Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.966
Filtrar
Más filtros

Intervalo de año de publicación
1.
N Engl J Med ; 388(19): 1779-1789, 2023 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-37163624

RESUMEN

BACKGROUND: Since 2010, Black persons in the United States have had a greater increase in opioid overdose-related mortality than other groups, but national-level evidence characterizing racial and ethnic disparities in the use of medications for opioid use disorder (OUD) is limited. METHODS: We used Medicare claims data from the 2016-2019 period for a random 40% sample of fee-for-service beneficiaries who were Black, Hispanic, or White; were eligible for Medicare owing to disability; and had an index event related to OUD (nonfatal overdose treated in an emergency department or inpatient setting, hospitalization with injection drug use-related infection, or inpatient or residential rehabilitation or detoxification care). We measured the receipt of medications to treat OUD (buprenorphine, naltrexone, and naloxone), the receipt of high-risk medications (opioid analgesics and benzodiazepines), and health care utilization, all in the 180 days after the index event. We estimated differences in outcomes according to race and ethnic group with adjustment for beneficiary age, sex, index event, count of chronic coexisting conditions, and state of residence. RESULTS: We identified 25,904 OUD-related index events among 23,370 beneficiaries, with 3937 events (15.2%) occurring among Black patients, 2105 (8.1%) among Hispanic patients, and 19,862 (76.7%) among White patients. In the 180 days after the index event, patients received buprenorphine after 12.7% of events among Black patients, after 18.7% of those among Hispanic patients, and after 23.3% of those among White patients; patients received naloxone after 14.4%, 20.7%, and 22.9%, respectively; and patients received benzodiazepines after 23.4%, 29.6%, and 37.1%, respectively. Racial differences in the receipt of medications to treat OUD did not change appreciably from 2016 to 2019 (buprenorphine receipt: after 9.1% of index events among Black patients vs. 21.6% of those among White patients in 2016, and after 14.1% vs. 25.5% in 2019). In all study groups, patients had multiple ambulatory visits in the 180 days after the index event (mean number of visits, 6.6 after events among Black patients, 6.7 after events among Hispanic patients, and 7.6 after events among White patients). CONCLUSIONS: Racial and ethnic differences in the receipt of medications to treat OUD after an index event related to this disorder among patients with disability were substantial and did not change over time. The high incidence of ambulatory visits in all groups showed that disparities persisted despite frequent health care contact. (Funded by the National Institute on Drug Abuse and the National Institute on Aging.).


Asunto(s)
Analgésicos Opioides , Benzodiazepinas , Disparidades en Atención de Salud , Antagonistas de Narcóticos , Trastornos Relacionados con Opioides , Anciano , Humanos , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/uso terapéutico , Benzodiazepinas/administración & dosificación , Benzodiazepinas/uso terapéutico , Buprenorfina/uso terapéutico , Medicare/estadística & datos numéricos , Naloxona/uso terapéutico , Trastornos Relacionados con Opioides/complicaciones , Trastornos Relacionados con Opioides/tratamiento farmacológico , Trastornos Relacionados con Opioides/epidemiología , Trastornos Relacionados con Opioides/etnología , Estados Unidos/epidemiología , Disparidades en Atención de Salud/etnología , Disparidades en Atención de Salud/estadística & datos numéricos , Sobredosis de Opiáceos/epidemiología , Sobredosis de Opiáceos/etnología , Sobredosis de Opiáceos/etiología , Sobredosis de Opiáceos/prevención & control , Negro o Afroamericano/estadística & datos numéricos , Blanco/estadística & datos numéricos , Hispánicos o Latinos/estadística & datos numéricos , Naltrexona/uso terapéutico , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/uso terapéutico
2.
N Engl J Med ; 384(2): 140-153, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-33497547

RESUMEN

BACKGROUND: The use of naltrexone plus bupropion to treat methamphetamine use disorder has not been well studied. METHODS: We conducted this multisite, double-blind, two-stage, placebo-controlled trial with the use of a sequential parallel comparison design to evaluate the efficacy and safety of extended-release injectable naltrexone (380 mg every 3 weeks) plus oral extended-release bupropion (450 mg per day) in adults with moderate or severe methamphetamine use disorder. In the first stage of the trial, participants were randomly assigned in a 0.26:0.74 ratio to receive naltrexone-bupropion or matching injectable and oral placebo for 6 weeks. Those in the placebo group who did not have a response in stage 1 underwent rerandomization in stage 2 and were assigned in a 1:1 ratio to receive naltrexone-bupropion or placebo for an additional 6 weeks. Urine samples were obtained from participants twice weekly. The primary outcome was a response, defined as at least three methamphetamine-negative urine samples out of four samples obtained at the end of stage 1 or stage 2, and the weighted average of the responses in the two stages is reported. The treatment effect was defined as the between-group difference in the overall weighted responses. RESULTS: A total of 403 participants were enrolled in stage 1, and 225 in stage 2. In the first stage, 18 of 109 participants (16.5%) in the naltrexone-bupropion group and 10 of 294 (3.4%) in the placebo group had a response. In the second stage, 13 of 114 (11.4%) in the naltrexone-bupropion group and 2 of 111 (1.8%) in the placebo group had a response. The weighted average response across the two stages was 13.6% with naltrexone-bupropion and 2.5% with placebo, for an overall treatment effect of 11.1 percentage points (Wald z-test statistic, 4.53; P<0.001). Adverse events with naltrexone-bupropion included gastrointestinal disorders, tremor, malaise, hyperhidrosis, and anorexia. Serious adverse events occurred in 8 of 223 participants (3.6%) who received naltrexone-bupropion during the trial. CONCLUSIONS: Among adults with methamphetamine use disorder, the response over a period of 12 weeks among participants who received extended-release injectable naltrexone plus oral extended-release bupropion was low but was higher than that among participants who received placebo. (Funded by the National Institute on Drug Abuse and others; ADAPT-2 ClinicalTrials.gov number, NCT03078075.).


Asunto(s)
Trastornos Relacionados con Anfetaminas/tratamiento farmacológico , Bupropión/administración & dosificación , Metanfetamina , Naltrexona/administración & dosificación , Administración Oral , Adolescente , Adulto , Anciano , Bupropión/efectos adversos , Preparaciones de Acción Retardada , Método Doble Ciego , Quimioterapia Combinada , Femenino , Humanos , Inyecciones , Masculino , Cumplimiento de la Medicación , Metanfetamina/orina , Persona de Mediana Edad , Naltrexona/efectos adversos , Antagonistas de Narcóticos , Adulto Joven
3.
J Pharmacol Exp Ther ; 389(1): 5-14, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-37699710

RESUMEN

Opioid addiction is a chronic relapsing disorder in which drug-seeking behavior during abstinence can be provoked by exposure to a µ-opioid receptor (MOR) agonist or opioid-associated cues. Opioid self-administration behavior in laboratory subjects can be reinstated by priming with MOR agonists or agonist-related stimuli, providing a procedure suitable for relapse-related studies. The opioid antagonist naltrexone has been forwarded as a medication that can forestall relapse and, in an extended-release formulation, has demonstrated some treatment success. However, chronic naltrexone treatment has not been extensively investigated in nonhuman subjects and aspects of its pharmacology remain uncertain. For example, the relative effectiveness of naltrexone in reducing the priming strength of opioid agonists differing in efficacy is not well understood. Here, using intravenous self-administration and warm-water tail withdrawal procedures, we investigated changes in the direct reinforcing effects of oxycodone and in the priming strength and antinociceptive effects of opioid agonists in squirrel monkeys (n = 4) during chronic treatment with naltrexone (0.2 mg/kg/d). Results show that naltrexone produced: 1) a 10-fold rightward shift in the dose-response function for the reinforcing effects of oxycodone, and 2) in reinstatement and antinociception experiments, comparable rightward shifts in the dose-response functions for higher-efficacy MOR agonists (methadone, heroin, and oxycodone) but rightward and downward shifts in the dose-response functions for lower-efficacy MOR agonists (buprenorphine, nalbuphine, and butorphanol). These results suggest that, although chronic naltrexone should be effective in forestalling relapse following exposure to lower- and higher-efficacy agonists, the inability of lower-efficacy agonists to surmount naltrexone antagonism may complicate the prescription of opioids for pain. SIGNIFICANCE STATEMENT: Although naltrexone is commonly used in the treatment of opioid use disorder, its ability to reduce the priming strength of opioid agonists has not been extensively investigated. This study shows that chronic naltrexone treatment induces rightward shifts in the reinstatement and antinociceptive properties of higher efficacy opioid agonists, but rightward and downward shifts for lower efficacy opioid agonists, suggesting lower efficacy agonists may not be able to surmount naltrexone-induced antagonism of these two effects, and perhaps naltrexone offers greater protection against lower efficacy agonists.


Asunto(s)
Analgésicos Opioides , Naltrexona , Humanos , Analgésicos Opioides/farmacología , Naltrexona/farmacología , Oxicodona , Comportamiento de Búsqueda de Drogas , Antagonistas de Narcóticos/farmacología , Recurrencia , Receptores Opioides mu/agonistas , Relación Dosis-Respuesta a Droga
4.
Int J Obes (Lond) ; 48(8): 1118-1125, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38724682

RESUMEN

OBJECTIVE: Little is known about the effect of a multi-drug weight loss strategy in obesity treatment, particularly combining bupropion/naltrexone and glucagon-like peptide 1 (GLP-1) analogue. The purpose of this study was to evaluate if there are any additive effects of prescribing bupropion/naltrexone on top of GLP-1 analogue as weight loss therapy. METHODS: This was a retrospective cohort study of adult patients with a body mass index (BMI) ≥ 30 kg/m2 prescribed GLP-1 analogue therapy at an obesity specialist clinic in Vancouver, Canada. We compared a 6 and 12-month change in total body weight loss (TBWL) for those receiving monotherapy from the initiation of GLP-1 analogue therapy with those receiving combination therapy from the initiation of bupropion/naltrexone added-on therapy. Patients prescribed combination therapy were stratified into responder (loss of ≥ 5% TBWL) and non-responder (TBWL < 5%) subgroups based on initial response to the GLP-1 analogue alone for any amount of time. RESULTS: The mean weight loss among patients prescribed GLP-1 analogue monotherapy at 12 months was 11.42 kg, SD 9.95 (9.6% TBWL). There was no significant difference between these two treatment strategies overall (HR 0.88, 95% CI 0.68 to 1.14, p = 0.35). However, when stratified by response to initial GLP analogue therapy, the addition of bupropion/naltrexone was associated with a statistically significant reduction in weight in both the responder (4.3% TBWL (p < 0.01)) and non-responder groups (4.0% TBWL (p < 0.01)). CONCLUSIONS: GLP-1 analogues are an effective treatment for weight loss, and the addition of bupropion/naltrexone is associated with greater weight loss including in patients who are initially non-responsive to GLP-1 analogues.


Asunto(s)
Bupropión , Quimioterapia Combinada , Naltrexona , Pérdida de Peso , Humanos , Bupropión/uso terapéutico , Estudios Retrospectivos , Naltrexona/uso terapéutico , Naltrexona/análogos & derivados , Pérdida de Peso/efectos de los fármacos , Masculino , Femenino , Persona de Mediana Edad , Adulto , Péptido 1 Similar al Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón/uso terapéutico , Obesidad/tratamiento farmacológico , Resultado del Tratamiento , Combinación de Medicamentos , Fármacos Antiobesidad/uso terapéutico
5.
Int J Obes (Lond) ; 48(5): 683-693, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38291203

RESUMEN

OBJECTIVES: This study aimed to assess the cost-effectiveness of weight-management pharmacotherapies approved by Canada Health, i.e., orlistat, naltrexone 32 mg/bupropion 360 mg (NB-32), liraglutide 3.0 mg and semaglutide 2.4 mg as compared to the current standard of care (SoC). METHODS: Analyses were conducted using a cohort with a mean starting age 50 years, body mass index (BMI) 37.5 kg/m2, and 27.6% having type 2 diabetes. Using treatment-specific changes in surrogate endpoints from the STEP trials (BMI, glycemic, blood pressure, lipids), besides a network meta-analysis, the occurrence of weight-related complications, costs, and quality-adjusted life-years (QALYs) were projected over lifetime. RESULTS: From a societal perspective, at a willingness-to-pay (WTP) threshold of CAD 50 000 per QALY, semaglutide 2.4 mg was the most cost-effective treatment, at an incremental cost-utility ratio (ICUR) of CAD 31 243 and CAD 29 014 per QALY gained versus the next best alternative, i.e., orlistat, and SoC, respectively. Semaglutide 2.4 mg extendedly dominated other pharmacotherapies such as NB-32 or liraglutide 3.0 mg and remained cost-effective both under a public and private payer perspective. Results were robust to sensitivity analyses varying post-treatment catch-up rates, longer treatment durations and using real-world cohort characteristics. Semaglutide 2.4 mg was the preferred intervention, with a likelihood of 70% at a WTP threshold of CAD 50 000 per QALY gained. However, when the modeled benefits of weight-loss on cancer, mortality, cardiovascular disease (CVD) or osteoarthritis surgeries were removed simultaneously, orlistat emerged as the best value for money compared with SoC, with an ICUR of CAD 35 723 per QALY gained. CONCLUSION: Semaglutide 2.4 mg was the most cost-effective treatment alternative compared with D&E or orlistat alone, and extendedly dominated other pharmacotherapies such as NB-32 or liraglutide 3.0 mg. Results were sensitive to the inclusion of the combined benefits of mortality, cancer, CVD, and knee osteoarthritis.


Asunto(s)
Fármacos Antiobesidad , Análisis Costo-Beneficio , Obesidad , Orlistat , Humanos , Canadá , Persona de Mediana Edad , Obesidad/tratamiento farmacológico , Obesidad/economía , Femenino , Fármacos Antiobesidad/uso terapéutico , Fármacos Antiobesidad/economía , Masculino , Orlistat/uso terapéutico , Años de Vida Ajustados por Calidad de Vida , Liraglutida/uso terapéutico , Liraglutida/economía , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/economía , Bupropión/uso terapéutico , Bupropión/economía , Naltrexona/uso terapéutico , Naltrexona/economía , Péptidos Similares al Glucagón/uso terapéutico , Péptidos Similares al Glucagón/economía
6.
J Transl Med ; 22(1): 630, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38970055

RESUMEN

Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a debilitating disease with a broad overlap of symptomatology with Post-COVID Syndrome (PCS). Despite the severity of symptoms and various neurological, cardiovascular, microvascular, and skeletal muscular findings, no biomarkers have been identified. The Transient receptor potential melastatin 3 (TRPM3) channel, involved in pain transduction, thermosensation, transmitter and neuropeptide release, mechanoregulation, vasorelaxation, and immune defense, shows altered function in ME/CFS. Dysfunction of TRPM3 in natural killer (NK) cells, characterized by reduced calcium flux, has been observed in ME/CFS and PCS patients, suggesting a role in ineffective pathogen clearance and potential virus persistence and autoimmunity development. TRPM3 dysfunction in NK cells can be improved by naltrexone in vitro and ex vivo, which may explain the moderate clinical efficacy of low-dose naltrexone (LDN) treatment. We propose that TRPM3 dysfunction may have a broader involvement in ME/CFS pathophysiology, affecting other organs. This paper discusses TRPM3's expression in various organs and its potential impact on ME/CFS symptoms, with a focus on small nerve fibers and the brain, where TRPM3 is involved in presynaptic GABA release.


Asunto(s)
Síndrome de Fatiga Crónica , Naltrexona , Canales Catiónicos TRPM , Humanos , Síndrome de Fatiga Crónica/tratamiento farmacológico , Canales Catiónicos TRPM/metabolismo , Naltrexona/uso terapéutico , Naltrexona/farmacología , Naltrexona/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Resultado del Tratamiento
7.
Eur J Clin Invest ; 54(4): e14122, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37929909

RESUMEN

BACKGROUND: Considering the conflicting effects of bupropion on parameters related to metabolic syndrome including glucose metabolism and lipid profile, in this meta-analysis study, we investigated the effects of this drug alone or in combination with naltrexone on glucose metabolism and lipid profile. METHODS: Scopus, PubMed/Medline, Web of Science and Embase databases were searched using standard keywords to identify all controlled trials investigating effects of bupropion alone and combined with naltrexone on the glucose and lipid profile. Pooled weighted mean difference and 95% confidence intervals were achieved by random-effects model. RESULTS: Twelve studies with 5152 participants' were included in this article. The pooled findings showed that bupropion alone or in combination with naltrexone would significantly reduce glucose (weighted mean difference (WMD): -2.25 mg/dL, 95% confidence interval (CI): -4.10, -0.40), insulin (WMD: -4.06 µU/mL, 95% CI: -6.09, -2.03), homeostatic model assessment for insulin resistance (HOMA-IR) (WMD: -0.58, 95% CI: -0.98, -0.19), triglyceride (TG) (WMD: -11.78 mg/dL, 95% CI: -14.48 to -9.08) and increase high-density lipoprotein (HDL) (WMD: 2.68 mg/dL, 95% CI: 2.13 to 3.24). A Greater reduction in glucose levels was observed with duration >26 weeks. Dose of bupropion intake ≤360 mg and intervention for more than 26 weeks decreased insulin level significantly. With regard to lipid profile, reduction of triglycerides is more significant with dose of bupropion greater than 360 mg and a shorter intervention length equal to 26 weeks. CONCLUSIONS: The addition of combination therapies such as bupropion and naltrexone to lifestyle modification can significantly improve glucose metabolism and some lipid parameters.


Asunto(s)
Bupropión , Naltrexona , Humanos , Suplementos Dietéticos , Glucosa , Insulina , Naltrexona/farmacología , Triglicéridos
8.
Eur J Clin Invest ; 54(3): e14118, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37924302

RESUMEN

BACKGROUND: Considering the conflicting effects of bupropion on parameters related to cardiovascular system including blood pressure and inflammation, in this meta-analysis study, we investigated the effects of this drug alone or in combination with naltrexone on systolic (SBP) and diastolic blood pressure (DBP) and C-reactive protein (CRP). METHODS: Scopus, PubMed/Medline, Web of Science and Embase databases were searched using standard keywords to identify all controlled trials investigating effects of bupropion alone and combined with naltrexone on the BP and CRP. Pooled weighted mean difference and 95% confidence intervals (CIs) were achieved by random-effects model analysis for the best estimation of outcomes. RESULTS: The pooled findings showed that that bupropion alone or in combination with naltrexone would significantly increase SBP (weighted mean difference (WMD): 1.34 mmHg, 95% CI: 0.38-2.29) and DBP (WMD: 0.93 mmHg, 95% CI 0.88-0.99) as well as decrease CRP (WMD: -0.89 mg/L, 95% CI -1.09 to -0.70). The findings of the subgroup also show the greater effect of bupropion on blood pressure (SBP and DBP) increase in a dose greater than 360 mg and a duration of intervention less equal to 26 weeks. In addition, the subgroup analysis showed that changes in SBP after receiving bupropion together with naltrexone were more compared to bupropion alone. CONCLUSIONS: The addition of combination therapies such as bupropion and naltrexone can significantly improve CRP levels. However, its effect on blood pressure requires proper management of this drug.


Asunto(s)
Proteína C-Reactiva , Hipertensión , Humanos , Presión Sanguínea , Naltrexona/farmacología , Naltrexona/uso terapéutico , Bupropión/uso terapéutico , Bupropión/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto , Análisis de Regresión , Hipertensión/tratamiento farmacológico
9.
J Gen Intern Med ; 39(8): 1342-1348, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38424347

RESUMEN

BACKGROUND: Treatment-seeking people with opioid use disorder (OUD) who are capable of pregnancy need accurate information about the potential impact of medication to treat OUD (MOUD) on fertility to make informed choices about treatment that are consistent with their reproductive wishes. There is a dearth of research on fertility associated with MOUD receipt in birthing people with OUD. OBJECTIVE: To estimate the association between treatment with MOUD and odds of conception among birthing people using national administrative claims. DESIGN: Retrospective case-crossover study using multi-state US administrative data (2006-2016). Dates of conception were estimated from delivery dates and served as "case" days for which MOUD exposures were compared to those on all other ("control") days of insurance enrollment. PARTICIPANTS: Treatment-seeking people with OUD with a delivery during the observation period. MAIN MEASURES: Odds ratios for conception from within-person fixed effects models were modeled as a function of exposure to MOUD (buprenorphine, methadone, extended-release depot naltrexone, or oral naltrexone) using conditional logistic regression. KEY RESULTS: A total of 21,928 births among 19,133 people with OUD were identified. In the sample, 5873 people received buprenorphine, 1825 methadone, 486 extended-release naltrexone, and 714 oral naltrexone. Participants could receive more than one type of MOUD. Mean age was 28.2 years (SD = 2.2; range = 16-45), with 76.2% having Medicaid. vs. commercial insurance. Compared to no MOUD, periods of methadone (aOR = 0.55 [95% CI = 0.48-0.63]) or buprenorphine receipt (aOR = 0.84 [0.77-0.91]) were associated with fewer conceptions. Treatment periods with extended-release depot naltrexone compared to no medication were associated with higher odds of conception (aOR = 1.75 [1.22-2.50]) and there was no significant difference in conception with oral naltrexone (aOR = 1.02 [0.67-1.54]). CONCLUSIONS: The association between MOUD and odds of conception among birthing people varied by type of MOUD, with extended-release naltrexone associated with higher odds of conceiving compared to no treatment. Clinical studies are urgently needed to investigate these findings further.


Asunto(s)
Buprenorfina , Metadona , Naltrexona , Tratamiento de Sustitución de Opiáceos , Trastornos Relacionados con Opioides , Índice de Embarazo , Humanos , Femenino , Embarazo , Trastornos Relacionados con Opioides/tratamiento farmacológico , Trastornos Relacionados con Opioides/epidemiología , Adulto , Estudios Retrospectivos , Tratamiento de Sustitución de Opiáceos/métodos , Naltrexona/uso terapéutico , Naltrexona/administración & dosificación , Buprenorfina/uso terapéutico , Buprenorfina/administración & dosificación , Metadona/uso terapéutico , Metadona/administración & dosificación , Adulto Joven , Estudios Cruzados , Estados Unidos/epidemiología , Analgésicos Opioides/uso terapéutico , Analgésicos Opioides/administración & dosificación , Complicaciones del Embarazo/tratamiento farmacológico , Antagonistas de Narcóticos/uso terapéutico , Antagonistas de Narcóticos/administración & dosificación , Adolescente
10.
Anesthesiology ; 141(2): 388-399, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38980158

RESUMEN

Substance use disorders, including alcohol use disorder, are a public health concern that affect more than 150 million people globally. The opioid antagonist naltrexone is being increasingly prescribed to treat opioid use disorder, alcohol use disorder, and chronic pain. Perioperative management of patients on naltrexone is inconsistent and remains a controversial topic, with mismanagement posing a significant risk to the long-term health of these patients. This scoping review was conducted to identify human studies in which the perioperative management of naltrexone was described. This review includes a systematic literature search involving Medline, Medline In-Process, Embase, PsycINFO, and Web of Science. Seventeen articles that describe perioperative naltrexone management strategies were included, including thirteen guidelines, one case report, and three randomized trials. Despite its use in patients with alcohol use disorder and chronic pain, no clinical studies, case reports, or guidelines addressed naltrexone use in these clinical populations. All of the guideline documents recommended the preoperative cessation of naltrexone, irrespective of dose, indication, or route of administration. None of these guideline documents were designed on the basis of a systematic literature search or a Delphi protocol. As described by the primary studies, perioperative pain relief varied depending on naltrexone dose and route of administration, time since last naltrexone administration, and underlying substance use disorder. None of the studies commented on the maintenance of recovery for the patient's substance use disorder in the context of perioperative naltrexone management. The current understanding of the risks and benefits of continuing or stopping naltrexone perioperatively is limited by a lack of high-quality evidence. In patients with risk factors for return to use of opioids or alcohol, the discontinuation of naltrexone should have a strong rationale. Future studies and guidelines should seek to address both acute pain management and maintaining recovery when discussing perioperative naltrexone management strategies.


Asunto(s)
Naltrexona , Antagonistas de Narcóticos , Atención Perioperativa , Naltrexona/uso terapéutico , Naltrexona/administración & dosificación , Humanos , Antagonistas de Narcóticos/uso terapéutico , Antagonistas de Narcóticos/administración & dosificación , Atención Perioperativa/métodos , Trastornos Relacionados con Opioides/prevención & control , Manejo del Dolor/métodos , Dolor Postoperatorio/tratamiento farmacológico
11.
Diabetes Obes Metab ; 26(8): 3147-3154, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38757729

RESUMEN

AIM: To investigate growth/differentiation factor 15 (GDF-15) levels in response to antiobesity medications, namely, liraglutide (Lira) and naltrexone/bupropion (N/B), in individuals with overweight or obesity. MATERIALS AND METHODS: This was a prospective, non-randomized clinical trial with a two-arm, parallel design. A total of 42 individuals with overweight or obesity without type 1 or type 2 diabetes mellitus were enrolled. The participants received either Lira 3 mg or N/B 32/360 mg, along with diet and exercise, according to comorbidities, cost and method of administration. Participants underwent clinical and laboratory measurements at baseline, as well as at the 3- and 6-month time points. Anthropometric measurements and body composition analysis via bioelectrical impendence analysis were performed. Total blood samples for GDF-15 and H-specific GDF-15 were collected in the fasting state and every 30 min for 3 h after the consumption of a standardized mixed meal. RESULTS: Overall, participants' weight was reduced by 9.29 ± 5.34 kg at Month 3 and 11.52 ± 7.52 kg at Month 6. Total and H-specific GDF-15 levels did not show significant changes during the mixed meal compared to values before the meal when all participants were examined at baseline, and at 3 and 6 month follow-ups. No statistical significance was found when participants were examined by subgroup (Lira vs. N/B). No significant differences between treatment groups in postprandial area under the curve (AUC) or incremental AUC values were found at baseline or in the follow-up months with regard to total and H-specific GDF-15 levels. CONCLUSION: Neither total nor H-specific GDF-15 levels are affected by Lira or N/B treatment in patients with overweight or obesity.


Asunto(s)
Fármacos Antiobesidad , Bupropión , Factor 15 de Diferenciación de Crecimiento , Liraglutida , Naltrexona , Obesidad , Humanos , Liraglutida/uso terapéutico , Femenino , Masculino , Factor 15 de Diferenciación de Crecimiento/sangre , Bupropión/uso terapéutico , Bupropión/administración & dosificación , Naltrexona/uso terapéutico , Naltrexona/administración & dosificación , Persona de Mediana Edad , Estudios Prospectivos , Obesidad/tratamiento farmacológico , Obesidad/sangre , Adulto , Fármacos Antiobesidad/uso terapéutico , Fármacos Antiobesidad/administración & dosificación , Sobrepeso , Pérdida de Peso/efectos de los fármacos , Combinación de Medicamentos , Periodo Posprandial
12.
Diabetes Obes Metab ; 26(5): 1687-1696, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38287140

RESUMEN

AIM: To characterize factors associated with the receipt of anti-obesity medication (AOM) prescription and fill. MATERIALS AND METHODS: This retrospective cohort study used electronic health records from 1 January 2015 to 30 June 2023, in a large health system in Ohio and Florida. Adults with a body mass index ≥30 kg/m2 who attended ≥1 weight-management programme or had an initial AOM prescription between 1 July 2015 and 31 December 2022, were included. The main measures were a prescription for an AOM (naltrexone-bupropion, orlistat, phentermine-topiramate, liraglutide 3.0 mg and semaglutide 2.4 mg) and an AOM fill during the study follow-up. RESULTS: We identified 50 678 adults, with a mean body mass index of 38 ± 8 kg/m2 and follow-up of 4.7 ± 2.4 years. Only 8.0% of the cohort had AOM prescriptions and 4.4% had filled prescriptions. In the multivariable analyses, being a man, Black, Hispanic and other race/ethnicity (vs. White), Medicaid, traditional Medicare, Medicare Advantage, self-pay and other insurance types (vs. private insurance) and fourth quartile of the area deprivation index (vs. first quartile) were associated with lower odds of a new prescription. Hispanic ethnicity, being a man, Medicaid, traditional Medicare and Medicare Advantage insurance types, liraglutide and orlistat (vs. naltrexone-buproprion) were associated with lower odds of AOM fill, while phentermine-topiramate was associated with higher odds. Among privately insured individuals, the insurance carrier was associated with both the odds of AOM prescription and fill. CONCLUSIONS: Significant disparities exist in access to AOM both at the prescribing stage and getting the prescription filled based on patient characteristics and insurance type.


Asunto(s)
Fármacos Antiobesidad , Medicare Part C , Anciano , Adulto , Humanos , Estados Unidos/epidemiología , Orlistat/uso terapéutico , Estudios Retrospectivos , Topiramato , Naltrexona/uso terapéutico , Liraglutida/uso terapéutico , Fármacos Antiobesidad/uso terapéutico , Fentermina
13.
Diabetes Obes Metab ; 26(6): 2102-2110, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38419410

RESUMEN

AIMS: To describe trends in the use of anti-obesity drugs in Norway during the period 2004-2022. MATERIALS AND METHODS: We assessed the annual utilization of any available drug indicated for obesity recorded in the nationwide Norwegian Prescribed Drug Register for adults (age 18-79 years) from 1 January 2004 to 31 December 2022. Prevalence was stratified by sex and age group (18-29 years and 10-year age groups thereafter). Additional analyses were performed in individuals initiating treatment with an anti-obesity drug and on the cost of the anti-obesity drugs since 2017. RESULTS: The prevalence of anti-obesity drug use decreased from 2009, when sibutramine and rimonabant were withdrawn from the market, and increased again after the approval of bupropion-naltrexone in 2017 and liraglutide in 2018. The use of the peripheral-acting anti-obesity drug orlistat decreased from 2004. In 2022, 1.04% of the adult Norwegian population (72.8% women) filled at least one prescription of bupropion-naltrexone, 0.91% used liraglutide (Saxenda; 74.2% women), and semaglutide without reimbursement was used by 0.68% (76.7% women). The prevalence increased with age, peaking in the age group 50 to 59 years, and decreased in older age groups. From 2017 to 2022, 2.8% of the adult residents initiated treatment with an anti-obesity drug. The total sale of those drugs increased from 1.1 million euros in 2017 to 91.8 million euros in 2022. CONCLUSIONS: The use of anti-obesity drugs in Norway has increased substantially in recent years, especially among women aged 40 to 59 years. Changes in availability and reimbursement have influenced the use of these drugs in recent years.


Asunto(s)
Fármacos Antiobesidad , Bupropión , Liraglutida , Naltrexona , Obesidad , Humanos , Adulto , Noruega/epidemiología , Persona de Mediana Edad , Femenino , Masculino , Fármacos Antiobesidad/uso terapéutico , Fármacos Antiobesidad/economía , Obesidad/tratamiento farmacológico , Obesidad/epidemiología , Adolescente , Anciano , Adulto Joven , Liraglutida/uso terapéutico , Bupropión/uso terapéutico , Naltrexona/uso terapéutico , Orlistat/uso terapéutico , Rimonabant/uso terapéutico , Péptidos Similares al Glucagón/uso terapéutico , Péptidos Similares al Glucagón/análogos & derivados , Costos de los Medicamentos/estadística & datos numéricos , Sistema de Registros , Prevalencia , Utilización de Medicamentos/tendencias , Utilización de Medicamentos/estadística & datos numéricos , Ciclobutanos
14.
Diabetes Obes Metab ; 26(6): 2466-2475, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38533552

RESUMEN

BACKGROUND: Metabolic syndrome (MetS), which can be induced or exacerbated by the current class of antipsychotic drugs, is highly prevalent in patients with schizophrenia and presents significant challenges to lifetime disease management. Supported by initial clinical results, trace amine-associated receptor 1 (TAAR1) agonists have emerged as potential novel treatments for schizophrenia. Notably, non-clinical studies have also shown weight-lowering and glucoregulatory effects of TAAR1 agonists, including the investigational agent ulotaront. However, the translatability of these findings to humans has not been adequately assessed. Given that delayed gastric emptying (GE) was identified as a potential mechanism contributing to the metabolic benefits of TAAR1 agonists in rodents, the aim of this study was to evaluate the effect of ulotaront on GE in patients with schizophrenia and concurrent MetS with prediabetes. METHODS: Patients with schizophrenia were randomized to receive a single oral dose of ulotaront (150 mg) and their previous antipsychotic (PA) in an open-label, crossover, two-sequence design (NCT05402111). Eligible participants fulfilled at least three of five MetS criteria and had prediabetes defined by elevated glycated haemoglobin (5.7-6.4%) and/or fasting homeostatic model assessment of insulin resistance (i.e. ≥2.22). Following an overnight fast and 4 h post-dose, participants ingested a 99mTc-sulphur colloid radiolabelled egg meal (320 kcal, 30% fat). GE was measured by scintigraphy over 4 h. Endpoints included GE of solids half-time (T1/2) and percentage gastric retention at 1, 2 and 4 h. RESULTS: Thirty-one adults were randomized and 27 completed the study. Ulotaront significantly delayed GE of solids [median GE T1/2 ulotaront at 139 min (119, 182) vs. the participant's PA of 124 min (109, 132), p = .006]. A significant increase in gastric retention was seen in the ulotaront versus the PA group at 1 h (80% vs. 75%, p = .015), 2 h (61% vs. 50%, p = .023) and 4 h (17% vs. 7%, p = .002) post-meal. CONCLUSION: Ulotaront delayed the GE of solids in patients with schizophrenia and concurrent MetS with prediabetes. Additional studies are needed to assess whether treatment with TAAR1 agonists is associated with weight loss and glucoregulatory improvement.


Asunto(s)
Antipsicóticos , Estudios Cruzados , Vaciamiento Gástrico , Síndrome Metabólico , Naltrexona/análogos & derivados , Estado Prediabético , Receptores Acoplados a Proteínas G , Esquizofrenia , Humanos , Vaciamiento Gástrico/efectos de los fármacos , Masculino , Femenino , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/complicaciones , Adulto , Persona de Mediana Edad , Síndrome Metabólico/complicaciones , Síndrome Metabólico/tratamiento farmacológico , Estado Prediabético/complicaciones , Estado Prediabético/tratamiento farmacológico , Antipsicóticos/uso terapéutico , Antipsicóticos/efectos adversos , Receptores Acoplados a Proteínas G/agonistas , Tetrahidronaftalenos/uso terapéutico , Tetrahidronaftalenos/farmacología
15.
Semin Neurol ; 44(4): 441-451, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38848746

RESUMEN

The rates of opioid use and opioid related deaths are escalating in the United States. Despite this, evidence-based treatments for Opioid Use Disorder are underutilized. There are three medications FDA approved for treatment of Opioid Use Disorder: Methadone, Buprenorphine, and Naltrexone. This article reviews the history, criteria, and mechanisms associated with Opioid Use Disorder. Pertinent pharmacology considerations, treatment strategies, efficacy, safety, and challenges of Methadone, Buprenorphine, and Naltrexone are outlined. Lastly, a practical decision making algorithm is discussed to address pertinent psychiatric and medical comorbidities when prescribing pharmacology for Opioid Use Disorder.


Asunto(s)
Buprenorfina , Metadona , Naltrexona , Antagonistas de Narcóticos , Tratamiento de Sustitución de Opiáceos , Trastornos Relacionados con Opioides , Humanos , Naltrexona/uso terapéutico , Naltrexona/farmacología , Buprenorfina/uso terapéutico , Buprenorfina/farmacología , Tratamiento de Sustitución de Opiáceos/métodos , Trastornos Relacionados con Opioides/tratamiento farmacológico , Metadona/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Antagonistas de Narcóticos/farmacología , Analgésicos Opioides/uso terapéutico , Analgésicos Opioides/farmacología
16.
Support Care Cancer ; 32(8): 504, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38985364

RESUMEN

PURPOSE: In this study, we aimed to evaluate the safety and effectiveness of naldemedine for treating opioid-induced constipation (OIC) in patients with advanced cancer, who are receiving palliative care, and particularly explored its early effects. METHODS: Palliative care teams and inpatient palliative care units across 14 institutions in Japan were included in this multicenter, prospective, observational study. Patients who were newly prescribed a daily oral dose of 0.2 mg naldemedine were enrolled. The spontaneous bowel movement (SBM) within 24 h after the first dose of naldemedine was considered the primary outcome, whereas, the secondary outcomes included weekly changes in SBM frequency and adverse events. RESULTS: A total of 204 patients were enrolled and 184 completed the 7-day study. The average age of the participants (103 males, 101 females) was 63 ± 14 years. The primary cancer was detected in the lungs (23.5%), gastrointestinal tract (13.7%), and urological organs (9.3%). A considerable proportion of patients (34.8%) had ECOG performance status of 3-4. Most patients were undergoing active cancer treatment, however, 40.7% of the patients were receiving the best supportive care. Within 24 h of the first naldemedine dose, 146 patients (71.6%, 95% CI: 65.4-77.8%) experienced SBMs. The weekly SBM counts increased in 62.7% of the participants. The major adverse events included diarrhea and abdominal pain, detected in 17.6% and 5.4% of the patients, respectively. However, no serious adverse events were observed. CONCLUSION: Conclusively, naldemedine is effective and safe for OIC treatments in real-world palliative care settings. TRIAL REGISTRATION NUMBER: UMIN000031381, registered 20/02/2018.


Asunto(s)
Analgésicos Opioides , Naltrexona , Antagonistas de Narcóticos , Neoplasias , Estreñimiento Inducido por Opioides , Cuidados Paliativos , Humanos , Masculino , Femenino , Persona de Mediana Edad , Estudios Prospectivos , Cuidados Paliativos/métodos , Anciano , Neoplasias/tratamiento farmacológico , Neoplasias/complicaciones , Estreñimiento Inducido por Opioides/tratamiento farmacológico , Naltrexona/análogos & derivados , Naltrexona/uso terapéutico , Naltrexona/administración & dosificación , Naltrexona/efectos adversos , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/uso terapéutico , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/uso terapéutico , Antagonistas de Narcóticos/efectos adversos , Japón , Adulto , Estreñimiento/inducido químicamente , Estreñimiento/tratamiento farmacológico , Anciano de 80 o más Años , Dolor en Cáncer/tratamiento farmacológico , Resultado del Tratamiento
17.
Pharmacopsychiatry ; 57(1): 21-29, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38052239

RESUMEN

INTRODUCTION: Pharmacotherapy with drugs like naltrexone or acamprosate is a well-evaluated element in the treatment of alcohol dependence (AD). However, in many countries, these medications are rarely administered. The objective of the present study was to identify from patients' perspective factors that prevent the initiation and compliance with pharmacological treatment of AD. METHODS: Patients from inpatient alcohol withdrawal treatment underwent a standardized interview. Questions included socio-demographic data, history of AD, treatment history, knowledge and personal experience regarding pharmacotherapy of AD, and personal views about the causes of AD. RESULTS: Three hundred patients (mean age 47.3 years, 27.7% female, mean duration of AD 8.9 years, 67% with a history of previous inpatient withdrawal treatment) were included. The majority of patients (58.7%) already knew drugs for the pharmacotherapy of AD. Thirty percent had ever used such medications, most often acamprosate. Except for disulfiram, pharmacotherapy of AD had lasted only a few weeks, on average. Medication usually had been applied without additional psychotherapy. No severe side effects were reported. Patients had often stopped pharmacotherapy on their own, when assuming they had reached stable abstinence. Openness to start pharmacotherapy for AD was currently stated by 67% of the total sample. In multiple logistic regression, openness was predicted by having a concept of AD as a medical disease and by a shorter duration of AD. DISCUSSION: To improve the administration of pharmacotherapy for AD implementation strategies should be systematically developed and evaluated with a focus on the concept of AD as a medical disease.


Asunto(s)
Disuasivos de Alcohol , Alcoholismo , Síndrome de Abstinencia a Sustancias , Humanos , Femenino , Persona de Mediana Edad , Masculino , Alcoholismo/tratamiento farmacológico , Acamprosato/uso terapéutico , Disuasivos de Alcohol/uso terapéutico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Naltrexona/uso terapéutico , Disulfiram/uso terapéutico , Taurina/uso terapéutico
18.
Cochrane Database Syst Rev ; 1: CD007024, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38180268

RESUMEN

BACKGROUND: Cocaine is a psychostimulant used by approximately 0.4% of the general population worldwide. Cocaine dependence is a chronic mental disorder characterised by the inability to control cocaine use and a host of severe medical and psychosocial complications. There is current no approved pharmacological treatment for cocaine dependence. Some researchers have proposed disulfiram, a medication approved to treat alcohol use disorder. This is an update of a Cochrane review first published in 2010. OBJECTIVES: To evaluate the efficacy and safety of disulfiram for the treatment of cocaine dependence. SEARCH METHODS: We updated our searches of the following databases to August 2022: the Cochrane Drugs and Alcohol Group Specialised Register, CENTRAL, MEDLINE, Embase, CINAHL, and PsycINFO. We also searched for ongoing and unpublished studies via two trials registries. We handsearched the references of topic-related systematic reviews and included studies. The searches had no language restrictions. SELECTION CRITERIA: We included randomised controlled trials that evaluated disulfiram alone or associated with psychosocial interventions versus placebo, no intervention, other pharmacological interventions, or any psychosocial intervention for the treatment of cocaine dependence. DATA COLLECTION AND ANALYSIS: We used standard methodological procedures expected by Cochrane. MAIN RESULTS: Thirteen studies (1191 participants) met our inclusion criteria. Disulfiram versus placebo or no treatment Disulfiram compared to placebo may increase the number of people who are abstinent at the end of treatment (point abstinence; risk ratio (RR) 1.58, 95% confidence interval (CI) 1.05 to 2.36; 3 datasets, 142 participants; low-certainty evidence). However, compared to placebo or no pharmacological treatment, disulfiram may have little or no effect on frequency of cocaine use (standardised mean difference (SMD) -0.11 standard deviations (SDs), 95% CI -0.39 to 0.17; 13 datasets, 818 participants), amount of cocaine use (SMD -0.00 SDs, 95% CI -0.30 to 0.30; 7 datasets, 376 participants), continuous abstinence (RR 1.23, 95% CI 0.80 to 1.91; 6 datasets, 386 participants), and dropout for any reason (RR 1.20, 95% CI 0.92 to 1.55; 14 datasets, 841 participants). The certainty of the evidence was low for all these outcomes. We are unsure about the effects of disulfiram versus placebo on dropout due to adverse events (RR 12.97, 95% CI 0.77 to 218.37; 1 study, 67 participants) and on the occurrence of adverse events (RR 3.00, 95% CI 0.35 to 25.98), because the certainty of the evidence was very low for these outcomes. Disulfiram versus naltrexone Disulfiram compared with naltrexone may reduce the frequency of cocaine use (mean difference (MD) -1.90 days, 95% CI -3.37 to -0.43; 2 datasets, 123 participants; low-certainty evidence) and may have little or no effect on amount of cocaine use (SMD 0.12 SDs, 95% CI -0.27 to 0.51, 2 datasets, 123 participants; low-certainty evidence). We are unsure about the effect of disulfiram versus naltrexone on dropout for any reason (RR 0.86, 95% CI 0.56 to 1.32, 3 datasets, 131 participants) and dropout due to adverse events (RR 0.50, 95% CI 0.07 to 3.55; 1 dataset, 8 participants), because the certainty of the evidence was very low for these outcomes. AUTHORS' CONCLUSIONS: Our results show that disulfiram compared to placebo may increase point abstinence. However, disulfiram compared to placebo or no pharmacological treatment may have little or no effect on frequency of cocaine use, amount of cocaine use, continued abstinence, and dropout for any reason. We are unsure if disulfiram has any adverse effects in this population. Caution is required when transferring our results to clinical practice.


Asunto(s)
Alcoholismo , Trastornos Relacionados con Cocaína , Cocaína , Humanos , Disulfiram/efectos adversos , Trastornos Relacionados con Cocaína/tratamiento farmacológico , Naltrexona , Alcoholismo/tratamiento farmacológico
19.
Alcohol Alcohol ; 59(5)2024 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-39242103

RESUMEN

AIMS: This study aimed to investigate acamprosate and naltrexone dispensing patterns in Australia. METHODS: A 10% representative sample of medications subsidized by the Australian Pharmaceutical Benefits Scheme (PBS) was used to identify individuals who were dispensed naltrexone or acamprosate between January 2006 and December 2023. Data were used to examine concurrent dispensing, medication switching and treatment episode length, as well as changes in prevalence and incidence over time. RESULTS: During the study, we identified 22 745 individuals with a total of 117 548 dispensed prescriptions (45.3% naltrexone, 43.0% acamprosate, and 11.7% concurrent dispensing). Alcohol pharmacotherapy dispensing occurred in 1354 per 100 000 individuals. It is estimated that 2.9% of individuals with an alcohol use disorder in Australia are receiving a PBS-listed pharmacological treatment. For both pharmacotherapies, individuals were most likely to be male (60.0%) and 35-54 years of age (56.0%). Individuals were more likely to switch from acamprosate to naltrexone rather than the reverse. From 2006 and 2023, the number of prevalent individuals treated with an alcohol pharmacotherapy significantly increased, driven mainly the use of naltrexone, which more than doubled over the study period. Incident naltrexone-treated individuals were more likely to remain on treatment for the recommended minimum 3-month period compared to acamprosate treated individuals, although overall dispensing for at least 3 months was low (5.1%). CONCLUSIONS: In Australia between 2006 and 2023, rates of naltrexone dispensing have substantially increased, while acamprosate dispensing showed minimal changes. However, the use of alcohol pharmacotherapies remains low compared with the likely prevalence of alcohol use disorders.


Asunto(s)
Acamprosato , Disuasivos de Alcohol , Alcoholismo , Naltrexona , Humanos , Acamprosato/uso terapéutico , Australia/epidemiología , Masculino , Femenino , Naltrexona/uso terapéutico , Persona de Mediana Edad , Adulto , Disuasivos de Alcohol/uso terapéutico , Alcoholismo/tratamiento farmacológico , Alcoholismo/epidemiología , Adulto Joven , Anciano , Adolescente
20.
Alcohol Alcohol ; 59(5)2024 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-39270736

RESUMEN

AIMS: We conducted a proof-of-concept randomized controlled trial of the mu-opioid receptor antagonist, naltrexone, augmented with the alpha-1 adrenergic receptor antagonist, prazosin, for alcohol use disorder in veterans. We sought a signal that the naltrexone plus prazosin combination regimen would be superior to naltrexone alone. METHODS: Thirty-one actively drinking veterans with alcohol use disorder were randomized 1:1:1:1 to naltrexone plus prazosin (NAL-PRAZ [n = 8]), naltrexone plus placebo (NAL-PLAC [n = 7]), prazosin plus placebo (PRAZ-PLAC [n = 7]), or placebo plus placebo (PLAC-PLAC [n = 9]) for 6 weeks. Prazosin was titrated over 2 weeks to a target dose of 4 mg QAM, 4 mg QPM, and 8 mg QHS. Naltrexone was administered at 50 mg QD. Primary outcomes were the Penn Alcohol Craving Scale (PACS), % drinking days (PDD), and % heavy drinking days (PHDD). RESULTS: In the NAL-PRAZ condition, % reductions from baseline for all three primary outcome measures exceeded 50% and were at least twice as large as % reductions in the NAL-PLAC condition (PACS: 57% vs. 26%; PDD: 51% vs. 22%; PHDD: 69% vs. 15%) and in the other two comparator conditions. Standardized effect sizes between NAL-PRAZ and NAL-PLAC for each primary outcome measure were >0.8. All but one participant assigned to the two prazosin containing conditions achieved the target prazosin dose of 16 mg/day and maintained that dose for the duration of the trial. CONCLUSION: These results suggest that prazosin augmentation of naltrexone enhances naltrexone benefit for alcohol use disorder. These results strengthen rationale for an adequately powered definitive randomized controlled trial.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 1 , Alcoholismo , Quimioterapia Combinada , Naltrexona , Antagonistas de Narcóticos , Prazosina , Prueba de Estudio Conceptual , Humanos , Naltrexona/uso terapéutico , Naltrexona/administración & dosificación , Prazosina/uso terapéutico , Prazosina/administración & dosificación , Masculino , Persona de Mediana Edad , Alcoholismo/tratamiento farmacológico , Antagonistas de Narcóticos/uso terapéutico , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Receptores Adrenérgicos alfa 1/uso terapéutico , Antagonistas de Receptores Adrenérgicos alfa 1/administración & dosificación , Femenino , Adulto , Resultado del Tratamiento , Veteranos , Método Doble Ciego
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA