Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.675
Filtrar
2.
Clin Toxicol (Phila) ; 62(10): 643-650, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39235169

RESUMEN

INTRODUCTION: Naloxone is an effective antidote, but its short half-life means repeated doses, and infusions are often required. We investigated the effectiveness of adding intramuscular naloxone to titrated intravenous naloxone in opioid overdose in preventing recurrence of respiratory depression. METHODS: This double-blinded randomised placebo-controlled trial was conducted in patients with suspected opioid poisoning and respiratory depression (respiratory rate <10 breaths/min or oxygen saturation <93%). Patients were randomised to receive either intramuscular naloxone 1,600 µg or saline placebo. All patients received titrated intravenous naloxone 100 µg and were managed on an opioid poisoning care pathway. The primary outcome was recurrence of respiratory depression within 4 h. Secondary outcomes were the proportion receiving naloxone infusions, number of naloxone boluses administered, reversal of respiratory depression at 10 min, and precipitation of opioid withdrawal (any symptom). RESULTS: Recurrence of respiratory depression within 4 h was less common in 28/69 (41%) patients receiving intramuscular naloxone versus 48/67 (72%) patients receiving placebo (difference 31%, 95% CI: 13-46%; P < 0.001). Fewer naloxone infusions (5/69; 7% versus 25/67; 37%, difference 30%, 95% CI: 15 to 55%; P < 0.001) and fewer naloxone doses were administered (median 2, IQR: 1 to 5, versus median 5, IQR: 2 to 8; P = 0.001) in the intramuscular group. Reversal of respiratory depression at 10 min was similar between groups (51/69; 74% intramuscular naloxone versus 47/67; 70% placebo; P = 0.703). Opioid withdrawal occurred in 35/69 (51%) given intramuscular naloxone compared to 28/67 (42%) in the placebo group (difference 9%; 95% CI: -8 to 27%; P = 0.308). DISCUSSION: The favourable pharmacokinetics of intramuscular naloxone, particularly its longer duration of activity, likely explains the improved effectiveness with lower recurrence of respiratory depression. CONCLUSION: The addition of intramuscular naloxone 1,600 µg to titrated intravenous naloxone prolonged effective reversal of respiratory depression, with fewer naloxone doses and infusions given, and no significant difference in patients developing withdrawal.


Asunto(s)
Naloxona , Antagonistas de Narcóticos , Insuficiencia Respiratoria , Humanos , Naloxona/administración & dosificación , Naloxona/uso terapéutico , Inyecciones Intramusculares , Método Doble Ciego , Masculino , Femenino , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/uso terapéutico , Adulto , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/tratamiento farmacológico , Persona de Mediana Edad , Sobredosis de Opiáceos/tratamiento farmacológico , Infusiones Intravenosas , Resultado del Tratamiento , Analgésicos Opioides/envenenamiento , Analgésicos Opioides/administración & dosificación
3.
CPT Pharmacometrics Syst Pharmacol ; 13(10): 1722-1733, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39283753

RESUMEN

The purpose of the analysis was to evaluate if 10 mg naloxone, administered intramuscularly, could reverse or prevent opioid-induced respiratory depression (OIRD), including OIRD associated with the administration of lethal doses of high-potency opioids. A naloxone population pharmacokinetic (PK) model was generated using data from two naloxone auto-injector (NAI) clinical PK studies. Mechanistic OIRD PK-pharmacodynamic (PD) models were constructed using published data for buprenorphine, morphine, and fentanyl. Due to the lack of published carfentanil data in humans, interspecies allometric scaling methods were used to predict carfentanil PK parameters in humans. A PD model of a combined effect-compartment and receptor kinetics model with a linear relationship between ventilation and carbon dioxide was used to predict the respiratory depression induced by carfentanil. Model-based simulations were performed using the naloxone population PK model and the constructed mechanistic OIRD PK-PD models. Changes in ventilation were assessed after opioid exposure and treatment with 2 mg naloxone or one or two doses of 10 mg naloxone. A higher percentage of subjects recovered back to the rescue ventilation thresholds and/or had a faster recovery to 40% or 70% of baseline ventilation with 10 mg compared with 2 mg naloxone. A second dose of 10 mg naloxone, administered 60 min post-opioid exposure, expedited recovery to 85% of baseline ventilation and delayed time to renarcotization compared with a single dose. In addition, when 10 mg naloxone was administered at 5, 15, 30, or 60 min before fentanyl or carfentanil exposure, rapid and profound OIRD was prevented.


Asunto(s)
Analgésicos Opioides , Fentanilo , Modelos Biológicos , Naloxona , Antagonistas de Narcóticos , Insuficiencia Respiratoria , Naloxona/farmacocinética , Naloxona/administración & dosificación , Naloxona/farmacología , Humanos , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/tratamiento farmacológico , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/farmacología , Antagonistas de Narcóticos/farmacocinética , Antagonistas de Narcóticos/administración & dosificación , Antagonistas de Narcóticos/farmacología , Fentanilo/farmacocinética , Fentanilo/administración & dosificación , Fentanilo/farmacología , Fentanilo/análogos & derivados , Masculino , Adulto , Simulación por Computador , Inyecciones Intramusculares , Femenino , Persona de Mediana Edad , Adulto Joven
5.
Crit Care Explor ; 6(7): e1123, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-39018285

RESUMEN

IMPORTANCE: The opioid crisis is impacting people across the country and deserves attention to be able to curb the rise in opioid-related deaths. OBJECTIVES: To evaluate practice patterns in opioid infusion administration and dosing for patients with acute respiratory failure receiving invasive mechanical ventilation. DESIGN: Retrospective cohort study. SETTING AND PARTICIPANTS: Patients from 21 hospitals in Kaiser Permanente Northern California and 96 hospitals in Philips electronic ICU Research Institute. MAIN OUTCOMES AND MEASURES: We assessed whether patients received opioid infusion and the dose of said opioid infusion. RESULTS: We identified patients with a diagnosis of acute respiratory failure who were initiated on invasive mechanical ventilation. From each patient, we determined if opioid infusions were administered and, among those who received an opioid infusion, the median daily dose of fentanyl infusion. We used hierarchical regression models to quantify variation in opioid infusion use and the median daily dose of fentanyl equivalents across hospitals. We included 13,140 patients in the KPNC cohort and 52,033 patients in the eRI cohort. A total of 7,023 (53.4%) and 16,311 (31.1%) patients received an opioid infusion in the first 21 days of mechanical ventilation in the KPNC and eRI cohorts, respectively. After accounting for patient- and hospital-level fixed effects, the hospital that a patient was admitted to explained 7% (95% CI, 3-11%) and 39% (95% CI, 28-49%) of the variation in opioid infusion use in the KPNC and eRI cohorts, respectively. Among patients who received an opioid infusion, the median daily fentanyl equivalent dose was 692 µg (interquartile range [IQR], 129-1341 µg) in the KPNC cohort and 200 µg (IQR, 0-1050 µg) in the eRI cohort. Hospital explained 4% (95% CI, 1-7%) and 20% (95% CI, 15-26%) of the variation in median daily fentanyl equivalent dose in the KPNC and eRI cohorts, respectively. CONCLUSIONS AND RELEVANCE: In the context of efforts to limit healthcare-associated opioid exposure, our findings highlight the considerable opioid exposure that accompanies mechanical ventilation and suggest potential under and over-treatment with analgesia. Our results facilitate benchmarking of hospitals' analgesia practices against risk-adjusted averages and can be used to inform usual care control arms of analgesia and sedation clinical trials.


Asunto(s)
Analgésicos Opioides , Fentanilo , Pautas de la Práctica en Medicina , Respiración Artificial , Insuficiencia Respiratoria , Humanos , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/uso terapéutico , Analgésicos Opioides/efectos adversos , Masculino , Estudios Retrospectivos , Femenino , Persona de Mediana Edad , Pautas de la Práctica en Medicina/estadística & datos numéricos , Anciano , Fentanilo/administración & dosificación , Fentanilo/uso terapéutico , Insuficiencia Respiratoria/terapia , Insuficiencia Respiratoria/tratamiento farmacológico , Insuficiencia Respiratoria/epidemiología , Estudios de Cohortes , California , Adulto , Unidades de Cuidados Intensivos
6.
Respir Res ; 25(1): 280, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39014448

RESUMEN

BACKGROUND: Morphine relieves dyspnea in various clinical circumstances. Whether or not this applies to patients admitted to intensive care units (ICUs) for acute respiratory failure (ARF) is unknown. We evaluated the efficacy and safety of low-dose morphine on dyspnea in patients admitted to the ICU for ARF. METHODS: In this single-center, double-blind, phase 2, randomized, controlled trial, we assigned non-intubated adults admitted to the ICU for ARF with severe dyspnea, defined by a visual analog scale for dyspnea (dyspnea-VAS) from zero (no dyspnea) to 100 mm (worst imaginable dyspnea) ≥40 mm, to receive a low dose of Morphine Hydrochloride (intravenous titration followed by subcutaneous relay) or Placebo. All patients received standard therapy, including etiological treatment and non-invasive respiratory support. RESULTS: Twenty-two patients were randomized, 11 in each group. The average dyspnea (median [interquartile range]) over 24 hours did not significantly differ between the two groups (40 [25 - 43] mm in the Morphine group vs. 40 [36 - 49] mm in the Placebo group, p=0.411). Dyspnea-VAS was lower in the Morphine group than in the Placebo group at the end of intravenous titration (30 [11 - 30] vs. 35 [30 - 44], p=0.044) and four hours later (18 [10 - 29] vs. 50 [30 - 60], p=0.043). The cumulative probability of intubation was higher in the Morphine group than in the Placebo group (p=0.046) CONCLUSION: In this phase 2 pilot trial, morphine did not improve 24-hour average dyspnea in adult patients with ARF, even though it had a statistically significant immediate effect. Of concern, Morphine use was associated with a higher intubation rate. TRIAL REGISTRATION: The protocol was declared on the ClinicalTrial.gov database (no. NCT04358133) and was published in September 2022.


Asunto(s)
Analgésicos Opioides , Disnea , Morfina , Humanos , Morfina/administración & dosificación , Método Doble Ciego , Disnea/tratamiento farmacológico , Disnea/diagnóstico , Masculino , Femenino , Persona de Mediana Edad , Anciano , Analgésicos Opioides/administración & dosificación , Analgésicos Opioides/efectos adversos , Insuficiencia Respiratoria/tratamiento farmacológico , Insuficiencia Respiratoria/diagnóstico , Resultado del Tratamiento , Adulto
7.
Medicine (Baltimore) ; 103(29): e38837, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39029082

RESUMEN

Opioids exert analgesic effects by agonizing opioid receptors and activating signaling pathways coupled to receptors such as G-protein and/or ß-arrestin. Concomitant respiratory depression (RD) is a common clinical problem, and improvement of RD is usually achieved with specific antagonists such as naloxone; however, naloxone antagonizes opioid analgesia and may produce more unknown adverse effects. In recent years, researchers have used various methods to isolate opioid receptor-mediated analgesia and RD, with the aim of preserving opioid analgesia while attenuating RD. At present, the focus is mainly on the development of new opioids with weak respiratory inhibition or the use of non-opioid drugs to stimulate breathing. This review reports recent advances in novel opioid agents, such as mixed opioid receptor agonists, peripheral selective opioid receptor agonists, opioid receptor splice variant agonists, biased opioid receptor agonists, and allosteric modulators of opioid receptors, as well as in non-opioid agents, such as AMPA receptor modulators, 5-hydroxytryptamine receptor agonists, phosphodiesterase-4 inhibitors, and nicotinic acetylcholine receptor agonists.


Asunto(s)
Analgésicos Opioides , Insuficiencia Respiratoria , Humanos , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/tratamiento farmacológico , Analgésicos Opioides/efectos adversos , Analgésicos Opioides/uso terapéutico , Receptores Opioides/efectos de los fármacos , Receptores Opioides/metabolismo , Receptores Opioides/agonistas , Antagonistas de Narcóticos/uso terapéutico , Antagonistas de Narcóticos/farmacología
8.
Medicine (Baltimore) ; 103(28): e38932, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38996125

RESUMEN

Corticosteroid therapy for oxygen-free coronavirus disease 2019 (COVID-19) is not recommended due to its negative prognostic impact, but the efficacy of corticosteroids when limited to COVID-19 pneumonia is unclear. We aimed to evaluate the efficacy of corticosteroid monotherapy for patients with COVID-19 pneumonia without supplemental oxygen. We retrospectively reviewed patients with oxygen-free COVID-19 pneumonia at our institute between September 2020 and August 2021 and assessed the use of corticosteroids and the timing of initiation. We classified the patients into the following 2 groups: those who were initiated corticosteroids without developing respiratory failure (early steroid group) and those who were not (standard of care [SOC] group). We used inverse probability of treatment weighting (IPW) to balance between the groups. The primary outcome was the incidence of respiratory failure. A total of 144 patient records were reviewed; 63 patients were in the early steroid group and 81 patients were in the SOC group. Of all patients, 14 (22.2%) and 27 (33.3%) patients in the early steroid and SOC group, respectively, required supplemental oxygen (P = .192). After adjusted by the IPW method, 10 (16.0%) and 32 (40.1%) patients in the early steroid and SOC groups, respectively, required supplemental oxygen (P = .004). The logistic regression analysis indicated that early corticosteroid use was significantly associated with a decreased incidence of respiratory failure (odds ratio; 0.17, 95% confidence intervals; 0.06-0.46, P < .001). Corticosteroid monotherapy may suppress the development of exacerbation requiring oxygen supply in patients with oxygen-free COVID-19 pneumonia.


Asunto(s)
Corticoesteroides , COVID-19 , Insuficiencia Respiratoria , Humanos , Femenino , Masculino , Estudios Retrospectivos , COVID-19/complicaciones , Insuficiencia Respiratoria/tratamiento farmacológico , Persona de Mediana Edad , Corticoesteroides/uso terapéutico , Corticoesteroides/administración & dosificación , Anciano , SARS-CoV-2 , Tratamiento Farmacológico de COVID-19 , Terapia por Inhalación de Oxígeno/métodos , Resultado del Tratamiento
11.
Trials ; 25(1): 328, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38760804

RESUMEN

BACKGROUND: The SARS CoV-2 pandemic has resulted in more than 1.1 million deaths in the USA alone. Therapeutic options for critically ill patients with COVID-19 are limited. Prior studies showed that post-infection treatment of influenza A virus-infected mice with the liponucleotide CDP-choline, which is an essential precursor for de novo phosphatidylcholine synthesis, improved gas exchange and reduced pulmonary inflammation without altering viral replication. In unpublished studies, we found that treatment of SARS CoV-2-infected K18-hACE2-transgenic mice with CDP-choline prevented development of hypoxemia. We hypothesize that administration of citicoline (the pharmaceutical form of CDP-choline) will be safe in hospitalized SARS CoV-2-infected patients with hypoxemic acute respiratory failure (HARF) and that we will obtain preliminary evidence of clinical benefit to support a larger Phase 3 trial using one or more citicoline doses. METHODS: We will conduct a single-site, double-blinded, placebo-controlled, and randomized Phase 1/2 dose-ranging and safety study of Somazina® citicoline solution for injection in consented adults of any sex, gender, age, or ethnicity hospitalized for SARS CoV-2-associated HARF. The trial is named "SCARLET" (Supplemental Citicoline Administration to Reduce Lung injury Efficacy Trial). We hypothesize that SCARLET will show that i.v. citicoline is safe at one or more of three doses (0.5, 2.5, or 5 mg/kg, every 12 h for 5 days) in hospitalized SARS CoV-2-infected patients with HARF (20 per dose) and provide preliminary evidence that i.v. citicoline improves pulmonary outcomes in this population. The primary efficacy outcome will be the SpO2:FiO2 ratio on study day 3. Exploratory outcomes include Sequential Organ Failure Assessment (SOFA) scores, dead space ventilation index, and lung compliance. Citicoline effects on a panel of COVID-relevant lung and blood biomarkers will also be determined. DISCUSSION: Citicoline has many characteristics that would be advantageous to any candidate COVID-19 therapeutic, including safety, low-cost, favorable chemical characteristics, and potentially pathogen-agnostic efficacy. Successful demonstration that citicoline is beneficial in severely ill patients with SARS CoV-2-induced HARF could transform management of severely ill COVID patients. TRIAL REGISTRATION: The trial was registered at www. CLINICALTRIALS: gov on 5/31/2023 (NCT05881135). TRIAL STATUS: Currently enrolling.


Asunto(s)
COVID-19 , Citidina Difosfato Colina , Ensayos Clínicos Controlados Aleatorios como Asunto , SARS-CoV-2 , Adulto , Femenino , Humanos , Masculino , Administración Intravenosa , Betacoronavirus , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/complicaciones , COVID-19/complicaciones , Tratamiento Farmacológico de COVID-19 , Citidina Difosfato Colina/uso terapéutico , Método Doble Ciego , Hospitalización , Hipoxia/tratamiento farmacológico , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Neumonía Viral/complicaciones , Insuficiencia Respiratoria/tratamiento farmacológico , Insuficiencia Respiratoria/virología , SARS-CoV-2/efectos de los fármacos , Resultado del Tratamiento
12.
PLoS One ; 19(5): e0297137, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38722851

RESUMEN

BACKGROUND: Inhaled nitric oxide (iNO) has a beneficial effect on hypoxemic respiratory failure. The increased use of concurrent iNO and milrinone was observed. We aimed to report the trends of iNO use in the past 15 years in Taiwan and compare the first-year outcomes of combining iNO and milrinone to the iNO alone in very low birth weight preterm (VLBWP) infants under mechanical ventilation. METHODS: This nationwide cohort study enrolled preterm singleton infants with birth weight <1500g treated with iNO from 2004 to 2019. Infants were divided into two groups, with a combination of intravenous milrinone (Group 2, n = 166) and without milrinone (Group 1, n = 591). After propensity score matching (PSM), each group's sample size is 124. The primary outcomes were all-cause mortality and the respiratory condition, including ventilator use and duration. The secondary outcomes were preterm morbidities within one year after birth. RESULTS: After PSM, more infants in Group 2 needed inotropes. The mortality rate was significantly higher in Group 2 than in Group 1 from one month after birth till 1 year of age (55.1% vs. 13.5%) with the adjusted hazard ratio of 4.25 (95%CI = 2.42-7.47, p <0.001). For infants who died before 36 weeks of postmenstrual age (PMA), Group 2 had longer hospital stays compared to Group 1. For infants who survived after 36 weeks PMA, the incidence of moderate and severe bronchopulmonary dysplasia (BPD) was significantly higher in Group 2 than in Group 1. For infants who survived until one year of age, the incidence of pneumonia was significantly higher in Group 2 (28.30%) compared to Group 1 (12.62%) (p = 0.0153). CONCLUSION: Combined treatment of iNO and milrinone is increasingly applied in VLBWP infants in Taiwan. This retrospective study did not support the benefits of combining iNO and milrinone on one-year survival and BPD prevention. A future prospective study is warranted.


Asunto(s)
Recién Nacido de muy Bajo Peso , Milrinona , Óxido Nítrico , Humanos , Milrinona/administración & dosificación , Milrinona/uso terapéutico , Recién Nacido , Óxido Nítrico/administración & dosificación , Óxido Nítrico/uso terapéutico , Masculino , Administración por Inhalación , Femenino , Estudios Retrospectivos , Taiwán/epidemiología , Recien Nacido Prematuro , Insuficiencia Respiratoria/tratamiento farmacológico , Insuficiencia Respiratoria/mortalidad , Lactante , Respiración Artificial , Resultado del Tratamiento , Hipoxia/tratamiento farmacológico
13.
J Med Chem ; 67(11): 9173-9193, 2024 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-38810170

RESUMEN

While in the process of designing more effective synthetic opioid rescue agents, we serendipitously identified a new chemotype of potent synthetic opioid. Here, we report that conformational constraint of a piperazine ring converts a mu opioid receptor (MOR) antagonist into a potent MOR agonist. The prototype of the series, which we have termed atoxifent (2), possesses potent in vitro agonist activity. In mice, atoxifent displayed long-lasting antinociception that was reversible with naltrexone. Repeated dosing of atoxifent produced antinociceptive tolerance and a level of withdrawal like that of fentanyl. In rats, while atoxifent produced complete loss of locomotor activity like fentanyl, it failed to produce deep respiratory depression associated with fentanyl-induced lethality. Assessment of brain biodistribution demonstrated ample distribution of atoxifent into the brain with a Tmax of approximately 0.25 h. These results indicate enhanced safety for atoxifent-like molecules compared to fentanyl.


Asunto(s)
Analgésicos Opioides , Fentanilo , Receptores Opioides mu , Insuficiencia Respiratoria , Animales , Ratones , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/tratamiento farmacológico , Analgésicos Opioides/farmacología , Analgésicos Opioides/síntesis química , Analgésicos Opioides/química , Ratas , Masculino , Fentanilo/farmacología , Fentanilo/síntesis química , Fentanilo/química , Relación Estructura-Actividad , Piperazinas/farmacología , Piperazinas/química , Piperazinas/síntesis química , Piperazinas/uso terapéutico , Piperazinas/farmacocinética , Humanos , Ratas Sprague-Dawley , Distribución Tisular , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos , Naltrexona/farmacología , Naltrexona/análogos & derivados , Naltrexona/síntesis química , Naltrexona/química , Naltrexona/uso terapéutico
14.
BMC Pulm Med ; 24(1): 160, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38566026

RESUMEN

BACKGROUND: The coronavirus disease 2019 (COVID-19) pandemic has had a significant impact on global health and economies, resulting in millions of infections and deaths. This retrospective cohort study aimed to investigate the effect of antifibrotic agents (nintedanib and pirfenidone) on 1-year mortality in COVID-19 patients with acute respiratory failure. METHODS: Data from 61 healthcare organizations in the TriNetX database were analyzed. Adult patients with COVID-19 and acute respiratory failure were included. Patients with a pre-existing diagnosis of idiopathic pulmonary fibrosis before their COVID-19 diagnosis were excluded. The study population was divided into an antifibrotic group and a control group. Propensity score matching was used to compare outcomes, and hazard ratios (HR) for 1-year mortality were calculated. RESULTS: The antifibrotic group exhibited a significantly lower 1-year mortality rate compared to the control group. The survival probability at the end of the study was 84.42% in the antifibrotic group and 69.87% in the control group. The Log-Rank test yielded a p-value of less than 0.001. The hazard ratio was 0.434 (95% CI: 0.264-0.712), indicating a significant reduction in 1-year mortality in the antifibrotic group. Subgroup analysis demonstrated significantly improved 1-year survival in patients receiving nintedanib treatment and during periods when the Wuhan strain was predominant. DISCUSSION: This study is the first to demonstrate a survival benefit of antifibrotic agents in COVID-19 patients with acute respiratory failure. Further research and clinical trials are needed to confirm the efficacy of these antifibrotic agents in the context of COVID-19 and acute respiratory failure.


Asunto(s)
COVID-19 , Fibrosis Pulmonar Idiopática , Insuficiencia Respiratoria , Adulto , Humanos , Antifibróticos , Estudios Retrospectivos , Prueba de COVID-19 , Fibrosis Pulmonar Idiopática/complicaciones , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/diagnóstico , Insuficiencia Respiratoria/tratamiento farmacológico , Piridonas/uso terapéutico , Resultado del Tratamiento
15.
Altern Ther Health Med ; 30(7): 40-44, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38430175

RESUMEN

Background: Chronic obstructive pulmonary disease (COPD) poses a significant global health burden. Finding effective interventions for COPD is crucial to alleviating this health burden and enhancing patient outcomes and quality of life. Objective: This study aimed to evaluate the therapeutic efficacy of the combination of non-invasive ventilation and naloxone in the management of acute respiratory failure among patients with COPD. Methods: A retrospective analysis was conducted on the clinical data of 102 COPD patients experiencing acute respiratory failure who were treated at our hospital between October 2020 and October 2022. Patients were categorized into an observation group (receiving non-invasive ventilation combined with naloxone) and a control group (receiving non-invasive ventilation alone). Parameters such as lung function, blood gas levels, endocrine hormone concentrations, treatment efficacy, and patient prognosis were carefully recorded and compared. Results: The observation group demonstrated enhanced lung function, optimized endocrine hormone levels, and improved blood gas parameters compared to the control group. Following treatment, the observation group exhibited significant reductions in plasma renin activity (PRA), angiotensin II (AngII), aldosterone (ALD), and norepinephrine (NE) levels. The total effective rate was notably higher in the observation group. This group also presented higher scores for Acute Physiology and Chronic Health Evaluation II (APACHE II) and Chronic Obstructive Pulmonary Disease Assessment Test (CAT). Conclusions: The combination of non-invasive ventilation with naloxone emerged as a significantly effective strategy in managing acute respiratory failure in COPD patients. This approach led to improvements in lung function, endocrine hormone levels, and blood gas parameters and highlights its potential as an impactful treatment strategy for COPD patients experiencing acute respiratory failure.


Asunto(s)
Naloxona , Ventilación no Invasiva , Enfermedad Pulmonar Obstructiva Crónica , Insuficiencia Respiratoria , Humanos , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/terapia , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Naloxona/uso terapéutico , Masculino , Femenino , Ventilación no Invasiva/métodos , Estudios Retrospectivos , Anciano , Insuficiencia Respiratoria/terapia , Insuficiencia Respiratoria/tratamiento farmacológico , Persona de Mediana Edad , Terapia Combinada , Antagonistas de Narcóticos/uso terapéutico
16.
Lancet Haematol ; 11(4): e276-e286, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38452788

RESUMEN

BACKGROUND: Hypomethylating agents combined with venetoclax are effective regimens in patients with acute myeloid leukaemia who are ineligible for intensive chemotherapy. Decitabine and cedazuridine (ASTX727) is an oral formulation of decitabine that achieves equivalent area-under-curve exposure to intravenous decitabine. We performed a single centre phase 2 study to evaluate the efficacy and safety of ASTX727 plus venetoclax. METHODS: This study enrolled patients with newly diagnosed (frontline treatment group) acute myeloid leukaemia who were ineligible for intensive chemotherapy (aged ≥75 years, an Eastern Cooperative Oncology Group [ECOG] performance status of 2-3, or major comorbidities) or relapsed or refractory acute myeloid leukaemia. Being aged 18 years or older and having an ECOG performance status of 2 or less were requirements for the relapsed or refractory disease treatment cohort, without any limits in the number of previous lines of therapy. Treatment consisted of ASTX727 (cedazuridine 100 mg and decitabine 35 mg) orally for 5 days and venetoclax 400 mg orally for 21-28 days in 28-day cycles. The primary outcome was overall response rate of ASTX727 plus venetoclax. Living patients who have not completed cycle one were not evaluable for response. Safety was analysed in all patients who started treatment. This study was registered on ClinicalTrials.gov (NCT04746235) and is ongoing. The data cutoff date for this analysis was Sept 22, 2023. FINDINGS: Between March 16, 2021, and Sept 18, 2023, 62 patients were enrolled (49 frontline and 13 relapsed or refractory) with a median age of 78 years (IQR 73-82). 36 (58%) were male; 53 (85%) were White, 4 (6%) Black, 2 (3%) Asian and 3 (5%) other or did not answer. 48 (77%) of 62 patients were European LeukemiaNet 2022 adverse risk, 24 (39%) had antecedent myelodysplastic syndromes, 12 (19%) had previously failed a hypomethylating agent, ten (16%) had therapy-related acute myeloid leukaemia, and 11 (18%) had TP53 mutations. The median follow-up time was 18·3 months (IQR 8·8-23·3). The overall response rate was 30 (64%) of 47 patients (95% CI 49-77) in frontline cohort and six (46%) of 13 patients (19-75) in relapsed or refractory cohort. The most common grade 3 or worse treatment-emergent adverse events were febrile neutropenia in 11 (18%) of 62 patients, pneumonia in eight (13%), respiratory failure in five (8%), bacteraemia in four (6%), and sepsis in four (6%). Three deaths occurred in patients in remission (one sepsis, one gastrointestinal haemorrhage, and one respiratory failure) and were potentially treatment related. INTERPRETATION: ASTX727 plus venetoclax is an active fully oral regimen and safe in most older or unfit patients with acute myeloid leukaemia. Our findings should be confirmed in larger multicentric studies. FUNDING: MD Anderson Cancer Center Support Grant, Myelodysplastic Syndrome/Acute Myeloid Leukaemia Moon Shot, Leukemia SPORE, Taiho Oncology, and Astex Pharmaceuticals.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes , Combinación de Medicamentos , Leucemia Mieloide Aguda , Insuficiencia Respiratoria , Sepsis , Sulfonamidas , Uridina/análogos & derivados , Humanos , Masculino , Anciano , Anciano de 80 o más Años , Femenino , Decitabina/efectos adversos , Resultado del Tratamiento , Leucemia Mieloide Aguda/diagnóstico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/tratamiento farmacológico , Sepsis/inducido químicamente , Sepsis/tratamiento farmacológico
17.
J Appl Physiol (1985) ; 136(4): 821-843, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38385184

RESUMEN

Opioids are well-known to cause respiratory depression, but despite clinical evidence of dysphagia, the effects of opioids on swallow excitability and motor pattern are unknown. We tested the effects of the clinically relevant opioid buprenorphine on pharyngeal swallow and respiratory drive in male and female rats. We also evaluated the utility of 5-HT1A agonists (8-OH-DPAT and buspirone) to improve swallowing and breathing following buprenorphine administration. Experiments were performed on 44 freely breathing Sprague-Dawley rats anesthetized with sodium pentobarbital. Bipolar fine wire electrodes were inserted into the mylohyoid, thyroarytenoid, posterior cricoarytenoid, thyropharyngeus, and diaphragm muscles to measure electromyographic (EMG) activity of swallowing and breathing. We evaluated the hypotheses that swallowing varies by stimulus, opioids depress swallowing and breathing, and that 5-HT1A agonists improve these depressions. Our results largely confirmed the following hypotheses: 1) swallow-related EMG activity was larger during swallows elicited by esophageal distension plus oral water infusion than by either stimulus alone. 2) Buprenorphine depressed swallow in both sexes, but females were more susceptible to total swallow suppression. 3) Female animals were also more vulnerable to opioid-induced respiratory depression. 4) 8-OH-DPAT rescued breathing following buprenorphine-induced respiratory arrest, and pretreatment with the partial 5-HT1A agonist buspirone prevented buprenorphine-induced respiratory arrest in female animals. 5) 8-OH-DPAT enhanced mylohyoid and thyropharyngeus EMG amplitude during swallow but did not restore excitability of the swallow pattern generator following total suppression by buprenorphine. Our results highlight sex-specific and behavior-specific effects of buprenorphine and provide preclinical evidence of a 5HT1A agonist for the treatment of respiratory depression and dysphagia.NEW & NOTEWORTHY This is the first study, to our knowledge, to evaluate sex-specific effects of opioid administration on pharyngeal swallow. We expand on a small but growing number of studies that report a lower threshold for opioid-induced respiratory depression in females compared with males, and we are the first to produce this effect with the partial µ-opioid-receptor agonist buprenorphine. This is the first demonstration, to our knowledge, that activation of 5-HT1A receptors can improve swallow and breathing outcomes following systemic buprenorphine administration.


Asunto(s)
Buprenorfina , Trastornos de Deglución , Insuficiencia Respiratoria , Ratas , Femenino , Masculino , Animales , Analgésicos Opioides/farmacología , Serotonina , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , 8-Hidroxi-2-(di-n-propilamino)tetralin/uso terapéutico , Buspirona/efectos adversos , Ratas Sprague-Dawley , Insuficiencia Respiratoria/inducido químicamente , Insuficiencia Respiratoria/tratamiento farmacológico , Buprenorfina/efectos adversos
18.
Ann Clin Microbiol Antimicrob ; 23(1): 15, 2024 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-38350983

RESUMEN

PURPOSE: Multidrug-resistant (MDR) bacteria impose a considerable health-care burden and are associated with bronchiectasis exacerbation. This study investigated the clinical outcomes of adult patients with bronchiectasis following MDR bacterial infection. METHODS: From the Chang Gung Research Database, we identified patients with bronchiectasis and MDR bacterial infection from 2008 to 2017. The control group comprised patients with bronchiectasis who did not have MDR bacterial infection and were propensity-score matched at a 1:2 ratio. The main outcomes were in-hospital and 3-year mortality. RESULTS: In total, 554 patients with both bronchiectasis and MDR bacterial infection were identified. The types of MDR bacteria that most commonly affected the patients were MDR- Acinetobacter baumannii (38.6%) and methicillin-resistant Staphylococcus aureus (18.4%), Extended-spectrum-beta-lactamases (ESBL)- Klebsiella pneumoniae (17.8%), MDR-Pseudomonas (14.8%), and ESBL-E. coli (7.5%). Compared with the control group, the MDR group exhibited lower body mass index scores, higher rate of chronic bacterial colonization, a higher rate of previous exacerbations, and an increased use of antibiotics. Furthermore, the MDR group exhibited a higher rate of respiratory failure during hospitalization (MDR vs. control, 41.3% vs. 12.4%; p < 0.001). The MDR and control groups exhibited in-hospital mortality rates of 26.7% and 7.6%, respectively (p < 0.001); 3-year respiratory failure rates of 33.5% and 13.5%, respectively (p < 0.001); and 3-year mortality rates of 73.3% and 41.5%, respectively (p < 0.001). After adjustments were made for confounding factors, the infection with MDR and MDR bacteria species were determined to be independent risk factors affecting in-hospital and 3-year mortality. CONCLUSIONS: MDR bacteria were discovered in patients with more severe bronchiectasis and were independently associated with an increased risk of in-hospital and 3-year mortality. Given our findings, we recommend that clinicians identify patients at risk of MDR bacterial infection and follow the principle of antimicrobial stewardship to prevent the emergence of resistant bacteria among patients with bronchiectasis.


Asunto(s)
Infecciones Bacterianas , Bronquiectasia , Staphylococcus aureus Resistente a Meticilina , Insuficiencia Respiratoria , Adulto , Humanos , Escherichia coli , Antibacterianos/uso terapéutico , Antibacterianos/farmacología , Bronquiectasia/tratamiento farmacológico , Bronquiectasia/epidemiología , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/epidemiología , Infecciones Bacterianas/microbiología , Fibrosis , Insuficiencia Respiratoria/tratamiento farmacológico , Farmacorresistencia Bacteriana Múltiple
19.
BMJ Case Rep ; 17(2)2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38378591

RESUMEN

Patients on dialysis have limited treatment options for advanced lung cancer because some chemotherapeutic agents are unavailable due to renal dysfunction. A man in his 70s on peritoneal dialysis presented with persistent fever refractory to antibiotics for 2 weeks. Subsequent whole-body CT showed a 5 cm diameter mass in the right lower lobe of the lung with right-sided pleural effusion and osteolytic metastasis of the right iliac bone. The patient was diagnosed with squamous cell carcinoma (cT3N2M1b, stage IVB) harbouring the p.Gly719Ala point mutation on exon 18 of the epidermal growth factor receptor. The patient developed severe respiratory failure due to malignant lymphangitis after a bronchoscopy. He received 30 mg/day of afatinib, resulting in tumour shrinkage and recovery from respiratory failure. We advocate for aggressive screening of driver oncogenes in patients with lung cancer on dialysis, including those with squamous cell lung cancer.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Pulmonares , Linfangitis , Insuficiencia Respiratoria , Masculino , Humanos , Afatinib/uso terapéutico , Linfangitis/tratamiento farmacológico , Linfangitis/etiología , Diálisis Renal , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Carcinoma de Células Escamosas/complicaciones , Carcinoma de Células Escamosas/tratamiento farmacológico , Insuficiencia Respiratoria/terapia , Insuficiencia Respiratoria/tratamiento farmacológico , Pulmón/patología , Mutación
20.
Artículo en Inglés | MEDLINE | ID: mdl-38222320

RESUMEN

Introduction: Nirmatrelvir-ritonavir (NMV-r) and molnupiravir (MOL) were developed as out-patient anti-viral for mild COVID-19. There was limited data on their role in treating COVID-19 for hospitalized patients, especially among adult patients who are unvaccinated and had chronic respiratory diseases. Methods: A territory-wide retrospective study was conducted in Hong Kong to compare the efficacy of NMV-r and MOL against COVID-19 in unvaccinated adult patients with asthma, chronic obstructive pulmonary disease, bronchiectasis and interstitial lung diseases presenting with moderate COVID-19 from 16th February 2022 to 15th March 2023. Results: A total of 1354 patients were included, 738 received NMV-r and 616 received MOL. NMV-r was more effective in reducing 90-day mortality with adjusted hazard ratios (aHR) of 0.508 (95% confidence interval [CI] = 0.314-0.822, p = 0.006). Patients who received NMV-r also had significantly shorter length of stay (LOS) than those receiving MOL, with median LOS of 4 (Interquartile range [IQR] = 2-7) for NMV-r and 6 (IQR = 3-10) for MOL (p-value < 0.001). There was no statistically significant difference in the development of respiratory failure and severe respiratory failure in the two groups. Discussion: NMV-r was more effective than MOL among unvaccinated adults with chronic respiratory diseases who were hospitalized for moderate COVID-19 without hypoxaemia on admission.


Asunto(s)
COVID-19 , Citidina/análogos & derivados , Hidroxilaminas , Lactamas , Leucina , Nitrilos , Prolina , Enfermedad Pulmonar Obstructiva Crónica , Trastornos Respiratorios , Insuficiencia Respiratoria , Adulto , Humanos , Estudios Retrospectivos , Ritonavir/efectos adversos , Tratamiento Farmacológico de COVID-19 , Pacientes Ambulatorios , Insuficiencia Respiratoria/diagnóstico , Insuficiencia Respiratoria/tratamiento farmacológico , Insuficiencia Respiratoria/etiología , Antivirales/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...