Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 18(8): e0277718, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37607205

RESUMO

Riluzole is the only treatment known to improve survival in patients with Amyotrophic Lateral Sclerosis (ALS). However, oral riluzole efficacy is modest at best, further it is known to have large inter-individual variability of serum concentration and clearance, is formulated as an oral drug in a patient population plagued with dysphagia, and has known systemic side-effects like asthenia (limiting patient compliance) and elevated liver enzymes. In this context, we postulated that continuous intrathecal (IT) infusion of low doses of riluzole could provide consistent elevations of the drug spinal cord (SC) concentrations at or above those achieved with oral dosing, without increasing the risk for adverse events associated with systemic drug exposure or off-target side effects in the brain. We developed a formulation of riluzole for IT delivery and conducted our studies in purpose-bred hound dogs. Our non-GLP studies revealed that IT infusion alone was able to increase SC concentrations above those provided by oral administration, without increasing plasma concentrations. We then conducted two GLP studies that combined IT infusion with oral administration at human equivalent dose, to evaluate SC and brain concentrations of riluzole along with assessments of safety and tolerability. In the 6-week study, the highest IT dose (0.2 mg/hr) was well tolerated by the animals and increased SC concentrations above those achieved with oral riluzole alone, without increasing brain concentrations. In the 6-month study, the highest dose tested (0.4 mg/hr) was not tolerated and yielded SC significantly above those achieved in all previous studies. Our data show the feasibility and safety profile of continuous IT riluzole delivery to the spinal cord, without concurrent elevated liver enzymes, and minimal brain concentrations creating another potential therapeutic route of delivery to be used in isolation or in combination with other therapeutics."


Assuntos
Esclerose Amiotrófica Lateral , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Humanos , Animais , Cães , Esclerose Amiotrófica Lateral/tratamento farmacológico , Riluzol/uso terapêutico , Encéfalo , Administração Oral
2.
Pharmacol Biochem Behav ; 204: 173157, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33647274

RESUMO

Opioid receptors modulate neurochemical and behavioral responses to drugs of abuse in nonclinical models. Samidorphan (SAM) is a new molecular entity that binds with high affinity to human mu- (µ), kappa- (κ), and delta- (δ) opioid receptors and functions as a µ-opioid receptor antagonist with partial agonist activity at κ- and δ-opioid receptors. Based on its in vitro profile, we hypothesized that SAM would block key neurobiological effects of drugs of abuse. Therefore, we assessed the effects of SAM on ethanol-, oxycodone-, cocaine-, and amphetamine-induced increases in extracellular dopamine (DAext) in the nucleus accumbens shell (NAc-sh), and ethanol and cocaine self-administration behavior in rats. In microdialysis studies, administration of SAM alone did not result in measurable changes in NAc-sh DAext when given across a large range of doses. However, SAM markedly decreased average and maximal increases in NAc-sh DAext produced by each of the drugs of abuse tested. In behavioral studies, SAM attenuated fixed-ratio ethanol self-administration and progressive ratio cocaine self-administration. These results highlight the potential of SAM to counteract the neurobiological and behavioral effects of several drugs of abuse with differing mechanisms of action.


Assuntos
Dopamina/metabolismo , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides/metabolismo , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Anfetamina/farmacologia , Animais , Cocaína/farmacologia , Etanol/farmacologia , Humanos , Masculino , Microdiálise/métodos , Naltrexona/farmacologia , Núcleo Accumbens/metabolismo , Oxicodona/farmacologia , Ratos , Ratos Wistar , Receptores Opioides mu/metabolismo , Autoadministração/métodos
3.
J Psychopharmacol ; 33(12): 1620-1627, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31512988

RESUMO

BACKGROUND: The combination of buprenorphine, a partial mu-opioid receptor agonist and a functional kappa-opioid receptor antagonist, with samidorphan, a functional mu-opioid receptor antagonist, is being developed as an adjunct therapy for major depressive disorder, in order to harness the mood-enhancing effects of opioids without unwanted side-effects such as a risk of addiction. Acute and subacute administration of the combination of buprenorphine and samidorphan is effective in reducing forced swim immobility in the Wistar-Kyoto rat, but the chronic effects have not been examined. AIMS AND METHODS: The purpose of this study was to assess if chronic (14-day) administration of buprenorphine (0.1 mg/kg, subcutaneous) alone or in combination with samidorphan (0.3 mg/kg, subcutaneous) maintains antidepressant-like activity in the olfactory bulbectomised rat model and the Wistar-Kyoto rat, two models that exhibit ongoing behavioural deficits in tests commonly used to study effects of antidepressants. RESULTS: Olfactory bulbectomised-induced hyperactivity was attenuated by chronic administration of buprenorphine alone and in combination with samidorphan, to that of sham control activity levels. Neither buprenorphine nor samidorphan altered stress-associated defecation in sham or olfactory bulbectomised rats in the open field. In Wistar-Kyoto rats, buprenorphine alone significantly reduced forced swim immobility and increased locomotor activity three hours post-final dosing. Buprenorphine plus samidorphan significantly reduced forced swim immobility without changing locomotor activity at this time point. Buprenorphine alone also significantly reduced forced swim immobility 24 h post-final dosing. CONCLUSION: Chronic treatment of buprenorphine alone or buprenorphine plus samidorphan is effective in reversing behavioural deficits in distinct non-clinical paradigms. These non-clinical results complement the antidepressant effect of this combination observed in clinical studies.


Assuntos
Antidepressivos/administração & dosagem , Buprenorfina/administração & dosagem , Depressão/tratamento farmacológico , Naltrexona/análogos & derivados , Animais , Antidepressivos/farmacologia , Comportamento Animal/efeitos dos fármacos , Buprenorfina/farmacologia , Modelos Animais de Doenças , Quimioterapia Combinada , Locomoção/efeitos dos fármacos , Masculino , Naltrexona/administração & dosagem , Naltrexona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Ratos , Ratos Endogâmicos WKY , Ratos Sprague-Dawley
4.
J Psychopharmacol ; 33(10): 1303-1316, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31294646

RESUMO

BACKGROUND: Olanzapine, regarded as one of the most efficacious antipsychotic medications for the treatment of schizophrenia, is associated with a high risk of weight gain and metabolic dysfunction. ALKS 3831, a clinical candidate for treatment of schizophrenia, is a combination of olanzapine and samidorphan, an opioid receptor antagonist. The addition of samidorphan is intended to mitigate weight gain and the metabolic dysregulation associated with the use of olanzapine. METHODS: Non-clinical studies were conducted to assess the metabolic effects of olanzapine and samidorphan alone and in combination at clinically relevant exposure levels. RESULTS: Chronic olanzapine administration in male and female rats shifted body composition by increasing adipose mass, which was accompanied by an increase in the rate of weight gain in female rats. Co-administration of samidorphan normalized body composition in both sexes and attenuated weight gain in female rats. In hyperinsulinemic euglycemic clamp experiments conducted prior to measurable changes in weight and/or body composition, olanzapine decreased hepatic insulin sensitivity and glucose uptake in muscle while increasing uptake in adipose tissue. Samidorphan appeared to normalize glucose utilization in both tissues, but did not restore hepatic insulin sensitivity. In subsequent studies, samidorphan normalized olanzapine-induced decreases in whole-body glucose clearance following bolus insulin administration. Results from experiments in female monkeys paralleled the effects in rats. CONCLUSIONS: Olanzapine administration increased weight gain and adiposity, both of which were attenuated by samidorphan. Furthermore, the combination of olanzapine and samidorphan prevented olanzapine-induced insulin insensitivity. Collectively, these data indicate that samidorphan mitigates several metabolic abnormalities associated with olanzapine in both the presence and the absence of weight gain.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Antipsicóticos/efeitos adversos , Transtornos do Metabolismo de Glucose , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Olanzapina/efeitos adversos , Aumento de Peso/efeitos dos fármacos , Animais , Combinação de Medicamentos , Feminino , Transtornos do Metabolismo de Glucose/induzido quimicamente , Transtornos do Metabolismo de Glucose/tratamento farmacológico , Transtornos do Metabolismo de Glucose/prevenção & controle , Macaca fascicularis , Masculino , Naltrexona/administração & dosagem , Naltrexona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Ratos , Ratos Sprague-Dawley , Fatores Sexuais
5.
Neuropharmacology ; 146: 327-336, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30553825

RESUMO

Modulation of the opioid system has re-emerged as a potential therapeutic avenue for treating depression, with efficacy of a fixed-dose combination of buprenorphine (BUP), a partial µ-opioid receptor (MOR) agonist and κ-opioid receptor (KOR) antagonist, and samidorphan (SAM), a potent MOR antagonist, as an adjuvant treatment in patients with major depressive disorder (MDD). To advance understanding of the mechanism of action underlying this combination, we examined BUP, SAM and their combination in a series of rat behavioural assays. We examined effects on locomotor activity in Sprague Dawley (SD) rats over an extended period of time in a home-cage tracking system, and behavioural despair (immobility) in the forced swim test (FST), a commonly-used test to study antidepressants, in SD and Wistar-Kyoto (WKY) rats. Strain differences in opioid receptor and prepropeptide mRNA expression in the brain (prefrontal cortex, amygdala, hippocampus and striatum) were examined using qRT-PCR. BUP produced locomotor hyperactivity in SD rats from 2 to 6 h following administration, which was attenuated by SAM. In SD rats, a low, but not a high, dose of SAM in combination with BUP counteracted swim-stress induced immobility. This effect was not seen with BUP alone. In contrast, BUP alone reduced immobility in WKY rats, and this effect was enhanced by a low, but not high, dose of SAM. In WKY rats, MOR mRNA expression was higher in the hippocampus and lower in the striatum vs. SD rats. KOR mRNA expression was higher in the amygdala and nociceptin receptor (NOP) mRNA expression was lower in the hippocampus vs. SD rats. Differences in opioid receptor expression may account for the differential behavioural profile of WKY and SD rats. In summary, administration of BUP, a MOR receptor agonist together with a MOR opioid-receptor antagonist, SAM, reduces behavioural despair in animal models traditionally used to study effects of antidepressants.


Assuntos
Comportamento Animal/efeitos dos fármacos , Buprenorfina/farmacologia , Depressão/tratamento farmacológico , Atividade Motora/efeitos dos fármacos , Naltrexona/análogos & derivados , Natação , Tonsila do Cerebelo/metabolismo , Animais , Depressão/metabolismo , Hipocampo/metabolismo , Masculino , Naltrexona/farmacologia , Peptídeos Opioides/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/metabolismo
6.
Neuropharmacology ; 146: 316-326, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30448421

RESUMO

Approximately two-thirds of major depressive disorder (MDD) patients do not respond adequately to current therapies. BUP/SAM (ALKS 5461), a combination of buprenorphine (BUP) and samidorphan (SAM), is a novel opioid system modulator in development as an adjunct treatment for MDD. Using a rat strain (Wistar Kyoto rat) that is predisposed to stress and has an inadequate response to selective serotonin reuptake inhibitors (SSRIs), we investigated the effect of BUP and SAM, individually and in combination, in established nonclinical assays used to study antidepressants (the forced swim test, FST) and anxiolytics (marble burying test). As opioids and their receptors are expressed in mesocorticolimbic regions of the brain, we analyzed extracellular concentrations of dopamine, serotonin, and/or their metabolites in brain areas associated with mood and motivation. BUP alone and in combination with SAM significantly reduced immobility in the FST. Similarly, the BUP/SAM combination significantly reduced immobility in SSRI (escitalopram)-treated rats. BUP/SAM also decreased burying behavior. SAM attenuated BUP-induced changes of extracellular levels of serotonin and dopamine in the medial prefrontal cortex and nucleus accumbens shell. The latter suggests that the addition of SAM to BUP may limit activation of the mesolimbic dopamine reward pathway and thereby reduce BUP's reinforcing properties. SAM alone had no effect on neurochemistry or immobility in the FST. Collectively, these data indicate that opioid system modulation may offer an alternative mechanism that does not rely on enhanced serotonergic neurotransmission in neurocircuits associated with antidepressant and anxiolytic activity in nonclinical models.


Assuntos
Analgésicos Opioides/farmacologia , Buprenorfina/farmacologia , Transtorno Depressivo Maior/tratamento farmacológico , Naltrexona/análogos & derivados , Animais , Antidepressivos/farmacologia , Comportamento Animal/efeitos dos fármacos , Citalopram/farmacologia , Dopamina/metabolismo , Combinação de Medicamentos , Quimioterapia Combinada , Masculino , Modelos Animais , Atividade Motora/efeitos dos fármacos , Naltrexona/farmacologia , Ratos , Ratos Endogâmicos WKY , Serotonina/metabolismo , Natação
7.
Drug Dev Res ; 79(5): 234-238, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30188587

RESUMO

Preclinical Research & Development Background: Samidorphan, a µ-opioid receptor antagonist, is in clinical development for central nervous system related diseases. The discriminative stimulus effects of samidorphan were assessed in rats trained to discriminate the effects of a known morphinan of abuse, morphine, from that of saline. METHODS: Escalating doses of samidorphan were substituted for morphine and rats were allowed to respond on two levers for food reward. Doses of samidorphan were chosen based on other pharmacodynamic assays in which samidorphan blocked the effects of morphine (such as blocking analgesia in a hot plate test; data not shown). In addition, a pharmacokinetic study was conducted to determine if these doses would reflect predicted exposure levels that translate to human equivalent doses. RESULTS: Rats discriminating morphine from vehicle responded predominantly on the vehicle lever after receiving samidorphan. In addition, samidorphan was rapidly absorbed, and plasma concentrations of the doses tested in this study bracket therapeutically relevant concentrations. CONCLUSIONS: In summary, samidorphan produced saline-like behavioral responses over a wide dose range.


Assuntos
Discriminação Psicológica , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides mu/antagonistas & inibidores , Animais , Relação Dose-Resposta a Droga , Humanos , Masculino , Morfina/farmacologia , Naltrexona/farmacocinética , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacocinética , Ratos Sprague-Dawley
8.
J Pharmacol Exp Ther ; 367(2): 267-281, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30108159

RESUMO

A combination of buprenorphine (BUP) and samidorphan (SAM) at a 1:1 (mg/mg) fixed-ratio dose is being investigated as an adjunctive treatment of major depressive disorder (BUP/SAM, ALKS 5461). Both [3H]BUP and [3H]SAM bound to the µ-, κ-, and δ-opioid receptors (MOR, KOR, and DOR, respectively) with Kd values of 3 nM or less. [3H]BUP dissociated from the MOR more slowly than [3H]SAM did. In the [35S]GTPγS assay, BUP was a partial agonist at the MOR, KOR, and DOR. SAM was an antagonist at the MOR and a partial agonist at the KOR and DOR. The pharmacology of the combination of SAM and BUP was characterized at ratios like the molar ratios of both compounds at steady state in humans. In all assessments, SAM reduced the efficacy of BUP at the MOR without altering its potency. At the KOR, SAM had no significant effect on the activity of BUP. In bioluminescent resonance energy transfer assays, SAM, naltrexone, and naloxone were partial agonists when the MOR was coupled to the Gα oB and Gα z, and were antagonists when coupled to Gα i At the KOR, SAM was a partial agonist activating Gα oA and Gα oB and a full agonist in stimulating Gα z SAM inhibited BUP's recruitment of ß-arrestin to the MOR, suggesting an attenuation of BUP's efficacy in activating G proteins correlated with an inhibition of ß-arrestin recruitment. The collective data suggest that SAM attenuates the efficacy of BUP under all conditions tested at the MOR and DOR but had little effect on BUP activity at the KOR.


Assuntos
Buprenorfina/farmacologia , Transtorno Depressivo Maior/tratamento farmacológico , Naltrexona/análogos & derivados , Animais , Células CHO , Linhagem Celular , Cricetulus , Combinação de Medicamentos , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Naloxona/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , beta-Arrestinas/metabolismo
9.
Brain Behav Immun ; 67: 152-162, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28844812

RESUMO

Patients receiving the cytokine immunotherapy, interferon-alpha (IFN-α) frequently present with neuropsychiatric consequences and cognitive impairments. Patients (25-80%) report symptoms of depression, including, anhedonia, irritability, fatigue and impaired motivation. Our lab has previously demonstrated treatment (170,000IU/kg sc, 3 times per week for 4weeks) of the pro-inflammatory cytokine, IFN-α, induced a depressive phenotype in rats in the forced swim test (FST). Here, we examine the biological mechanisms underlying behavioral changes induced by IFN-α, which may be reflective of mechanisms underlying inflammation associated depression. We also investigate the potential of 3-carboxamido seco-nalmefene (3CS-nalmefene), a novel opioid modulator (antagonist at mu and partial agonist at kappa and delta opioid receptors in vitro), to reverse IFN-α induced changes. In vitro radioligand receptor binding assays and the [35S] GTPγS were performed to determine the affinity of 3CS-nalmefene for the mu, kappa and delta opioid receptors. IFN-α treatment increased circulating and central markers of inflammation and hypothalamic-pituitaryadrenal (HPA) axis activity (IL-6, IL-1ß and corticosterone) while increasing immobility in the FST, impairing of object displacement learning in the object exploration task (OET), and decreasing neuronal proliferation and brain-derived neurotrophic factor (BDNF) in the hippocampus. Treatment with 3CS-nalmefene (0.3mg/kg/sc twice per day, 3 times per week for 4weeks) prevented IFN-α-induced immobility in the FST and impaired object displacement learning. In addition, 3CS-nalmefene prevented IFN-α-induced increases in inflammation and hyperactivity of the HPA-axis, the IFN-α-induced reduction in both neuronal proliferation and BDNF expression in the hippocampus. Overall, these preclinical data would support the hypothesis that opioid receptor modulation is a relevant target for treatment of depression.


Assuntos
Antidepressivos/administração & dosagem , Transtorno Depressivo/tratamento farmacológico , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/administração & dosagem , Receptores Opioides/agonistas , Animais , Ansiedade/induzido quimicamente , Ansiedade/tratamento farmacológico , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proliferação de Células/efeitos dos fármacos , Depressão/induzido quimicamente , Depressão/tratamento farmacológico , Transtorno Depressivo/induzido quimicamente , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Interferon-alfa/administração & dosagem , Masculino , Naltrexona/administração & dosagem , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos Wistar
10.
Physiol Behav ; 167: 28-34, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27591842

RESUMO

Depression is a debilitating psychiatric disorder that is highly comorbid with anxiety. Depression is twice as prevalent in women as in men, however, females remain underrepresented in preclinical research. The stress hyperresponsive Wistar-Kyoto (WKY) rat displays hypolocomotion in a novel aversive environment and depressive- and anxiety-like behaviours, which have been mostly characterised in males. The current study characterised behaviour in male and female rats in a battery of behavioural paradigms. Adult male and female WKY rats were tested in the open field and forced swim tests (tests with a locomotor component); and the marble burying, novelty-induced hypophagia and sucrose preference tests (tests with a minimal locomotor component) and 24h home-cage locomotor activity was also monitored. The tests were compared against the Sprague-Dawley (SD) strain, a commonly used "control" strain. SD, but not WKY, females exhibited higher home-cage locomotor activity compared to males. In the open field, WKY rats of both sexes exhibited a significant reduction in locomotor activity and increased anxiety-like behaviour as demonstrated by reduced time in the aversive inner zone of the open field, compared to SD counterparts. In the marble burying test, WKY females, but not males, exhibited a trend towards increased burying, indicative of anxiety-like/neophobic behaviour. In comparison, WKY males, but not females, exhibited enhanced novelty-induced hypophagia, indicative of increased anxiety-like behaviour compared to SD rats. In the forced swim test, WKY rats of both sexes spent more time immobile compared with SD counterparts, indicating depressive-like behaviour. However, in comparison to SD rats, WKY males, but not females, exhibited anhedonic-like behaviour. In conclusion, WKY rats exhibit depressive- and anxiety-like behaviours that are complex and nuanced depending on the sex of the rat and testing conditions. This study supports the use of a varied test battery to fully characterise depression/anxiety-like behaviour in male and female rats.


Assuntos
Ansiedade/fisiopatologia , Depressão/fisiopatologia , Análise de Variância , Animais , Modelos Animais de Doenças , Feminino , Preferências Alimentares , Locomoção/fisiologia , Masculino , Escalas de Graduação Psiquiátrica , Ratos , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Fatores Sexuais , Natação/psicologia
13.
Pharm Res ; 31(8): 2065-77, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24558010

RESUMO

PURPOSE: Long-acting injectables (LAIs) are increasingly recognized as an effective therapeutic approach for treating chronic conditions. Many LAIs are formulated to create a poorly soluble depot from which the active agent is delivered over time. This long residing depot can cause localized chronic-active inflammation in the tissue, which has not been well defined in the literature. The purpose of this work is to establish an experimental baseline for describing these responses. METHODS: Non-human primates and rodents were used to examine the response to LAI formulations of two clinically relevant atypical antipsychotics, aripiprazole monohydrate and olanzapine pamoate monohydrate. RESULTS: A foreign body response develops with elevations of key cytokines such as IL-1α, IL-1ß, TNFα, and IL6 at the site of injection. However, the tissue response for the two atypical antipsychotics compounds diverge as evidenced by quantitative differences observed in cytokine levels at various time points after dosing. CONCLUSIONS: Our studies show that, while the drugs are in the same therapeutic class, the response to each of these compounds can be distinguished qualitatively and quantitatively, supporting the idea that the injection site reaction involves a multiplicity of factors including the properties of the compound and cellular dynamics at the site of injection.


Assuntos
Antipsicóticos/administração & dosagem , Antipsicóticos/metabolismo , Mediadores da Inflamação/metabolismo , Piperazinas/administração & dosagem , Piperazinas/metabolismo , Quinolonas/administração & dosagem , Quinolonas/metabolismo , Animais , Aripiprazol , Relação Dose-Resposta a Droga , Injeções Subcutâneas , Macaca fascicularis , Masculino , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia
14.
Comp Med ; 62(3): 209-17, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22776054

RESUMO

Self-injurious behavior (SIB) is a spontaneous behavior that threatens the health and wellbeing of multiple species. In humans, the opioid antagonist naltrexone hydrochloride has been used successfully to modulate the endogenous opioid system and reduce the occurrence of SIB. This study is the first to assess the efficacy of extended-release naltrexone in the pharmacologic treatment of SIB in rhesus macaques (Macaca mulatta). In an acute pharmacokinetic study of 4 macaques, we determined the mean naltrexone plasma concentration was maintained above the therapeutic level (2 ng/mL) after administration of a single dose (20 mg/kg) of 28-d extended-release naltrexone throughout the release period. For a subsequent treatment study, we selected 8 singly housed macaques known to engage in SIB. The study comprised a 4-wk baseline phase; an 8-wk treatment phase, during which each macaque received 2 doses of extended-release naltrexone 28 d apart; and a 4-wk posttreatment phase. Plasma samples were collected and analyzed weekly for naltrexone concentrations throughout the treatment and posttreatment phases. In addition, total of 6 h of video was analyzed per animal per phase of the study. Compared with baseline phases, both the frequency and the percentage of time spent displaying SIB decreased during the treatment phase, and the percentage of time remained decreased during the posttreatment phase. In contrast, extended-release naltrexone did not alter the expression of other abnormal, anxiety-related, or agonistic behaviors nor were levels of inactivity affected. The present study supports the use of naltrexone in the treatment of SIB in rhesus macaques.


Assuntos
Naltrexona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Comportamento Autodestrutivo/prevenção & controle , Animais , Macaca mulatta , Masculino , Naltrexona/farmacocinética , Antagonistas de Entorpecentes/farmacocinética
15.
Pharmacol Biochem Behav ; 100(3): 530-7, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22056608

RESUMO

Naltrexone is a µ-opioid receptor antagonist that has been extensively studied for its ability to block the rewarding effects of ethanol. Opioid receptors are widely distributed within the gastrointestinal tract (GIT). Typically, naltrexone is administered by parenteral routes in nonclinical studies. We initially tested if opioid receptors within the GIT would influence the ability of oral naltrexone to inhibit ethanol oral self-administration in rats using the co-administration of oral loperamide, a peripherally restricted opioid agonist. As expected, oral naltrexone only had modest effects on ethanol intake, and the response was not dose-dependent. However in rats, treatment with loperamide prior to the administration of naltrexone resulted in a suppression of ethanol intake which approached that observed with naltrexone given by the subcutaneous (SC) route. Importantly, administration of loperamide prior to administration of naltrexone did not alter blood concentrations of naltrexone. We then evaluated if oral loperamide would enhance effects of baclofen (a GABA(B) receptor agonist) and AM-251 (a CB-1 receptor antagonist) and found that pre-treatment with loperamide did potentiate the action of both drugs to reduce ethanol self-administration. Finally, the specific opioid receptor type involved was investigated using selective µ- and κ-receptor antagonists to determine if these would affect the ability of the AM-251 and loperamide combination to block ethanol drinking behavior. The effect of loperamide was blocked by ALKS 37, a peripherally restricted µ-receptor antagonist. These data suggest an important role for opioid receptors within the GIT in modulating central reward pathways and may provide new insights into strategies for treating reward disorders, including drug dependency.


Assuntos
Dissuasores de Álcool/uso terapêutico , Consumo de Bebidas Alcoólicas/prevenção & controle , Baclofeno/uso terapêutico , Loperamida/uso terapêutico , Naltrexona/uso terapêutico , Piperidinas/uso terapêutico , Pirazóis/uso terapêutico , Receptores Opioides mu/agonistas , Administração Oral , Dissuasores de Álcool/administração & dosagem , Dissuasores de Álcool/sangue , Dissuasores de Álcool/farmacocinética , Animais , Animais não Endogâmicos , Baclofeno/administração & dosagem , Comportamento Animal/efeitos dos fármacos , Sinergismo Farmacológico , Quimioterapia Combinada , Agonistas dos Receptores de GABA-B/administração & dosagem , Agonistas dos Receptores de GABA-B/uso terapêutico , Loperamida/administração & dosagem , Loperamida/antagonistas & inibidores , Masculino , Naltrexona/administração & dosagem , Naltrexona/sangue , Naltrexona/farmacocinética , Antagonistas de Entorpecentes/sangue , Antagonistas de Entorpecentes/farmacocinética , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/uso terapêutico , Piperidinas/administração & dosagem , Pirazóis/administração & dosagem , Ratos , Ratos Wistar , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides mu/antagonistas & inibidores
16.
Behav Pharmacol ; 22(1): 87-90, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21160425

RESUMO

Amphetamines are the second most highly abused illicit drugs worldwide, yet there is no pharmacological treatment for amphetamine abuse and dependence. Preclinical studies and, more recently, human studies, suggest that the opioid receptor antagonist, naltrexone, might be useful in the treatment of amphetamine abuse. Naltrexone, an opioid receptor antagonist, is currently used for the treatment of alcohol dependence. The aim of this study was to explore the ability of naltrexone to modify self-administration of amphetamine or ethanol in rhesus monkeys. Monkeys were trained to respond to intravenous injections of either D-amphetamine (0.003 mg/kg/injection) or ethanol (0.05 g/kg/injection) on a fixed ratio 30 schedule. Naltrexone (0.01-1 mg/kg) was administered intramuscularly 30 min before the start of treatment test sessions. Naltrexone dose-dependently decreased both amphetamine and ethanol self-administration. These findings support the potential use of naltrexone as therapy for amphetamine and polydrug abuse.


Assuntos
Anfetamina/administração & dosagem , Comportamento Animal/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/administração & dosagem , Etanol/administração & dosagem , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Animais , Condicionamento Operante/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Macaca mulatta , Masculino , Autoadministração
17.
Addict Biol ; 14(4): 408-18, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19489752

RESUMO

Opioid receptor antagonism has been shown to attenuate behavioral and neurochemical effects of amphetamine in humans and rodents. The effects of acute (oral or subcutaneous) or extended-release naltrexone (XR-NTX) were tested on the reward-enhancing effects of amphetamine using the intracranial self-stimulation (ICSS) paradigm. Acute exposure to drugs of abuse reduces the locus of rise (LOR) in the ICSS procedure, reflecting enhanced brain stimulation reward (BSR). Rats were treated once a day with naltrexone orally (PO; 5.0 mg/kg) or subcutaneously (SC; 0.5 mg/kg) for four consecutive days and tested with D-amphetamine (0.5 mg/kg, intraperitoneal) in the ICSS paradigm 30 minutes later on days 1 and 4. Separate groups of rats received XR-NTX (50 mg/kg, SC) or placebo microspheres (similar mass to XR-NTX, SC) on day 0 and tested with D-amphetamine in the ICSS paradigm on days 4, 14, 21, 28 and 41 after administration. Naltrexone plasma concentrations were determined for each amphetamine testing session using liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). In rats pretreated with naltrexone acutely, amphetamine-potentiated BSR did not differ from vehicle-pretreated rats on either day 1 or day 4 (25-30% decrease in LOR). In XR-NTX-pretreated rats, amphetamine-potentiated BSR was reduced by 64 and 70% on days 4 and 14, respectively, compared to placebo microsphere-treated controls. This effect dissipated by day 21. Naltrexone plasma concentrations were comparable across all treatment groups (14-30 ng/ml) on days 1, 4 and 14. In summary, an extended-release formulation of naltrexone results in significant attenuation of psychostimulant-enhanced BSR that is not observed with acute naltrexone.


Assuntos
Encéfalo/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/antagonistas & inibidores , Estimulantes do Sistema Nervoso Central/farmacologia , Dextroanfetamina/antagonistas & inibidores , Dextroanfetamina/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Administração Oral , Animais , Estimulantes do Sistema Nervoso Central/administração & dosagem , Dextroanfetamina/administração & dosagem , Vias de Administração de Medicamentos , Esquema de Medicação , Cromatografia Gasosa-Espectrometria de Massas , Injeções Intraperitoneais , Injeções Subcutâneas , Masculino , Naltrexona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Ratos , Ratos Sprague-Dawley , Recompensa
18.
Pharmacol Biochem Behav ; 89(4): 515-22, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18342360

RESUMO

A monthly extended-release formulation of the opioid antagonist naltrexone (XR-NTX) is approved for treatment of alcohol dependence. There is little research regarding overriding chronic (>21 days) competitive opioid receptor blockade with opioids for acute pain. Using the hot plate test after XR-NTX or placebo microsphere administration, rats were treated with an opioid analgesic to determine the dose required to produce the maximum response latency (MRL; 60 s). Rats were later treated with the same opioid to determine any potential effects on respiration rate or locomotor activity. In naïve rats, 15 mg/kg morphine, 0.1 mg/kg fentanyl and 8 mg/kg hydrocodone produced MRL. In XR-NTX treated rats, morphine produced 36% and 46% MRL at 90 mg/kg on days 4 and 19 and 96% MRL at 45 mg/kg on day 39. Fentanyl produced 100% MRL at 2.0 mg/kg on days 4 and 19 and at 0.5 mg/kg on day 39. Hydrocodone (80 mg/kg) produced 69%, 80% and 100% MRL on days 4, 19 and 39. Compared to placebo, these doses did not further depress respiration or alter locomotor activity. Thus, opioid receptor blockade with XR-NTX can be overcome in rats with higher doses of opioids without further affecting respiration or locomotor activity.


Assuntos
Analgésicos Opioides/administração & dosagem , Naltrexona/administração & dosagem , Antagonistas de Entorpecentes/administração & dosagem , Alcoolismo/tratamento farmacológico , Analgésicos Opioides/efeitos adversos , Animais , Preparações de Ação Retardada , Interações Medicamentosas , Fentanila/administração & dosagem , Fentanila/efeitos adversos , Humanos , Hidrocodona/administração & dosagem , Hidrocodona/efeitos adversos , Masculino , Morfina/administração & dosagem , Morfina/efeitos adversos , Atividade Motora/efeitos dos fármacos , Naltrexona/sangue , Antagonistas de Entorpecentes/sangue , Dor/tratamento farmacológico , Medição da Dor , Pletismografia Total , Ratos , Ratos Sprague-Dawley , Respiração/efeitos dos fármacos
19.
J Aerosol Med ; 20(2): 112-26, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17536950

RESUMO

The purpose of this study was to characterize the toxicity, pharmacokinetics, and pharmacodynamics of human insulin inhalation powder (HIIP) in beagle dogs when administered daily as an aerosolized dry powder formulation for 26 weeks via head-only inhalation. Conscious beagle dogs were exposed for 15 mins/day to an air control, placebo, maximal placebo (approximately three-fold the placebo dose), or one of three doses of HIIP (mean inhaled doses of 80, 240, or 701 microg/kg/day for the HIIP-low, HIIP-mid, and HIIP-high dose, respectively), The mass median aerodynamic diameters (MMAD) were between 2 and 3 microm and geometric standard deviation (GSD) values were approximately 2 across the groups, which is the ideal size range for favorable lung deposition. All groups were comprised of four dogs/sex, with the air control, HIIP-high, and maximal placebo groups having an additional two dogs/sex as recovery subgroups. Concentrations of serum insulin and glucose were determined from blood samples obtained following the first and last exposure for evaluation of the pharmacokinetics and pharmacodynamics of HIIP. Dose-related exposure (C(max), AUC) to inhaled insulin was observed with rapid absorption and no apparent gender differences or accumulation after repeated inhalation exposures for 26 weeks. The expected pharmacological effect of insulin was observed with dose-related decreases in serum glucose levels following HIIP administration. There were no toxic effects observed including no HIIP or placebo treatment-related effects on mean body weights, absolute body weight changes, clinical observations, food consumption, respiratory function parameters, ophthalmic examinations, electrocardiograms, heart rates, clinical pathology, or urinalysis. Similarly, there were no HIIP or placebo treatment-related effects on pulmonary assessments that included respiratory function parameters, bronchial alveolar lavage assessments, organ weights, or macroscopic and microscopic evaluations, including lung cell proliferation indices. HIIP was considered to have either low or no immunogenic potential in dogs. The no-observed-adverse-effect level (NOAEL) and maximum tolerated dose were the average inhaled dose of 701 microg insulin/kg/day.


Assuntos
Glicemia/análise , Insulina/farmacocinética , Administração por Inalação , Animais , Cães , Feminino , Insulina/administração & dosagem , Insulina/sangue , Insulina/farmacologia , Masculino , Pós/administração & dosagem , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...