Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 386
Filtrar
1.
J Neurosci Nurs ; 56(4): 136-142, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38976832

RESUMO

ABSTRACT: INTRODUCTION: Nurses have a central role in educating patients and families about treatment options and how to integrate them into action plans for neurologic conditions. In recent years, a growing number of intranasal formulations have become available as rescue therapy for neurologic conditions or symptoms including migraine, opioid overdose, and seizures. Rescue therapies do not replace maintenance medications or emergency care but are designed to enable rapid treatment of urgent or disabling conditions in community settings. Yet, discussion of rescue therapies for neurologic conditions remains limited in nursing literature. CONTENT: Intranasal formulations are specifically formulated for delivery and absorption in the nose and have several characteristics that are well suited as rescue therapies for neurologic conditions. Intranasal formulations include triptans for migraine, naloxone and nalmefene for opioid overdose, and benzodiazepines for seizure clusters in patients with epilepsy. Therapeutic attributes discussed here include ease of use in community settings by nonmedical professionals, relatively rapid onset of action, and favorable safety profile and patient experience. This information is critical for nurses to make informed decisions about rescue therapy options, incorporate these into plans of care, and educate patients, care partners, and other healthcare providers. CONCLUSION: Rescue therapies are increasingly important in the care of people with neurologic conditions. Various formulations are available and continue to evolve, offering easy and quick ways for nurses, patients, and nonmedical care partners to administer critical rescue medications. For nurses overseeing medication management, the attributes of intranasal rescue therapies should be considered in the context of providing patients with the right care at the right time.


Assuntos
Administração Intranasal , Doenças do Sistema Nervoso , Humanos , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/antagonistas & inibidores , Benzodiazepinas/administração & dosagem , Benzodiazepinas/uso terapêutico , Naloxona/administração & dosagem , Naloxona/uso terapêutico , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/uso terapêutico , Doenças do Sistema Nervoso/induzido quimicamente , Doenças do Sistema Nervoso/tratamento farmacológico , Triptaminas/uso terapêutico , Triptaminas/administração & dosagem
2.
Ann N Y Acad Sci ; 1520(1): 105-114, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36514207

RESUMO

Studies conducted in rodents indicate a crucial role of the opioid circuit in mediating objective hedonic reactions to primary rewards. However, it remains unclear whether opioid transmission is also essential to experience pleasure with more abstract rewards, such as music. We addressed this question using a double-blind within-subject pharmacological design in which opioid levels were up- and downregulated by administering an opioid agonist (oxycodone) and antagonist (naltrexone), respectively, before healthy participants (n = 21) listened to music. Participants also performed a monetary incentive delay (MID) task to control for the effectiveness of the treatment and the specificity of the effects. Our results revealed that the pharmacological intervention did not modulate subjective reports of pleasure, nor the occurrence of chills. On the contrary, psychophysiological (objective) measures of emotional arousal, such as skin conductance responses (SCRs), were bidirectionally modulated in both the music and MID tasks. This modulation specifically occurred during reward consumption, with greater pleasure-related SCR following oxycodone than naltrexone. These findings indicate that opioid transmission does not modulate subjective evaluations but rather affects objective reward-related psychophysiological responses. These findings raise new caveats about the role of the opioidergic system in the modulation of pleasure for more abstract or cognitive forms of rewarding experiences, such as music.


Assuntos
Analgésicos Opioides , Música , Prazer , Analgésicos Opioides/agonistas , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/metabolismo , Música/psicologia , Naltrexona , Oxicodona , Prazer/fisiologia , Recompensa , Método Duplo-Cego , Voluntários Saudáveis , Humanos
3.
Braz. J. Pharm. Sci. (Online) ; 59: e22764, 2023. graf
Artigo em Inglês | LILACS | ID: biblio-1439499

RESUMO

Abstract Remifentanil is a modern fentanyl analogue with ultrashort-action granted by an esterase-labile methyl propanoate chain. Here, we present the development of a continuous flow methodology for the key N-alkylation step of remifentanil preparation in a biphasic, "slug-flow" regime. We screened parameters under microwave-assisted reactions, translated conditions to flow settings, and obtained remifentanil under 15-min residence time in a 1-mL microreactor, with a space-time yield of 89 mg/mL·h and 94% yield.


Assuntos
Preparações Farmacêuticas/análise , Remifentanil/farmacologia , Analgésicos Opioides/antagonistas & inibidores , Fluxo Contínuo
5.
Neuropeptides ; 90: 102185, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34419803

RESUMO

It has been shown that systemic and local administration of ultra-low dose morphine induced a hyperalgesic response via mu-opioid receptors. However, its exact mechanism(s) has not fully been clarified. It is documented that mu-opioid receptors functionally couple to T-type voltage dependent Ca+2 channels. Here, we investigated the role of T-type calcium channels, amiloride and mibefradil, on the induction of low-dose morphine hyperalgesia in male Wistar rats. The data showed that morphine (0.01 µg i.t. and 1 µg/kg i.p.) could elicit hyperalgesia as assessed by the tail-flick test. Administration of amiloride (5 and 10 µg i.t.) and mibefradil (2.5 and 5 µg i.t.) completely blocked low-dose morphine-induced hyperalgesia in spinal dorsal horn. Amiloride at doses of 1 and 5 mg/kg (i.p.) and mibefradil (9 mg/kg ip) 10 min before morphine (1 µg/kg i.p.) inhibited morphine-induced hyperalgesia. Our results indicate a role for T-type calcium channels in low dose morphine-induced hyperalgesia in rats.


Assuntos
Analgésicos Opioides/efeitos adversos , Canais de Cálcio Tipo T/efeitos dos fármacos , Hiperalgesia/induzido quimicamente , Morfina/efeitos adversos , Amilorida/farmacologia , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/antagonistas & inibidores , Animais , Relação Dose-Resposta a Droga , Injeções Intraperitoneais , Masculino , Mibefradil/farmacologia , Morfina/administração & dosagem , Morfina/antagonistas & inibidores , Medição da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Células do Corno Posterior/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores Opioides mu
6.
Neuropharmacology ; 185: 108437, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33316279

RESUMO

BACKGROUND: Antagonism of peripheral opioid receptors by methylnaltrexone (MNTX) was recently proposed as a potential mechanism to attenuate the development of opioid analgesic tolerance based on experiments conducted in mice. However, reports indicate that MNTX is demethylated to naltrexone (NTX) in mice, and NTX may subsequently cross the blood-brain barrier to antagonize centrally-mediated opioid effects. The goal of this study was to determine whether MNTX alters centrally-mediated behaviors elicited by the opioid analgesics, morphine and oxycodone, and to quantify concentrations of MNTX and NTX in blood and brain following their administration in mice. METHODS: Combinations of MNTX and morphine were tested under acute and chronic conditions in thermal nociceptive assays. Effects of MNTX and NTX pretreatment were assessed in an oxycodone discrimination operant procedure. Blood and brain concentrations of these antagonists were quantified after their administration using liquid chromatography-mass spectrometry. RESULTS: MNTX dose-dependently attenuated acute and chronic morphine antinociception. MNTX and NTX dose-dependently antagonized the discriminative stimulus effects of oxycodone. MNTX and NTX were detected in both blood and brain after administration of MNTX, confirming its demethylation and demonstrating that MNTX itself can cross the blood-brain barrier. CONCLUSIONS: These results provide converging behavioral and analytical evidence that MNTX administration in mice attenuates centrally-mediated effects produced by opioid analgesics and results in functional concentrations of MNTX and NTX in blood and brain. Collectively, these findings indicate that MNTX cannot be administered systemically in mice for making inferences that its effects are peripherally restricted.


Assuntos
Analgésicos Opioides/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Morfina/farmacologia , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/farmacologia , Oxicodona/farmacologia , Analgésicos Opioides/antagonistas & inibidores , Animais , Barreira Hematoencefálica/metabolismo , Relação Dose-Resposta a Droga , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/antagonistas & inibidores , Naltrexona/metabolismo , Naltrexona/farmacologia , Antagonistas de Entorpecentes/metabolismo , Oxicodona/antagonistas & inibidores , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Compostos de Amônio Quaternário/metabolismo , Compostos de Amônio Quaternário/farmacologia
7.
Molecules ; 25(18)2020 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-32957550

RESUMO

The opioid receptors are members of the G-protein-coupled receptor (GPCR) family and are known to modulate a variety of biological functions, including pain perception. Despite considerable advances, the mechanisms by which opioid agonists and antagonists interact with their receptors and exert their effect are still not completely understood. In this report, six new hybrids of the Dmt-Tic pharmacophore and cyclic peptides, which were shown before to have a high affinity for the µ-opioid receptor (MOR) were synthesized and characterized pharmacologically in calcium mobilization functional assays. All obtained ligands turned out to be selective antagonists of the δ-opioid receptor (DOR) and did not activate or block the MOR. The three-dimensional structural determinants responsible for the DOR antagonist properties of these analogs were further investigated by docking studies. The results indicate that these compounds attach to the DOR in a slightly different orientation with respect to the Dmt-Tic pharmacophore than Dmt-TicΨ[CH2-NH]Phe-Phe-NH2 (DIPP-NH2[Ψ]), a prototypical DOR antagonist peptide. Key pharmacophoric contacts between the DOR and the ligands were maintained through an analogous spatial arrangement of pharmacophores, which could provide an explanation for the predicted high-affinity binding and the experimentally observed functional properties of the novel synthetic ligands.


Assuntos
Dipeptídeos/metabolismo , Peptídeos Opioides/metabolismo , Peptídeos Cíclicos/metabolismo , Receptores Opioides delta/metabolismo , Tetra-Hidroisoquinolinas/metabolismo , Analgésicos Opioides/antagonistas & inibidores , Animais , Humanos , Ligantes , Simulação de Acoplamento Molecular , Antagonistas de Entorpecentes/química , Antagonistas de Entorpecentes/metabolismo , Peptídeos Opioides/síntese química , Peptídeos Cíclicos/síntese química , Receptores Opioides/química , Relação Estrutura-Atividade
9.
Neuropharmacology ; 171: 108110, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32344007

RESUMO

MT-45 is a synthetic opioid that was developed in the 1970s as an analgesic compound. However, in recent years MT-45 has been associated with multiple deaths in Europe and has been included in the class of novel psychoactive substances known as novel synthetic opioids (NSOs). Little is known about the pharmaco-toxicological effects of MT-45. Therefore, we used a dynamic mass redistribution (DMR) assay to investigate the pharmacodynamic profile of this NSO in vitro compared with morphine. We then used in vivo studies to investigate the effect of the acute systemic administration of MT-45 (0.01-15 mg/kg i.p.) on motor and sensorimotor (visual, acoustic and tactile) responses, mechanical and thermal analgesia, muscle strength and body temperature in CD-1 male mice. Higher doses of MT-45 (6-30 mg/kg i.p.) were used to investigate cardiorespiratory changes (heart rate, respiratory rate, SpO2 saturation and pulse distention). All effects of MT-45 were compared with those of morphine. In vitro DMR assay results demonstrated that at human recombinant opioid receptors MT-45 behaves as a potent selective mu agonist with a slightly higher efficacy than morphine. In vivo results showed that MT-45 progressively induces tail elevation at the lowest dose tested (0.01 mg/kg), increased mechanical and thermal antinociception (starting from 1 to 6 mg/kg), decreased visual sensorimotor responses (starting from 3 to 6 mg/kg) and reduced tactile responses, modulated motor performance and induced muscle rigidity at higher doses (15 mg/kg). In addition, at higher doses (15-30 mg/kg) MT-45 impaired the cardiorespiratory functions. All effects were prevented by the administration of the opioid receptor antagonist naloxone. These findings reveal the risks associated with the ingestion of opioids and the importance of studying these drugs and undertaking more clinical studies of the current molecules to better understand possible therapeutic interventions in the case of toxicity.


Assuntos
Analgésicos Opioides/farmacologia , Piperazinas/farmacologia , Agressão/efeitos dos fármacos , Analgésicos Opioides/antagonistas & inibidores , Animais , Células CHO , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Hemodinâmica/efeitos dos fármacos , Temperatura Alta , Humanos , Camundongos , Morfina/antagonistas & inibidores , Morfina/farmacologia , Atividade Motora/efeitos dos fármacos , Força Muscular/efeitos dos fármacos , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos , Estimulação Física , Piperazinas/antagonistas & inibidores , Receptores Opioides/efeitos dos fármacos , Respiração/efeitos dos fármacos , Sensação/efeitos dos fármacos
10.
Toxicol Lett ; 325: 34-42, 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32070766

RESUMO

Carfentanil is an ultra-potent opioid with an analgesic potency 10,000 times that of morphine but has received little scientific investigation. Three experiments were conducted to evaluate the toxicity of carfentanil and the efficacy of naloxone in adult male African green monkeys. The first experiment determined the ED50 (found to be 0.71 µg/kg) of subcutaneous carfentanil for inducing bradypnea and/or loss of posture. Experiment 2 attempted to establish the ED50 of naloxone for rapidly reversing the bradypnea/loss of posture induced by carfentanil (1.15 µg/kg). Experiment 3 evaluated the effects of carfentanil (0.575 µg/kg) alone, the safety of naloxone (71-2841 µg/kg), and the efficacy of naloxone (71-710 µg/kg) administration at two time points following carfentanil (1.15 µg/kg) on operant choice reaction time. Collectively, these experiments characterized the temporal progression of carfentanil-induced toxic signs, determined the range of naloxone doses that restored respiratory and gross behavioral function, and determined the time course and range of naloxone doses that partially or completely reversed the effects of carfentanil on operant choice reaction time performance in African green monkeys. These results have practical relevance for the selection of opioid antagonists, initial doses, and expected functional outcomes following treatment of synthetic opioid overdose in a variety of operational/emergency response contexts.


Assuntos
Analgésicos Opioides/toxicidade , Fentanila/análogos & derivados , Naloxona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Analgésicos Opioides/antagonistas & inibidores , Animais , Apneia/induzido quimicamente , Apneia/prevenção & controle , Comportamento Animal/efeitos dos fármacos , Chlorocebus aethiops , Condicionamento Operante/efeitos dos fármacos , Overdose de Drogas/tratamento farmacológico , Fentanila/antagonistas & inibidores , Fentanila/toxicidade , Masculino , Naloxona/toxicidade , Antagonistas de Entorpecentes/toxicidade , Tempo de Reação/efeitos dos fármacos
13.
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
14.
J Am Chem Soc ; 141(26): 10489-10503, 2019 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-31187995

RESUMO

Opioid abuse in the United States has been declared a national crisis and is exacerbated by an inexpensive, readily available, and illicit supply of synthetic opioids. Specifically, fentanyl and related analogues such as carfentanil pose a significant danger to opioid users due to their high potency and rapid acting depression of respiration. In recent years these synthetic opioids have become the number one cause of drug-related deaths. In our research efforts to combat the public health threat posed by synthetic opioids, we have developed monoclonal antibodies (mAbs) against the fentanyl class of drugs. The mAbs were generated in hybridomas derived from mice vaccinated with a fentanyl conjugate vaccine. Guided by a surface plasmon resonance (SPR) binding assay, we selected six hybridomas that produced mAbs with 10-11 M binding affinity for fentanyl, yet broad cross-reactivity with related fentanyl analogues. In mouse antinociception models, our lead mAb (6A4) could blunt the effects of both fentanyl and carfentanil in a dose-responsive manner. Additionally, mice pretreated with 6A4 displayed enhanced survival when subjected to fentanyl above LD50 doses. Pharmacokinetic analysis revealed that the antibody sequesters large amounts of these drugs in the blood, thus reducing drug biodistribution to the brain and other tissue. Lastly, the 6A4 mAb could effectively reverse fentanyl/carfentanil-induced antinociception comparable to the opioid antagonist naloxone, the standard of care drug for treating opioid overdose. While naloxone is known for its short half-life, we found the half-life of 6A4 to be approximately 6 days in mice, thus monoclonal antibodies could theoretically be useful in preventing renarcotization events in which opioid intoxication recurs following quick metabolism of naloxone. Our results as a whole demonstrate that monoclonal antibodies could be a desirable treatment modality for synthetic opioid overdose and possibly opioid use disorder.


Assuntos
Analgésicos Opioides/antagonistas & inibidores , Anticorpos Monoclonais/farmacologia , Antagonistas de Entorpecentes/farmacologia , Dor Nociceptiva/tratamento farmacológico , Transtornos Relacionados ao Uso de Opioides/tratamento farmacológico , Analgésicos Opioides/administração & dosagem , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Camundongos , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/química , Ressonância de Plasmônio de Superfície
15.
Br J Anaesth ; 122(6): e168-e179, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30915997

RESUMO

BACKGROUND: Opioids are potent painkillers but come with serious adverse effects ranging from addiction to potentially lethal respiratory depression. A variety of drugs with separate mechanisms of action are available to prevent or reverse opioid-induced respiratory depression (OIRD). METHODS: The authors reviewed human studies on reversal of OIRD using models that describe and predict the time course of pharmacokinetics (PK) and pharmacodynamics (PD) of opioids and reversal agents and link PK to PD. RESULTS: The PKPD models differ in their basic structure to capture the specific pharmacological mechanisms by which reversal agents interact with opioid effects on breathing. The effect of naloxone, a competitive opioid receptor antagonist, is described by the combined effect-compartment receptor-binding model to quantify rate limitation at the level of drug distribution and receptor kinetics. The effects of reversal agents that act through non-opioidergic pathways, such as ketamine and the experimental drug GAL021, are described by physiological models, in which stimulants act at CO2 chemosensitivity, CO2-independent ventilation, or both. The PKPD analyses show that although all reversal strategies may be effective under certain circumstances, there are conditions at which reversal is less efficacious and sometimes even impossible. CONCLUSIONS: Model-based drug development is needed to design an 'ideal' reversal agent-that is, one that is not influenced by opioid receptor kinetics, does not interfere with opioid analgesia, has a rapid onset of action with long-lasting effects, and is devoid of adverse effects.


Assuntos
Analgésicos Opioides/efeitos adversos , Antagonistas de Entorpecentes/farmacologia , Insuficiência Respiratória/induzido quimicamente , Analgésicos Opioides/antagonistas & inibidores , Corpo Carotídeo/efeitos dos fármacos , Doxapram/farmacologia , Desenho de Fármacos , Humanos , Modelos Biológicos , Naloxona/farmacologia , Insuficiência Respiratória/prevenção & controle , Triazinas/farmacologia
16.
Neurosci Res ; 144: 48-55, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30120960

RESUMO

Chemokines related neuroinflammation and N-methyl-d-aspartate receptor (NMDAR) mediated nociceptive transmission are pivotal determinants in the pathogenesis of opioid-induced hyperalgesia (OIH), but little is known about specific mechanism and treatment. Chemokine CXCL12 with its receptor CXCR4 is implicated in different pathological pain, moreover, neurotoxicity of CXCL12 is associated with NMDAR activation. Recent studies recapitulate the anti-nociception of Annexin 1 (ANXA1) in inflammatory pain. This study examined whether ANXA1 prevented remifentanil-caused OIH through modulating CXCL12 and NMDAR pathway in rats. Acute exposure to remifentanil induced mechanical allodynia and thermal hyperalgesia, which was accompanied by the increase of spinal ANXA1 and CXCL12/CXCR4 expression. Central injection of Anxa12-26 attenuated behavioral OIH in a dose-dependent manner, facilitated ANXA1 production, and inhibited up-regulation of CXCL12/CXCR4 level and NR2B-containing NMDAR phosphorylation. Moreover, pretreatment with AMD3100 reduced hyperalgesia and NR2B-containing NMDAR phosphorylation. Also, exogenous CXCL12 elicited pain hypersensitivity and NMDAR activation in naïve rats, which was reversed by the supplemental delivery of Anxa12-26. These current findings indicate the participation of spinal CXCL12/CXCR4 and NR2B-containing NMDAR pathway in anti-hyperalgesic action of ANXA1 in OIH.


Assuntos
Anexina A1/farmacologia , Quimiocina CXCL12/metabolismo , Receptores CXCR4/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Remifentanil/antagonistas & inibidores , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/farmacologia , Animais , Anexina A1/metabolismo , Benzilaminas , Quimiocina CXCL12/antagonistas & inibidores , Quimiocina CXCL12/biossíntese , Ciclamos , Interações Medicamentosas , Compostos Heterocíclicos/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Masculino , Fragmentos de Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/biossíntese , Remifentanil/farmacologia , Transdução de Sinais/efeitos dos fármacos
17.
J Pharmacol Exp Ther ; 368(1): 88-99, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30401680

RESUMO

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.


Assuntos
Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/antagonistas & inibidores , Cinamatos/uso terapêutico , Comportamento de Procura de Droga/efeitos dos fármacos , Derivados da Morfina/uso terapêutico , Receptores Opioides mu/antagonistas & inibidores , Analgésicos Opioides/metabolismo , Animais , Cinamatos/farmacologia , Comportamento de Procura de Droga/fisiologia , Feminino , Macaca mulatta , Masculino , Derivados da Morfina/farmacologia , Naltrexona/farmacologia , Naltrexona/uso terapêutico , Antagonistas de Entorpecentes/farmacologia , Antagonistas de Entorpecentes/uso terapêutico , Receptores Opioides mu/metabolismo , Autoadministração , Fatores de Tempo
18.
J Pharmacol Exp Ther ; 368(2): 282-291, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30409833

RESUMO

Fentanyl is an extremely potent synthetic opioid that has been increasingly used to adulterate heroin, cocaine, and counterfeit prescription pills, leading to an increase in opioid-induced fatal overdoses in the United States, Canada, and Europe. A vaccine targeting fentanyl could offer protection against the toxic effects of fentanyl in both recreational drug users and others in professions at risk of accidental exposure. This study focuses on the development of a vaccine consisting of a fentanyl-based hapten (F) conjugated to keyhole limpet hemocyanin (KLH) carrier protein or to GMP-grade subunit KLH (sKLH). Immunization with F-KLH in mice and rats reduced fentanyl-induced hotplate antinociception, and in rats reduced fentanyl distribution to the brain compared with controls. F-KLH did not reduce the antinociceptive effects of equianalgesic doses of heroin or oxycodone in rats. To assess the vaccine effect on fentanyl toxicity, rats immunized with F-sKLH or unconjugated sKLH were exposed to increasing subcutaneous doses of fentanyl. Vaccination with F-sKLH shifted the dose-response curves to the right for both fentanyl-induced antinociception and respiratory depression. Naloxone reversed fentanyl effects in both groups, showing that its ability to reverse respiratory depression was preserved. These data demonstrate preclinical selectivity and efficacy of a fentanyl vaccine and suggest that vaccines may offer a therapeutic option in reducing fentanyl-induced side effects.


Assuntos
Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/metabolismo , Fentanila/antagonistas & inibidores , Fentanila/metabolismo , Vacinas/farmacologia , Analgésicos Opioides/toxicidade , Animais , Bradicardia/sangue , Bradicardia/induzido quimicamente , Bradicardia/prevenção & controle , Fentanila/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Sprague-Dawley , Insuficiência Respiratória/sangue , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/prevenção & controle , Vacinas/uso terapêutico
19.
Curr Pain Headache Rep ; 22(11): 72, 2018 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-30151695

RESUMO

PURPOSE OF REVIEW: Testing the efficacy of academic detailing in improving the practice of prescribing naloxone for patients on high-dose opioids. RECENT FINDINGS: Academic detailing has been identified as an effective method for improving health care practices through focused community education. We found that academic detailing is an effective method in improving health care providers' knowledge about the importance of prescribing naloxone for patients on high-dose opioids. We also found that prescribers prescribed more naloxone after our education program. This study reflects the importance of education and academic detailing in resolving health problems. Academic detailing can provide effective preventive tools that can reduce the incidence of health problems we encounter.


Assuntos
Serviços de Saúde Comunitária/organização & administração , Prescrição Inadequada/prevenção & controle , Naloxona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/antagonistas & inibidores , Overdose de Drogas/tratamento farmacológico , Overdose de Drogas/epidemiologia , Prescrições de Medicamentos/normas , Uso de Medicamentos , Educação em Saúde , Pessoal de Saúde/educação , Humanos , Inquéritos e Questionários , Wisconsin/epidemiologia
20.
Eur J Pharmacol ; 833: 275-282, 2018 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-29886241

RESUMO

Fentanyl is an opioid commonly prescribed for cancer pain. Using melanoma-bearing mice, we investigated whether peripheral action would contribute to fentanyl analgesia in cancer pain. Intravenous injection of fentanyl inhibited mechanical nociception in healthy mice, which was markedly inhibited by the opioid antagonist naloxone, but not naloxone methiodide, a peripherally acting opioid antagonist. Melanoma-bearing mice showed mechanical allodynia and spontaneous licking, a pain-related behavior, which were suppressed by intravenous and local injections of fentanyl. Both naloxone and naloxone methiodide inhibited the analgesic effect of intravenous fentanyl to the same degree. Electrophysiological analysis showed that melanoma growth increased the spontaneous and mechanical stimuli-evoked activity of the tibial nerve, which were inhibited by intravenous fentanyl. There was a greater expression of µ- opioid receptors in skin with a melanoma mass than in the contralateral normal skin. In addition, we found µ-opioid receptors in cultured melanoma cells. There was no difference between the number of µ-opioid receptors in the dorsal root ganglia and spinal cord of the melanoma-bearing and contralateral skin side. These results suggest that the analgesic effect of systemic fentanyl is produced via central and peripheral µ- opioid receptors in cancer pain, and cancer cells are a key site of peripheral action.


Assuntos
Analgésicos Opioides/farmacologia , Dor do Câncer/tratamento farmacológico , Fentanila/farmacologia , Hiperalgesia/tratamento farmacológico , Receptores Opioides mu/metabolismo , Potenciais de Ação/efeitos dos fármacos , Analgésicos Opioides/antagonistas & inibidores , Analgésicos Opioides/uso terapêutico , Animais , Dor do Câncer/etiologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Fentanila/antagonistas & inibidores , Fentanila/uso terapêutico , Humanos , Hiperalgesia/etiologia , Injeções Intramusculares , Injeções Intravenosas , Masculino , Melanoma/complicações , Camundongos , Camundongos Endogâmicos C57BL , Naloxona/análogos & derivados , Naloxona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Compostos de Amônio Quaternário/farmacologia , Receptores Opioides , Pele/patologia , Neoplasias Cutâneas/complicações , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Nervo Tibial/efeitos dos fármacos , Nervo Tibial/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...