Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Anal Toxicol ; 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38836589

RESUMEN

BACKGROUND: In recent years, potential therapeutic applications of several different cannabinoids, such as Δ9-tetrahydrocannabinol (Δ9-THC), its isomer Δ8-THC and Δ9-tetrahydrocannabivarin (Δ9-THCV), have been investigated. Nevertheless, to establish dose-effect relationship and to gain knowledge of their pharmacokinetics and metabolism, sensitive and specific analytical assays are needed to measure these compounds in patients. For this reason, we developed and validated an online extraction high-performance liquid chromatography- tandem mass spectrometry (LC/LC-MS/MS) method for the simultaneous quantification of 13 cannabinoids and metabolites including the Δ8 and Δ9 isomers of THC, THCV and those of their major metabolites in human plasma. METHODS: Plasma was fortified with cannabinoids at varying concentrations within the working range of the respective compound and 200 µL were extracted using a simple one-step protein precipitation procedure. The extracts were analyzed using online trapping LC/LC-atmospheric pressure chemical ionization (APCI)-MS/MS running in the positive multiple reaction monitoring (MRM) mode. RESULTS: The lower limit of quantification ranged from 0.5 to 2.5 ng/mL and the upper limit of quantification was 400 ng/mL for all analytes. Inter-day analytical accuracy and imprecision ranged from 82.9 to 109% and 4.3 to 20.3% (coefficient of variance), respectively. Of 534 plasma samples following controlled oral administration of Δ8-THCV, 236 were positive for Δ8-THCV (median; interquartile ranges: 3.5 ng/mL; 1.8 - 11.9 ng/mL), 383 for the major metabolite (-)-11-nor-9-carboxy-Δ8-tetrahydrocannabivarin (Δ8-THCV-COOH) (95.4 ng/mL; 20.7 - 328 ng/mL), 260 for (-)-11-nor-9-carboxy-Δ9-tetrahydrocannabivarin (Δ9-THCV-COOH) (5.8 ng/mL; 2.5 - 16.1 ng/mL), 157 for (-)-11-hydroxy-Δ8-tetrahydrocannabivarin (11-OH-Δ8-THCV) (1.7 ng/mL; 1.0 - 3.7 ng/mL), 49 for Δ8-THC-COOH (1.7 ng/mL; 1.4 - 2.3 ng/mL) and 42 for Δ9-THCV (1.3 ng/mL; 0.8 - 1.6 ng/mL). CONCLUSIONS: We developed and validated the first LC/LC-MS/MS assay for the specific quantification of Δ8-THC, Δ9-THC and THCV isomers and their respective metabolites in human plasma. Δ8-THCV-COOH, 11-hydroxy-Δ8-THCV and Δ9-THCV-COOH were the major Δ8-THCV metabolites in human plasma after oral administration of 98.6% pure Δ8-THCV.

2.
Psychopharmacology (Berl) ; 241(6): 1227-1236, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38383903

RESUMEN

RATIONALE: Understanding mechanisms of drug use decisions will inform the development of treatments for opioid use disorder (OUD). Decision-making experiments using neurobehavioral approaches require many trials or events of interest for statistical analysis, but the pharmacokinetics of most opioids limit dosing in humans. OBJECTIVES: This experiment characterized the effects of repeated infusions of the ultra-short acting opioid remifentanil in people with OUD and physical opioid dependence. METHODS: An inpatient study using a within-subjects, single-blind, escalating, within-session, pre-post design was conducted. Seven (3 female) subjects were maintained on oral oxycodone (40-60 mg, 4x/day = 160-240 total mg/day) for seven days prior to the dose-ranging session. Subjects received infusions of three ascending remifentanil doses (0.03, 0.1, 0.3 mcg/kg/infusion in 2 subjects; 0.1, 0.3, 1.0 mcg/kg/infusion in 5 subjects) every minute for 40 min per dose, with infusions administered over 5 s to model naturalistic delivery rates. End tidal carbon dioxide, respiration rate, oxygen saturation (SpO2) and heart rate were measured continuously. Blood pressure (BP), pupil diameter and self-reported drug effects were measured every 5 min. RESULTS: Pupil diameter, SpO2 and systolic BP decreased, and ratings on prototypic subjective effects questionnaire items increased, as a function of remifentanil dose. The number of infusions held because of sedation or physiological parameters exceeding predetermined cutoffs also increased with dose. CONCLUSIONS: This experiment established doses and procedures for the safe delivery of rapid, repeated remifentanil infusions to individuals with OUD and physical fentanyl dependence, which can be applied to the mechanistic study of opioid use decisions.


Asunto(s)
Analgésicos Opioides , Presión Sanguínea , Relación Dosis-Respuesta a Droga , Fentanilo , Frecuencia Cardíaca , Trastornos Relacionados con Opioides , Piperidinas , Remifentanilo , Humanos , Remifentanilo/administración & dosificación , Remifentanilo/farmacología , Femenino , Masculino , Adulto , Trastornos Relacionados con Opioides/tratamiento farmacológico , Fentanilo/administración & dosificación , Fentanilo/farmacocinética , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacocinética , Piperidinas/administración & dosificación , Piperidinas/farmacocinética , Piperidinas/farmacología , Método Simple Ciego , Frecuencia Cardíaca/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Infusiones Intravenosas , Persona de Mediana Edad , Autoinforme , Adulto Joven , Oxicodona/administración & dosificación , Oxicodona/farmacocinética
3.
Cannabis Cannabinoid Res ; 8(S1): S71-S82, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37721990

RESUMEN

Introduction: Tetrahydrocannabivarin (THCV) is an understudied cannabinoid that appears to have effects that vary as a function of dose. No human study has evaluated the safety and nature of effects in a wide range of THCV doses. Methods: This was a two-phase, dose-ranging, placebo-controlled trial of the Δ8 isomer of oral THCV in healthy adults. Phase 1 utilized an unblinded, single-ascending dose design (n=3). Phase 2 used a double-blind, randomized, within-participant crossover design (n=18). Participants received single acute doses of placebo and 12.5, 25, 50, 100, and 200 mg of THCV. Safety measures and subjective and cognitive effects were assessed predose and up to 8 h postdose. Results: Most adverse events (AEs; 55/60) were mild. Euphoric mood was the most common AE. The 12.5, 25, and 200 mg doses produced significantly lower minimum times to complete the digit vigilance test (ps=0.01). The 25 mg dose showed elevations on mean ratings of "energetic" at 1-, 2-, and 4-h postdose, but the maximum postdose rating for this dose did not achieve statistical significance relative to placebo ([95% confidence interval]=3.2 [-0.5 to 6.9], p=0.116). The 100 and 200 mg doses showed elevations on ratings of "feel a drug effect" and "like the drug effect." Almost all urine drug screens (78/79) at 8 h postdose in the active THCV conditions tested positive for tetrahydrocannabinol (THC). Conclusion: All THCV doses displayed a favorable safety profile. Several THCV doses showed a preliminary signal for improved sustained attention, but the effect was not dose dependent. Though mild and not associated with impairment, THC-like effects were observed at higher THCV doses. Oral THCV-containing products could lead to positive urine drug screens for THC. ClinicalTrials.gov ID: NCT05210634.


Asunto(s)
Cannabinoides , Emociones , Adulto , Humanos , Voluntarios Sanos , Método Doble Ciego , Euforia
5.
Subst Use Misuse ; 58(1): 66-76, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36453437

RESUMEN

Background and objectives: Cannabis is the most used federally illicit substance. Due to widespread medicinal use and state-level legalization, public perceptions of cannabis have shifted toward the assumption that cannabis is safe. However, cannabinoids can cause adverse medical complications that may lead people to seek treatment. This study characterized cannabinoid poisoning-related medical encounters, poisoning involving cannabinoids and other psychoactive substances, and cannabinoid poisoning-related cardiac complications. Methods: Administrative billing data for emergency department visits and inpatient hospitalizations in acute care facilities with a discharge date from January 1, 2017 to December 31, 2019 were used to characterize cannabinoid poisoning events in Kentucky, identified by ICD-10-CM diagnosis code T40.7X. Results: There were 1,490 encounters of cannabinoid poisoning; patients were primarily non-Hispanic White males, ages 15-44, who had Medicaid and lived in a metropolitan area. Of those, 31.21% involved poisoning with a second psychoactive substance, primarily stimulants and/or opioids, and 17.72% experienced a cardiac complication. Cannabinoid-polydrug poisoning was associated with inpatient treatment (χ2=199.18, p < 0.001) and cardiac complications (χ2=4.58, p < 0.001). Discussion and Conclusions: These results are consistent with other state-level data. Patients who were diagnosed with cannabis-polydrug poisoning, compared to cannabis alone poisoning, had greater odds of hospital admission and cardiac complications, and longer length of hospital stays. Scientific Significance: The health risks of cannabinoid use must be more broadly recognized, while timely and accurate data need to be shared to guide policies on cannabis access. Future research on cannabinoid poisoning should consider the involvement of other psychoactive drugs.


Asunto(s)
Cannabinoides , Cannabis , Alucinógenos , Masculino , Estados Unidos , Humanos , Adolescente , Adulto Joven , Adulto , Cannabinoides/efectos adversos , Kentucky/epidemiología , Pacientes Internos , Cannabis/efectos adversos , Hospitalización , Servicio de Urgencia en Hospital
6.
Drug Alcohol Depend ; 237: 109522, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35690015

RESUMEN

BACKGROUND: Regulation has not kept pace with the growth of the hemp-derived CBD market. We have evaluated the risk of Δ9-tetrahydrocannabinol (Δ9-THC) contamination in 80 unregulated products with comparison to a regulated control, Epidiolex®. METHODS: Local and national brands of hemp-derived oil products were purchased online and from local retailers in central Kentucky (which carry both national and local brands). These were extracted by solvent extraction and quantified by liquid-chromatography tandem mass-spectrometry (LC-MS/MS) using a validated method. RESULTS: Of the 80 unregulated products and Epidiolex®, Δ9-THC was detected above the limit of quantification (LOQ = 0.005 mg/mL) of the assay in 52 samples, ranging from 0.008 mg/mL to 2.071 mg/mL. Twenty-one of the products tested were labelled as "THC-Free", and 5 of these products contained detectable levels of Δ9-THC ranging from 0.015 mg/mL to 0.656 mg/mL. CONCLUSIONS: Consumers are taking hemp-derived CBD products without understanding the risks of unintentional consumption of Δ9-THC. This accidental use of Δ9-THC could have adverse effects on health and safety as well as potential legal consequences (e.g., child custody, impaired driving), as Δ9-THC drug test findings could impact employment, military, and sport eligibility status.


Asunto(s)
Cannabidiol , Dronabinol , Niño , Cromatografía Liquida/métodos , Dronabinol/análisis , Humanos , Detección de Abuso de Sustancias , Espectrometría de Masas en Tándem/métodos
7.
J Cannabis Res ; 4(1): 28, 2022 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-35658956

RESUMEN

BACKGROUND: The legalization of hemp in the USA has led to tremendous growth in the availability of hemp-derived products, particularly cannabidiol (CBD) products. The lack of regulatory oversight in this industry has resulted in the marketing and sale of CBD products with questionable ingredients and quality. The aim of the current study was to examine the CBD content in 80 commercially available hemp-derived CBD products purchased from online and local retailers. Epidiolex® was also included in the study as a positive control. METHODS: Hemp-derived CBD products were selected to represent products readily available to residents of Central Kentucky. The samples were comprised of local and national brands produced in a variety of locations inside and outside of Kentucky. The products were analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS), and the analytical findings were compared to the label claims for CBD content. Descriptive statistics and normal-based confidence intervals were calculated using Microsoft Excel. RESULTS: The label claims for CBD content ranged from 7.5 to 60 mg/mL, while LC-MS/MS analysis detected a range of 2.9 to 61.3 mg/mL. Of the 80 products evaluated, 37 contained CBD concentrations that were at least ± 10% different than the concentration listed on the label (range of 0.9 to 30.6 mg/mL from label claim) - 12 products contained < 90%, while 25 products contained > 110%. The degree of concordance for the samples tested using ± 10% tolerance from label claim was 54%. CONCLUSIONS: These data suggest that additional regulation is required to ensure label accuracy as nearly half of the products in this study were not properly labelled (i.e., not within a ± 10% margin of error). Consumers and practitioners should remain cautious of unregulated and often-mislabeled CBD products due to the risks of taking too much CBD (e.g., drug-drug interactions, liver enzyme elevations, increased side effects) and the consequences of taking too little (e.g., no clinical benefits due to underdosing). The results of this study support the continued need for good manufacturing practices and testing standards for CBD products.

8.
Cannabis Cannabinoid Res ; 6(5): 362-365, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34662224

RESUMEN

Delta-8-tetrahydrocannabinol (Δ8-THC) is chemically and functionally similar to delta-9-tetrahydrocannabinol (Δ9-THC) (the primary psychoactive cannabinoid in the cannabis plant) and is currently widely available "over-the-counter" across the United States due to unregulated sales. However, these products have a questionable legal status based on current U.S. laws, as Δ8-THC is considered a Schedule I drug by the federal Drug Enforcement Administration (DEA). Despite this designation, Δ8-THC products (e.g., gummies, edibles, oils, and vapes) are largely unregulated and are sold in gas stations, online, and other marketplaces (most often outside of authorized dispensaries) and are marketed as legal hemp products. This problem arises from a purposeful misinterpretation of the 2018 Farm Bill, which some interpret as legalization of non-Δ9-THC cannabinoids (notably, Δ8-THC). The widespread availability of Δ8-THC products has not been without health consequences. The lack of regulation means that there are no required warning labels or packaging protections in place and no mandated laboratory analysis to assure label accuracy or product purity. As Δ8-THC produces physiological and toxicological effects that are similar to Δ9-THC, high-dose exposure of Δ8-THC (e.g., consuming a full bag of Δ8-THC gummies) has resulted in recent reports of medical emergencies, including calls to poison control centers and presentations to emergency departments, with some pediatric patients arriving unconscious and unresponsive. Several states and regulatory agencies have called for legislation to regulate Δ8-THC, but little progress has occurred nationally thus far.


Asunto(s)
Cannabinoides , Cannabis , Cannabis/efectos adversos , Niño , Dronabinol/análisis , Humanos , Etiquetado de Productos , Embalaje de Productos , Estados Unidos
9.
Psychopharmacology (Berl) ; 238(9): 2503-2514, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34106317

RESUMEN

AIMS: Intravenous (IV) misuse of the µ opioid analgesic oxymorphone has caused significant public health harms; however, no controlled data on its IV abuse potential are available. The primary aims of this pilot study were to directly compare IV oxymorphone to IV oxycodone, morphine, and hydromorphone on a subjective measure of drug liking and to assess relative potency. METHODS: Participants (n = 6) with opioid use disorder, physical dependence, and current IV use completed this two-site, within-subject, double-blind, placebo-controlled, inpatient pilot study. During each session, one IV dose (mg/70 kg) was administered: oxymorphone (1.8, 3.2, 5.6, 10, 18, 32), hydromorphone (1.8, 3.2, 5.6, 10, 18), oxycodone (18, 32, 56), morphine (18, 32), and placebo. Data were collected before and for 6 h after dosing. Primary outcomes included safety/physiological effects, subjective reports of drug liking, and relative potency estimates. RESULTS: All active test drugs produced prototypical, dose-related µ opioid agonist effects (e.g., miosis). Oxymorphone was more potent than the comparator opioids on several measures, including drug liking and respiratory depression (p < 0.05). Across abuse-related subjective outcomes, oxymorphone was 2.3-2.8-fold more potent than hydromorphone and 12.5-14-fold more potent than oxycodone (p < 0.05). CONCLUSIONS: Despite the relatively small sample size, this pilot study detected robust oxymorphone effects. Oxymorphone was far more potent than the comparator opioids, particularly on abuse potential outcomes. Overall, these findings may help explain surveillance reports that demonstrate, after adjusting for prescription availability, oxymorphone is injected at the highest frequency, relative to other prescription opioids.


Asunto(s)
Trastornos Relacionados con Opioides , Oximorfona , Analgésicos Opioides/efectos adversos , Humanos , Trastornos Relacionados con Opioides/tratamiento farmacológico , Oxicodona , Oximorfona/efectos adversos , Proyectos Piloto
10.
Front Psychiatry ; 12: 805002, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35069295

RESUMEN

Appalachian Kentucky was at the epicenter of the prescription opioid epidemic in the early 2000's. As we enter the third decade of the epidemic, patterns have begun to emerge as people who use drugs (PWUD) transition from use of opioids to other drugs. The purpose of this analysis was to examine longitudinal changes in methamphetamine use in an ongoing cohort of rural people who use drugs (PWUD) in Appalachian Kentucky. All but five of the cohort participants (N = 503) reported nonmedical prescription opioid use (NMPOU) at baseline and those 498 are included in this longitudinal analysis encompassing eight waves of data (2008-2020). Past 6-month use of methamphetamine was the dependent variable. Given the correlated nature of the data, mixed effects logistic regression was utilized to examine changes in methamphetamine use over time. Significant increases in methamphetamine use were observed over the past decade in this cohort of PWUD, especially in recent years (2017-2020). Prevalence of recent use at baseline and each of the follow-up visits was as follows: 9.4, 5.6, 5.0, 5.4, 8.1, 6.8, 6.9, and 33.1%, respectively (p < 0.001). On the contrary, significant reductions in NMPO and heroin use were observed in the same time period. The odds of methamphetamine use at the most recent visit were 25.8 times greater than at baseline (95% CI: 14.9, 44.6) and 52.6% of those reporting methamphetamine use reported injecting the drug. These results provide further evidence of "twin epidemics" of methamphetamine use among NMPOU. While problematic on several fronts, of particular concern is the lack of effective treatment options for methamphetamine use disorder. As policies around the opioid epidemic continue to evolve, particular attention should be paid to the surge in stimulant use in opioid-endemic areas.

11.
Psychopharmacology (Berl) ; 238(1): 9-28, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33221931

RESUMEN

RATIONALE: Cannabidiol (CBD) products lacking regulatory approval are being used to self-treat a myriad of conditions and for their unsubstantiated health benefits. The scientific evidence supporting these claims largely arises not from controlled clinical trials, but from the recognition that CBD has numerous biological targets. Yet, CBD is commonly consumed and often in over-the-counter products that are unapproved and of unknown composition. Epidiolex® is the only product that has undergone rigorous pharmacokinetic assessment and testing in clinical trials; it was approved as a non-scheduled drug by the U.S. Food and Drug Administration for the treatment of intractable childhood-onset seizures. However, studies investigating CBD for other medical conditions are limited in number and often lack the scientific rigor, controls, or sample sizes required to draw clinically meaningful conclusions. Although Epidiolex® is safe for human consumption, recent changes in regulation of commercially available CBD products have resulted in limited quality control and products marketed with unknown CBD bioavailability. Even scientifically rigorous studies have used different sources of CBD and different suspension vehicles for administration, making it difficult to compare results among studies and resolve mixed outcomes. OBJECTIVES: This paper reviews the molecular targets, pharmacokinetics, and safety and abuse liability of CBD; additionally, the extant evidence on its potential therapeutic effects for neurological disorders, pain, inflammation, conditions related to immune function, psychiatric disorders, and substance use are described.


Asunto(s)
Cannabidiol , Trastornos Mentales/tratamiento farmacológico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Dolor/tratamiento farmacológico , Cannabidiol/efectos adversos , Cannabidiol/farmacocinética , Cannabidiol/farmacología , Niño , Humanos , Trastornos Relacionados con Sustancias/tratamiento farmacológico , Estados Unidos , United States Food and Drug Administration
12.
Psychopharmacology (Berl) ; 238(2): 539-549, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33169203

RESUMEN

RATIONALE: Epidemiological data indicate that drivers testing positive for an opioid drug are twice as likely to cause a fatal car crash; however, there are limited controlled data available. OBJECTIVES: The primary aim of this study was to assess the effects of a therapeutic dose range of oxycodone alone and in combination with alcohol on simulated driving performance. METHODS: Healthy participants (n = 10) completed this within-subject, double-blind, placebo-controlled, randomized outpatient study. Six 7-h sessions were completed during which oxycodone (0, 5, 10 mg, p.o.) was administered 30 min before alcohol (0, 0.8 g/kg (15% less for women), p.o.) for a total of 6 test conditions. Driving assessments and participant-, observer-rated, psychomotor and physiological measures were collected in regular intervals before and after drug administration. RESULTS: Oxycodone alone (5, 10 mg) did not produce any changes in driving outcomes or psychomotor task performance, relative to placebo (p > 0.05); however, 10 mg oxycodone produced increases in an array of subjective ratings, including sedation and impairment (p < 0.05). Alcohol alone produced driving impairment (e.g., decreased lateral control) (p < 0.05); however, oxycodone did not potentiate alcohol-related driving or subjective effects. CONCLUSIONS: These preliminary data suggest that acute doses of oxycodone (5, 10 mg) do not significantly impair acuity on laboratory-based simulated driving models; however, 10 mg oxycodone produced increases in self-reported outcomes that are not compatible with safe driving behavior (e.g., sedation, impairment). Additional controlled research is needed to determine how opioid misuse (higher doses; parenteral routes of administration) impacts driving risk.


Asunto(s)
Conducir bajo la Influencia/psicología , Etanol/efectos adversos , Modelos Psicológicos , Oxicodona/efectos adversos , Desempeño Psicomotor/efectos de los fármacos , Adulto , Estudios Cruzados , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Sinergismo Farmacológico , Etanol/administración & dosificación , Femenino , Humanos , Masculino , Trastornos Relacionados con Opioides/psicología , Oxicodona/administración & dosificación
13.
Eur Neuropsychopharmacol ; 36: 206-216, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32273144

RESUMEN

The endogenous opioid and cannabinoid receptor systems are widely distributed and co-localized throughout central and peripheral nervous system regions. A large body of preclinical evidence suggests that there are functional interactions between these two systems that may be leveraged to address various health conditions. Numerous animal studies have shown that cannabinoid agonists (e.g., delta-9-tetrahydrocannabinol [Δ9-THC]) enhance the analgesic effects of µ-opioid analgesics as evidenced by decreasing the opioid dose required for analgesia (i.e., opioid sparing) and extending the duration of the opioid analgesia. In contrast, controlled human laboratory studies and clinical trials have not demonstrated robust analgesic or opioid-sparing effects from opioid-cannabinoid combinations. Meta-analyses of the literature (clinical trials, controlled laboratory studies; some non-controlled studies/case reports) have examined the effects of cannabis/cannabinoids for pain relief in those taking a wide variety of analgesics, including prescription opioid medications. These data do not strongly support the use of cannabinoids for chronic pain nor do prospective studies demonstrate significant cannabinoid-mediated opioid-sparing effects. Preclinical studies have also suggested a role for cannabinoids for the treatment of opioid withdrawal. Controlled laboratory and clinical studies suggest that there may be a modest signal for Δ9-THC to suppress some opioid signs and symptoms but they are not completely ameliorated and there may also be concerns around safety of Δ9-THC administration in a state of heightened autonomic arousal as occurs with opioid withdrawal. Despite anecdotal and correlational reports suggesting a benefit of cannabis on reducing opioid overdose, there is no strong data supporting this contention and emerging reports have conflicting results. In summary, there is a groundswell of public advocacy supporting the use of cannabis and cannabinoids to replace opioid analgesics or to reduce opioid use; however, the extant controlled clinical data do not support the role of cannabinoids for opioid replacement or opioid-sparing effects when treating opioid use disorder or chronic pain.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Agonistas de Receptores de Cannabinoides/administración & dosificación , Dolor Crónico/tratamiento farmacológico , Dronabinol/administración & dosificación , Manejo del Dolor/métodos , Animales , Dolor Crónico/diagnóstico , Dolor Crónico/epidemiología , Ensayos Clínicos como Asunto/métodos , Humanos , Manejo del Dolor/tendencias , Estudios Prospectivos
14.
Addict Biol ; 25(4): e12799, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31240842

RESUMEN

Preclinical data indicate that selective kappa opioid receptor antagonists reduce nicotine self-administration and withdrawal symptoms. The aim of the current study was to determine whether treatment with CERC-501, an orally available, potent, and selective kappa opioid receptor antagonist, could alleviate nicotine withdrawal and craving and mitigate mood alterations associated with nicotine withdrawal in humans. Healthy, adult cigarette smokers were enrolled into this randomized, multisite, double-blind, placebo-controlled, crossover study. Participants completed two 8-day treatment phases during which they received either CERC-501 (15 mg, p.o., once daily) or placebo. On the seventh day of each dosing phase, participants were admitted as inpatients for an 18-hour cigarette abstinence period followed by experimental testing. The primary outcome measures were (a) performance on the McKee Smoking Lapse test (ie, latency to smoke in exchange for money) and (b) number of cigarettes self-administered during a 60-minute ad lib smoking period. Other outcomes included measures of craving, mood, anxiety, nicotine withdrawal, and subjective effects of cigarette smoking. A total of 71 participants who smoked an average of approximately 23 cigarettes per day were enrolled, and 56 subjects completed the study. CERC-501 was well tolerated, but it did not significantly alter the latency to start smoking (CERC-501: 16.5 min vs placebo: 17.7 min) or the number of cigarettes smoked (CERC-501: 3.3 cigarettes vs placebo: 3.1 cigarettes). Compared with placebo, CERC-501 also did not affect cigarette craving, mood, anxiety, nicotine withdrawal, or subjective effects of smoking. These findings do not support a role for CERC-501 in the treatment of nicotine use disorder.


Asunto(s)
Benzamidas/farmacología , Fumar Cigarrillos/metabolismo , Antagonistas de Narcóticos/farmacología , Pirrolidinas/farmacología , Síndrome de Abstinencia a Sustancias/metabolismo , Tabaquismo/metabolismo , Adulto , Afecto/efectos de los fármacos , Ansiedad/fisiopatología , Ansia/efectos de los fármacos , Estudios Cruzados , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nicotina/efectos adversos , Agonistas Nicotínicos/efectos adversos , Distribución Aleatoria , Receptores Opioides kappa/antagonistas & inhibidores , Cese del Hábito de Fumar , Síndrome de Abstinencia a Sustancias/etiología , Síndrome de Abstinencia a Sustancias/fisiopatología , Tabaquismo/fisiopatología
15.
Psychopharmacology (Berl) ; 236(11): 3341-3352, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31201479

RESUMEN

RATIONALE: Dozens of preclinical studies have reported cannabinoid agonist potentiation of the analgesic effects of µ-opioid agonists. OBJECTIVES: The aim of this study was to determine if a cannabinoid agonist could potentiate opioid analgesia in humans using several laboratory pain models. METHODS: Healthy participants (n = 10) with/out current drug use/pain conditions completed this within-subject, double-blind, placebo-controlled, randomized outpatient study. Nine 8-h sessions were completed during which dronabinol (0, 2.5, 5 mg, p.o.) was administered 1 h before oxycodone (0, 5, 10 mg, p.o.) for a total of 9 test conditions. Outcomes included sensory threshold and tolerance from four experimental pain models (cold pressor, pressure algometer, hot thermode, cold hyperalgesia), along with participant- and observer-rated, performance and physiological effects. RESULTS: Oxycodone produced miosis (p < 0.05) and analgesic responses (e.g., pressure algometer [p < 0.05]), while dronabinol did not (p > 0.05). Depending on the dose combination, dronabinol attenuated or did not alter oxycodone analgesia; for example, dronabinol (2.5 mg) decreased the analgesic effects of oxycodone (10 mg) on pressure tolerance. Conversely, dronabinol increased oxycodone subjective effects (e.g., drug liking) (p < 0.05); oxycodone (5 mg) ratings of "high" were potentiated by 5 mg dronabinol (p < 0.05; placebo = 1.1 [± 0.7]; 5 mg oxycodone = 4.7 [± 2.2]; 5 mg dronabinol = 9.9 [± 8.4]; 5 mg oxycodone + 5 mg dronabinol = 37.4 [± 11.3]). CONCLUSIONS: This study indicates that dronabinol did not enhance the analgesic effects of oxycodone and increased abuse- and impairment-related subjective effects. These data suggest that dronabinol may not be an effective or appropriate opioid adjuvant; it could potentially increase opioid dose requirements, while increasing psychoactive opioid effects.


Asunto(s)
Analgesia/métodos , Analgesia/psicología , Analgésicos Opioides/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/psicología , Adolescente , Adulto , Analgésicos Opioides/uso terapéutico , Agonistas de Receptores de Cannabinoides/uso terapéutico , Estudios Cruzados , Autoevaluación Diagnóstica , Método Doble Ciego , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Dolor/tratamiento farmacológico , Dolor/psicología , Dimensión del Dolor/métodos , Desempeño Psicomotor/efectos de los fármacos , Desempeño Psicomotor/fisiología , Adulto Joven
16.
Drug Alcohol Depend ; 192: 59-66, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30218864

RESUMEN

BACKGROUND: It is common for cannabis users to also use tobacco. While data suggest that tobacco users have more difficulty achieving cannabis cessation, secondary analyses of clinical trial data sets may provide insight into the moderating variables contributing to this relationship, as well as changes in tobacco use during cannabis treatment. Those were the aims of this secondary analysis. METHODS: The parent study was a multi-site trial of N-acetylcysteine for cannabis dependence conducted within the National Drug Abuse Treatment Clinical Trials Network. Participants were treatment-seeking adults (ages 18-50) who met criteria for cannabis dependence (N = 302). For cigarette smokers (n = 117), tobacco use was assessed via timeline follow-back and nicotine dependence was assessed via the Fagerström Test for Nicotine Dependence (FTND). Outcome measures included: 1) changes in tobacco use based on treatment assignment, nicotine dependence, and concurrent cannabis reduction/abstinence, and 2) independent associations between nicotine dependence and cannabis abstinence. RESULTS: Cigarette smokers accounted for 39% of the sample (117/302), with a median FTND score of 3.0 (10-point scale). Among those with lower baseline nicotine dependence scores, cigarette smoking was reduced in the active treatment group compared to placebo. Those with moderate/high levels of nicotine dependence showed slight increases in smoking following active treatment. Nicotine dependence did not affect cannabis cessation. CONCLUSIONS: Cigarette smoking during cannabis treatment was affected, but depended on baseline nicotine dependence severity, though dependence levels did not impact cannabis abstinence. Interventions that address both tobacco and cannabis are needed, especially due to an increasing prevalence of cannabis use.


Asunto(s)
Fumar Cigarrillos/epidemiología , Abuso de Marihuana/tratamiento farmacológico , Abuso de Marihuana/epidemiología , Cese del Hábito de Fumar/métodos , Tabaquismo/epidemiología , Acetilcisteína/uso terapéutico , Adolescente , Adulto , Fumar Cigarrillos/psicología , Femenino , Humanos , Masculino , Abuso de Marihuana/psicología , Marihuana Medicinal/uso terapéutico , Persona de Mediana Edad , Cese del Hábito de Fumar/psicología , Uso de Tabaco/epidemiología , Uso de Tabaco/psicología , Tabaquismo/diagnóstico , Tabaquismo/psicología , Adulto Joven
17.
Drug Alcohol Depend ; 177: 249-257, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28623823

RESUMEN

BACKGROUND: Cannabis use disorder (CUD) is a prevalent and impairing condition, and established psychosocial treatments convey limited efficacy. In light of recent findings supporting the efficacy of N-acetylcysteine (NAC) for CUD in adolescents, the objective of this trial was to evaluate its efficacy in adults. METHODS: In a 12-week double-blind randomized placebo-controlled trial, treatment-seeking adults ages 18-50 with CUD (N=302), enrolled across six National Drug Abuse Treatment Clinical Trials Network-affiliated clinical sites, were randomized in a 1:1 ratio to a 12-week course of NAC 1200mg (n=153) or placebo (n=149) twice daily. All participants received contingency management (CM) and medical management. The primary efficacy measure was the odds of negative urine cannabinoid tests during treatment, compared between NAC and placebo participants. RESULTS: There was not statistically significant evidence that the NAC and placebo groups differed in cannabis abstinence (odds ratio=1.00, 95% confidence interval 0.63-1.59, p=0.984). Overall, 22.3% of urine cannabinoid tests in the NAC group were negative, compared with 22.4% in the placebo group. Many participants were medication non-adherent; exploratory analysis within medication-adherent subgroups revealed no significant differential abstinence outcomes by treatment group. CONCLUSIONS: In contrast with prior findings in adolescents, there is no evidence that NAC 1200mg twice daily plus CM is differentially efficacious for CUD in adults when compared to placebo plus CM. This discrepant finding between adolescents and adults with CUD may have been influenced by differences in development, cannabis use profiles, responses to embedded behavioral treatment, medication adherence, and other factors.


Asunto(s)
Acetilcisteína/uso terapéutico , Abuso de Marihuana/diagnóstico , Abuso de Marihuana/tratamiento farmacológico , Adolescente , Adulto , Cannabis , Método Doble Ciego , Femenino , Depuradores de Radicales Libres/uso terapéutico , Humanos , Masculino , Abuso de Marihuana/psicología , Fumar Marihuana/tratamiento farmacológico , Fumar Marihuana/psicología , Cumplimiento de la Medicación/psicología , Sulpirida , Resultado del Tratamiento , Adulto Joven
18.
Drug Alcohol Depend ; 172: 9-13, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-28088032

RESUMEN

BACKGROUND: Cannabidiol (CBD) is a naturally occurring constituent of the marijuana plant. In the past few years, there has been great interest in the therapeutic effects of isolated CBD and it is currently being explored for numerous disease conditions (e.g., pain, epilepsy, cancer, various drug dependencies). However, CBD remains a Schedule I drug on the U.S. Controlled Substances Act (CSA). Despite its status, there are no well-controlled data available regarding its abuse liability. METHODS: Healthy, frequent marijuana users (n=31) were enrolled in this within subject, randomized, placebo-controlled, double-blind, multisite study that administered oral cannabidiol (0, 200, 400, 800mg) alone and in combination with smoked marijuana (0.01%, 5.3-5.8% THC). Participants received one dose combination across 8 once-weekly outpatient sessions (7.5h). The primary findings on the drug interaction effects were previously reported (Haney et al., 2016). The present study is a secondary analysis of the data to examine the abuse liability profile of oral cannabidiol (200, 400, 800mg) in comparison to oral placebo and active smoked marijuana (5.3-5.8% THC). RESULTS: Active marijuana reliably produced abuse-related subjective effects (e.g., high) (p<0.05). However, CBD was placebo-like on all measures collected (p>0.05). CONCLUSIONS: Overall, CBD did not display any signals of abuse liability at the doses tested and these data may help inform U.S. regulatory decisions regarding CBD schedule on the CSA.


Asunto(s)
Cannabidiol/farmacología , Abuso de Marihuana/psicología , Fumar Marihuana/psicología , Administración Oral , Adulto , Método Doble Ciego , Interacciones Farmacológicas , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Desempeño Psicomotor/efectos de los fármacos , Adulto Joven
19.
Curr Top Behav Neurosci ; 34: 33-58, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27356522

RESUMEN

While opioids are very effective analgesics for treating acute pain, humans have struggled with opiate addiction for millenia. An opium abuse epidemic in the early 1900's led the US government to develop a systematic research infrastructure and scientific plan to produce new compounds with analgesic properties but without abuse liability. This review describes the techniques that were developed for testing in the human laboratory, including empirically derived outcome measures and required elements for human abuse potential assessment. The evaluation and characterization of semi-synthetic and synthetic opioids, including full mu opioid agonists, partial agonists and mixed agonist-antagonists, are described across several decades of research. Finally, the prescription opioid epidemic beginning in the 1990's in the US led to a resurgence in abuse potential evaluations, and the application of these methods to the study of novel abuse-deterrent formulations is discussed.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Trastornos Relacionados con Opioides/prevención & control , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Buprenorfina/uso terapéutico , Química Farmacéutica , Humanos , Estados Unidos
20.
J Clin Psychopharmacol ; 36(4): 324-32, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27300254

RESUMEN

RATIONALE: Accurate assessment of medication adherence is critical for determination of medication efficacy in clinical trials, but most current methods have significant limitations. This study tests a subtherapeutic (microdose) of acetazolamide as a medication ingestion marker because acetazolamide is rapidly absorbed and excreted without metabolism in urine and can be noninvasively sampled. METHODS: In a double-blind, placebo-controlled, residential study, 10 volunteers received 15 mg oral acetazolamide for 4 consecutive days. Acetazolamide pharmacokinetics were assessed on day 3, and its pharmacokinetic and pharmacodynamic interactions with a model medication (30 mg oxycodone) were examined on day 4. The rate of acetazolamide elimination into urine was followed for several days after dosing cessation. RESULTS: Erythrocyte sequestration (half-life = 50.2 ± 18.5 h, mean ± SD, n = 6), resulted in the acetazolamide microdose exhibiting a substantially longer plasma half-life (24.5 ± 5.6 hours, n = 10) than previously reported for therapeutic doses (3-6 hours). After cessation of dosing, the rate of urinary elimination decreased significantly (F3,23 = 247: P < 0.05, n = 6) in a predictable manner with low intersubject variability and a half-life of 16.1 ± 3.8 h (n = 10). For each of 4 consecutive mornings after dosing cessation, the rates of urinary acetazolamide elimination remained quantifiable.There was no overall effect of acetazolamide on the pharmacodynamics, Cmax, Tmax, or elimination half-life of the model medication tested. Acetazolamide may have modestly increased overall oxycodone exposure (20%, P < 0.05) compared with one of the 2 days when oxycodone was given alone, but there were no observed effects of acetazolamide on oxycodone pharmacodynamic responses. CONCLUSIONS: Coformulation of a once-daily trial medication with an acetazolamide microdose may allow estimation of the last time of medication consumption for up to 96 hours postdose. Inclusion of acetazolamide may therefore provide an inexpensive new method to improve estimates of medication adherence in clinical trials.


Asunto(s)
Acetazolamida/farmacología , Acetazolamida/farmacocinética , Inhibidores de Anhidrasa Carbónica/farmacología , Inhibidores de Anhidrasa Carbónica/farmacocinética , Cumplimiento de la Medicación , Narcóticos/farmacología , Oxicodona/farmacología , Acetazolamida/administración & dosificación , Adulto , Biomarcadores , Inhibidores de Anhidrasa Carbónica/administración & dosificación , Estudios Cruzados , Método Doble Ciego , Interacciones Farmacológicas , Femenino , Humanos , Masculino , Narcóticos/administración & dosificación , Oxicodona/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA