and CFA decreased reinforcement rate without affecting fentanyl choice. CONCLUSIONS: Results extend the range of conditions across which pain-related manipulations fail to increase opioid seeking in rats and suggest that enhanced opioid-addiction risk in humans with chronic pain involves factors other than enhanced opioid reinforcement and relapse.


Assuntos
Analgésicos Opioides/administração & dosagem , Comportamento de Escolha/efeitos dos fármacos , Comportamento de Procura de Droga/efeitos dos fármacos , Medição da Dor/psicologia , Dor/psicologia , Reforço Psicológico , Animais , Comportamento de Escolha/fisiologia , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Comportamento de Procura de Droga/fisiologia , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/fisiologia , Feminino , Fentanila/farmacologia , Masculino , Transtornos Relacionados ao Uso de Opioides/psicologia , Dor/tratamento farmacológico , Medição da Dor/métodos , Ratos , Autoadministração/métodos
10.
J Neurosci Methods ; 354: 109110, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33705855

RESUMO

BACKGROUND: Preclinical drug self-administration procedures are commonly used to investigate expression, mechanisms, and treatment of substance use disorders. NEW METHOD: The aims were to back-translate an intravenous drug-vs-food choice procedure primarily utilized in monkeys to male and female rats and to develop a surgical method for sustained intravenous catheter patency suitable for long-term drug-choice studies. RESULTS: The surgical protocol resulted in a median intravenous jugular catheter patency in male and female rats of 126 days (range: 25-365 days). Drug-vs-food choice was established with opioids (fentanyl and heroin), psychostimulants (cocaine, methamphetamine, and amphetamine), and an opioid/psychostimulant mixture (fentanyl + methamphetamine). The average time from catheter implantation to stable choice behavior across all drugs was 27 sessions (range: 16-44 sessions). Choice behavior stabilized more quickly for cocaine and fentanyl than for other drugs. Manipulations of both environmental variables (e.g., response requirement or food reinforcer magnitude) and pharmacological variables (e.g., extended access drug self-administration or continuous buprenorphine treatment via osmotic pump) significantly shifted opioid-vs-food choice consistent with previous monkey studies. COMPARISON WITH EXISTING METHODS: Duration of intravenous catheter patency in rats was suitable for long-term, within-subject drug choice studies. Effects of environmental and pharmacological manipulations in rats confirmed and extended previous results from monkeys. CONCLUSIONS: The concordance of behavioral results between rats and monkeys using the present drug-vs-food choice procedure supports its utility to improve our basic understanding of the expression and mechanisms of substance use disorders towards to development of more effective therapeutics.


Assuntos
Cocaína , Preparações Farmacêuticas , Transtornos Relacionados ao Uso de Substâncias , Animais , Comportamento de Escolha , Relação Dose-Resposta a Droga , Feminino , Preferências Alimentares , Masculino , Ratos , Autoadministração , Transtornos Relacionados ao Uso de Substâncias/etiologia
11.
Psychopharmacology (Berl) ; 238(4): 1017-1028, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33404739

RESUMO

RATIONALE: Strategies are needed to decrease the abuse liability of mu opioid receptor (MOR) agonists. One strategy under consideration is to combine MOR agonists with kappa opioid receptor (KOR) agonists. OBJECTIVES: The effects of KOR agonists (U50488, nalfurafine) on fentanyl-vs.-food choice were compared under conditions where the KOR agonists were added to the intravenously self-administered fentanyl (contingent delivery) or administered as subcutaneous pretreatments (non-contingent delivery) in male and female rats. METHODS: Rats were trained to respond under a concurrent schedule of fentanyl (0, 0.32-10 µg/kg/infusion) and food reinforcement. In experiment 1, U50488 and nalfurafine were co-administered with fentanyl as fixed-proportion mixtures (contingent administration). In experiment 2, U50488 (1-10 mg/kg) and nalfurafine (3.2-32 µg/kg) were administered as acute pretreatments (non-contingent administration). The selective KOR antagonist, nor-BNI (32 mg/kg), was administered prior to contingent and non-contingent KOR-agonist treatment in experiment 3. RESULTS: Both U50488 and nalfurafine decreased fentanyl choice when administered contingently, demonstrating that KOR agonists punish opioid choice. However, evidence for punishment corresponded with an elimination of operant responding in the majority of rats. Non-contingent U50488 and nalfurafine administration only decreased the number of choices made during the behavioral session without altering fentanyl choice. Contingent and non-contingent KOR-agonist effects on fentanyl choice were both attenuated by nor-BNI. CONCLUSIONS: These results illustrate that the effects of KOR agonists on fentanyl reinforcement are dependent upon the contingencies under which they are administered.


Assuntos
Analgésicos Opioides/farmacologia , Fentanila/farmacologia , Receptores Opioides kappa/agonistas , Reforço Psicológico , Animais , Relação Dose-Resposta a Droga , Feminino , Masculino , Morfinanos/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Sprague-Dawley , Compostos de Espiro/farmacologia
12.
Artigo em Inglês | MEDLINE | ID: mdl-31932466

RESUMO

This review describes methods for preclinical evaluation of candidate medications to treat opioid use disorder (OUD). The review is founded on the propositions that (1) drug self-administration procedures provide the most direct method for assessment of medication effectiveness, (2) procedures that assess choice between opioid and nondrug reinforcers are especially useful, and (3) states of opioid dependence and withdrawal profoundly influence both opioid reinforcement and effects of candidate medications. Effects of opioid medications and vaccines on opioid choice in nondependent and opioid-dependent subjects are reviewed. Various nonopioid medications have also been examined, but none yet have been identified that safely and reliably reduce opioid choice. Future research will focus on (1) strategies for increasing safety and/or effectiveness of opioid medications (e.g., G-protein-biased µ-opioid agonists), and (2) continued development of nonopioid medications (e.g., clonidine) that might serve as adjunctive agents to current opioid medications.


Assuntos
Analgésicos Opioides/agonistas , Analgésicos Opioides/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Entorpecentes/agonistas , Entorpecentes/uso terapêutico , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Síndrome de Abstinência a Substâncias/tratamento farmacológico , Analgésicos Opioides/administração & dosagem , Buprenorfina/uso terapêutico , Comportamento de Escolha , Desenvolvimento de Medicamentos , Medicina Baseada em Evidências , Humanos , Metadona/uso terapêutico , Naltrexona/uso terapêutico , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Entorpecentes/administração & dosagem , Transtornos Relacionados ao Uso de Opioides/etiologia , Receptores Opioides mu/efeitos dos fármacos , Autoadministração , Síndrome de Abstinência a Substâncias/prevenção & controle , Resultado do Tratamento
13.
Drug Alcohol Depend ; 218: 108348, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33268227

RESUMO

AIM: Opioid-targeted vaccines are under consideration as candidate Opioid Use Disorder medications. We recently reported that a fentanyl-targeted vaccine produced a robust and long-lasting attenuation of fentanyl-vs-food choice in rats. In the current study, we evaluated an optimized fentanyl-targeted vaccine in rhesus monkeys to determine whether vaccine effectiveness to attenuate fentanyl choice translated to a species with greater phylogenetic similarity to humans. METHODS: Adult male (2) and female (3) rhesus monkeys were trained to respond under a concurrent schedule of food (1 g pellets) and intravenous fentanyl (0, 0.032-1 µg/kg/injection) reinforcement during daily 2 h sessions. Fentanyl choice dose-effect functions were determined daily and 7-day buprenorphine treatments (0.0032-0.032 mg/kg/h IV; n = 4-5) were determined for comparison to vaccine effects. Subsequently, a fentanyl-CRM197 conjugate vaccine was administered at week 0, 3, 8, 15 over a 29-week experimental period during which fentanyl choice dose-effect functions continued to be determined daily. RESULTS: Buprenorphine significantly decreased fentanyl choice and reciprocally increased food choice. Vaccination eliminated fentanyl choice and increased food choice in four-of-the-five monkeys. A transient and less robust vaccine effect was observed in the fifth monkey. Fentanyl-specific antibody concentrations peaked after the third vaccination to approximately 50 µg/mL while anti-fentanyl antibody affinity increased to a sustained low nanomolar level. CONCLUSION: These results translate fentanyl vaccine effectiveness from rats to rhesus monkeys to decrease fentanyl-vs-food choice, albeit with greater individual differences observed in monkeys. These results support the potential and further clinical evaluation of this fentanyl-targeted vaccine as a candidate Opioid Use Disorder medication.


Assuntos
Analgésicos Opioides/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Fentanila/farmacologia , Vacinas , Animais , Comportamento de Escolha/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Alimentos , Macaca mulatta , Masculino , Transtornos Relacionados ao Uso de Opioides , Filogenia , Ratos , Reforço Psicológico , Autoadministração
15.
ACS Chem Neurosci ; 11(9): 1300-1310, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32271538

RESUMO

Opioid-targeted vaccines represent an emerging treatment strategy for opioid use disorder. To determine whether concurrent vaccination against two commonly abused opioids (fentanyl and heroin) would confer broader spectrum opioid coverage, the current study evaluated dual fentanyl/heroin conjugate vaccine effectiveness using a warm water tail-withdrawal and a fentanyl/heroin-vs-food choice procedure in male and female rats across a 105-day observation period. Vaccine administration generated titers of high-affinity antibodies to both fentanyl and heroin sufficient to decrease the antinociceptive potency of fentanyl (25-fold), heroin (4.6-fold), and a 1:27 fentanyl/heroin mixture (7.5-fold). Vaccination did not alter the antinociceptive potency of the structurally dissimilar opioid agonist methadone. For comparison, continuous treatment with a naltrexone dose (0.032 mg/kg/h) shown previously to produce clinically relevant plasma-naltrexone levels decreased the antinociceptive potency of fentanyl, heroin, and the 1:27 fentanyl/heroin mixture by approximately 20-fold. Naltrexone treatment also shifted the potency of 1:27 fentanyl/heroin mixture in a drug-vs-food choice self-administration procedure 4.3-fold. In contrast, vaccination did not attenuate 1:27 fentanyl/heroin mixture self-administration in the drug-vs-food choice procedure. These data demonstrate that a vaccine can simultaneously attenuate the thermal antinociceptive effects of two structurally dissimilar opioids. However, the vaccine did not attenuate fentanyl/heroin mixture self-administration, suggesting a greater magnitude of vaccine responsiveness is required to decrease opioid reinforcement relative to antinociception.


Assuntos
Fentanila , Vacinas , Analgésicos Opioides , Animais , Relação Dose-Resposta a Droga , Feminino , Heroína , Masculino , Naltrexona/farmacologia , Ratos
16.
CNS Drugs ; 34(5): 449-461, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32248427

RESUMO

The ongoing opioid crisis, now into its second decade, represents a global public health challenge. Moreover, the opioid crisis has manifested despite clinical access to three approved opioid use disorder medications: the full opioid agonist methadone, the partial opioid agonist buprenorphine, and the opioid antagonist naltrexone. Although current opioid use disorder medications are underutilized, the ongoing opioid crisis has also identified the need for basic research to develop both safer and more effective opioid use disorder medications. Emerging preclinical evidence suggests that opioid-targeted vaccines or immunopharmacotherapies may be promising opioid use disorder therapeutics. One premise for this article is to critically examine whether vaccine effectiveness evaluated using preclinical antinociceptive endpoints is predictive of vaccine effectiveness on abuse-related endpoints such as drug self-administration, drug discrimination, and conditioned place preference. A second premise is to apply decades of knowledge in the preclinical evaluation of candidate small-molecule therapeutics for opioid use disorder to the preclinical evaluation of candidate opioid use disorder immunopharmacotherapies. We conclude with preclinical experimental design attributes to enhance preclinical-to-clinical translatability and potential future directions for immunopharmacotherapies to address the dynamic illicit opioid environment.


Assuntos
Imunoterapia/métodos , Transtornos Relacionados ao Uso de Opioides/terapia , Vacinas/administração & dosagem , Animais , Saúde Global , Humanos , Epidemia de Opioides/prevenção & controle , Transtornos Relacionados ao Uso de Opioides/imunologia , Vacinas/imunologia
17.
Psychopharmacology (Berl) ; 237(5): 1471-1480, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32006048

RESUMO

RATIONALE: Combinations of mu and kappa opioid receptor (KOR) agonists have been proposed as potential analgesic formulations with reduced abuse liability. The current studies extend previous work by investigating the typical KOR agonist, salvinorin A, and the atypical KOR agonist, nalfurafine, as deterrents of oxycodone self-administration using a progressive ratio (PR) schedule of reinforcement. METHODS: In separate experiments, adult male rhesus monkeys (N = 4/experiment) were trained under a PR schedule of reinforcement to self-administer cocaine (0.1 mg/kg/injection) and saline on alternating days. Oxycodone (0.01-0.1 mg/kg/injection) alone and combined with salvinorin A (experiment 1; 0.006, 0.012 mg/kg/injection) or nalfurafine (experiment 2; 0.0001-0.00032 mg/kg/injection) were tested within the alternating cocaine and saline baseline. The mechanism of nalfurafine's effects on oxycodone self-administration was investigated via pretreatment with the KOR antagonist, nor-binaltorphimine (nor-BNI; 10 mg/kg; i.m.). RESULTS: All subjects self-administered oxycodone alone above saline levels at sufficiently large doses, and combining salvinorin A or nalfurafine with oxycodone reduced the mean number of injections per session to saline levels (experiment 1) or to levels that were significantly lower than oxycodone alone (experiment 2). The ability of nalfurafine to reduce oxycodone self-administration was reversed by pretreatment with nor-BNI. CONCLUSIONS: These results demonstrate that KOR agonists, including the clinically used KOR agonist, nalfurafine, can punish self-administration of a prescription opioid analgesic, oxycodone, in rhesus monkeys and that nalfurafine's punishing effect is KOR-dependent. Combinations of KOR agonists with prescription opioids may have reduced abuse liability.


Assuntos
Analgésicos Opioides/administração & dosagem , Comportamento Aditivo/tratamento farmacológico , Diterpenos Clerodânicos/administração & dosagem , Morfinanos/administração & dosagem , Oxicodona/administração & dosagem , Receptores Opioides kappa/agonistas , Compostos de Espiro/administração & dosagem , Animais , Comportamento Aditivo/psicologia , Cocaína/administração & dosagem , Relação Dose-Resposta a Droga , Macaca mulatta , Masculino , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/administração & dosagem , Reforço Psicológico , Autoadministração
18.
Drug Alcohol Depend ; 208: 107848, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31982193

RESUMO

BACKGROUND: The current opioid crisis has reinvigorated preclinical research in the evaluation of non-opioid candidate treatments for opioid use disorder (OUD). Emerging evidence suggests 5-HT2C receptor agonists may attenuate the abuse-related effects of opioids. This study evaluated effectiveness of 7-day treatment with the clinically available 5-HT2C agonist lorcaserin (Belviq®) on heroin-vs.-food choice in rhesus monkeys. Lorcaserin effects were compared to effects produced by 7-day saline substitution and by 7-day treatment with the opioid antagonist naltrexone. METHODS: Adult male (1) and female (6) rhesus monkeys were trained to respond under a concurrent schedule of food delivery (1 g pellets, fixed-ratio 100 schedule) and intravenous heroin injections (0-0.032 mg/kg/injection, fixed-ratio 10 schedule) during daily 2 h sessions. Heroin choice dose-effect functions were determined daily before and following 7-day saline substitution or 7-day continuous treatment with naltrexone (0.0032-0.032 mg/kg/h, IV) or lorcaserin (0.032-0.32 mg/kg/h, IV). RESULTS: Under baseline conditions, increasing heroin doses maintained a dose-dependent increase in heroin choice. Both saline substitution and 7-day naltrexone treatment significantly attenuated heroin choice and produced a reciprocal increase in food choice. Continuous lorcaserin (0.32 mg/kg/h) treatment significantly increased heroin choice. CONCLUSIONS: In contrast to saline substitution and naltrexone, lorcaserin treatment was ineffective to reduce heroin-vs.-food choice. These preclinical results do not support the therapeutic potential and continued evaluation of lorcaserin as a candidate OUD treatment.


Assuntos
Analgésicos Opioides/administração & dosagem , Benzazepinas/administração & dosagem , Comportamento de Escolha/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/psicologia , Heroína/administração & dosagem , Animais , Comportamento de Escolha/fisiologia , Relação Dose-Resposta a Droga , Comportamento Alimentar/fisiologia , Feminino , Macaca mulatta , Masculino , Autoadministração
19.
Drug Alcohol Depend ; 204: 107501, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31479865

RESUMO

BACKGROUND: One emerging strategy to address the opioid crisis includes opioid-targeted immunopharmacotherapies. This study compared effectiveness of a heroin-tetanus toxoid (TT) conjugate vaccine to antagonize heroin, 6-acetylmorphine (6-AM), morphine, and fentanyl antinociception in rats. METHODS: Adult male and female Sprague Dawley rats received three doses of active or control vaccine at weeks 0, 2, and 4. Vaccine pharmacological selectivity was assessed by comparing opioid dose-effect curves in 50 °C warm-water tail-withdrawal procedure before and after active or control heroin-TT vaccine. Route of heroin administration [subcutaneous (SC) vs. intravenous [IV)] was also examined as a determinant of vaccine effectiveness. Continuous naltrexone treatment (0.0032-0.032 mg/kg/h) effects on heroin, 6-AM, and morphine antinociceptive potency were also determined as a benchmark for minimal vaccine effectiveness. RESULTS: The heroin-TT vaccine decreased potency of SC heroin (5-fold), IV heroin (3-fold), and IV 6-AM (3-fold) for several weeks without affecting IV morphine or SC and IV fentanyl potency. The control vaccine did not alter potency of any opioid. Naltrexone dose-dependently decreased antinociceptive potency of SC heroin, and treatment with 0.01 mg/kg/h naltrexone produced similar, approximate 8-fold decreases in potencies of SC and IV heroin, IV 6-AM, and IV morphine. The combination of naltrexone and active vaccine was more effective than naltrexone alone to antagonize SC heroin but not IV heroin. CONCLUSIONS: The heroin-TT vaccine formulation examined is less effective, but more selective, than chronic naltrexone to attenuate heroin antinociception in rats. Furthermore, these results provide an empirical framework for future preclinical opioid vaccine research to benchmark effectiveness against naltrexone.


Assuntos
Fentanila/antagonistas & inibidores , Heroína/antagonistas & inibidores , Heroína/farmacologia , Derivados da Morfina/antagonistas & inibidores , Morfina/antagonistas & inibidores , Naltrexona/farmacologia , Vacinação , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Feminino , Fentanila/farmacologia , Masculino , Morfina/farmacologia , Derivados da Morfina/farmacologia , Antagonistas de Entorpecentes/farmacologia , Ratos , Toxoide Tetânico/farmacologia
20.
Neuropsychopharmacology ; 44(10): 1681-1689, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31043682

RESUMO

The current opioid crisis remains a significant public health issue and there is a critical need for biomedical research to develop effective and easily deployable candidate treatments. One emerging treatment strategy for opioid use disorder includes immunopharmacotherapies or opioid-targeted vaccines. The present study determined the effectiveness of a fentanyl-tetanus toxoid conjugate vaccine to alter fentanyl self-administration using a fentanyl-vs.-food choice procedure in male and female rats under three experimental conditions. For comparison, continuous 7-day naltrexone (0.01-0.1 mg/kg/h) and 7-day clonidine (3.2-10 µg/kg/h) treatment effects were also determined on fentanyl-vs.-food choice. Male and female rats responded for concurrently available 18% diluted Ensure® (liquid food) and fentanyl (0-10 µg/kg/infusion) infusions during daily sessions. Under baseline and saline treatment conditions, fentanyl maintained a dose-dependent increase in fentanyl-vs.-food choice. First, fentanyl vaccine administration significantly blunted fentanyl reinforcement and increased food reinforcement for 15 weeks in non-opioid dependent rats. Second, surmountability experiments by increasing the unit fentanyl dose available during the self-administration session 10-fold empirically determined that the fentanyl vaccine produced an approximate 22-fold potency shift in fentanyl-vs.-food choice that was as effective as the clinically approved treatment naltrexone. Clonidine treatment significantly increased fentanyl-vs.-food choice. Lastly, fentanyl vaccine administration prevented the expression of withdrawal-associated increases in fentanyl-vs.-food choice following introduction of extended 12 h fentanyl access sessions. Overall, these results support the potential and further consideration of immunopharmacotherapies as candidate treatments to address the current opioid crisis.


Assuntos
Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/imunologia , Comportamento Animal/efeitos dos fármacos , Comportamento de Escolha/efeitos dos fármacos , Comportamento de Procura de Droga/efeitos dos fármacos , Fentanila/administração & dosagem , Fentanila/imunologia , Vacinas Conjugadas/farmacologia , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Clonidina/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Feminino , Alimentos , Masculino , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Transtornos Relacionados ao Uso de Opioides , Ratos , Reforço Psicológico , Autoadministração , Síndrome de Abstinência a Substâncias/fisiopatologia , Toxoide Tetânico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...