Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
N Engl J Med ; 390(13): 1186-1195, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38598573

ABSTRACT

BACKGROUND: Nirmatrelvir in combination with ritonavir is an antiviral treatment for mild-to-moderate coronavirus disease 2019 (Covid-19). The efficacy of this treatment in patients who are at standard risk for severe Covid-19 or who are fully vaccinated and have at least one risk factor for severe Covid-19 has not been established. METHODS: In this phase 2-3 trial, we randomly assigned adults who had confirmed Covid-19 with symptom onset within the past 5 days in a 1:1 ratio to receive nirmatrelvir-ritonavir or placebo every 12 hours for 5 days. Patients who were fully vaccinated against Covid-19 and who had at least one risk factor for severe disease, as well as patients without such risk factors who had never been vaccinated against Covid-19 or had not been vaccinated within the previous year, were eligible for participation. Participants logged the presence and severity of prespecified Covid-19 signs and symptoms daily from day 1 through day 28. The primary end point was the time to sustained alleviation of all targeted Covid-19 signs and symptoms. Covid-19-related hospitalization and death from any cause were also assessed through day 28. RESULTS: Among the 1296 participants who underwent randomization and were included in the full analysis population, 1288 received at least one dose of nirmatrelvir-ritonavir (654 participants) or placebo (634 participants) and had at least one postbaseline visit. The median time to sustained alleviation of all targeted signs and symptoms of Covid-19 was 12 days in the nirmatrelvir-ritonavir group and 13 days in the placebo group (P = 0.60). Five participants (0.8%) in the nirmatrelvir-ritonavir group and 10 (1.6%) in the placebo group were hospitalized for Covid-19 or died from any cause (difference, -0.8 percentage points; 95% confidence interval, -2.0 to 0.4). The percentages of participants with adverse events were similar in the two groups (25.8% with nirmatrelvir-ritonavir and 24.1% with placebo). In the nirmatrelvir-ritonavir group, the most commonly reported treatment-related adverse events were dysgeusia (in 5.8% of the participants) and diarrhea (in 2.1%). CONCLUSIONS: The time to sustained alleviation of all signs and symptoms of Covid-19 did not differ significantly between participants who received nirmatrelvir-ritonavir and those who received placebo. (Supported by Pfizer; EPIC-SR ClinicalTrials.gov number, NCT05011513.).


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , COVID-19 Vaccines , COVID-19 , Adult , Humans , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , COVID-19/diagnosis , COVID-19/prevention & control , COVID-19/therapy , Diarrhea/chemically induced , Ambulatory Care , Dysgeusia/chemically induced , Vaccination , COVID-19 Vaccines/therapeutic use
2.
N Engl J Med ; 386(15): 1397-1408, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35172054

ABSTRACT

BACKGROUND: Nirmatrelvir is an orally administered severe acute respiratory syndrome coronavirus 2 main protease (Mpro) inhibitor with potent pan-human-coronavirus activity in vitro. METHODS: We conducted a phase 2-3 double-blind, randomized, controlled trial in which symptomatic, unvaccinated, nonhospitalized adults at high risk for progression to severe coronavirus disease 2019 (Covid-19) were assigned in a 1:1 ratio to receive either 300 mg of nirmatrelvir plus 100 mg of ritonavir (a pharmacokinetic enhancer) or placebo every 12 hours for 5 days. Covid-19-related hospitalization or death from any cause through day 28, viral load, and safety were evaluated. RESULTS: A total of 2246 patients underwent randomization; 1120 patients received nirmatrelvir plus ritonavir (nirmatrelvir group) and 1126 received placebo (placebo group). In the planned interim analysis of patients treated within 3 days after symptom onset (modified intention-to treat population, comprising 774 of the 1361 patients in the full analysis population), the incidence of Covid-19-related hospitalization or death by day 28 was lower in the nirmatrelvir group than in the placebo group by 6.32 percentage points (95% confidence interval [CI], -9.04 to -3.59; P<0.001; relative risk reduction, 89.1%); the incidence was 0.77% (3 of 389 patients) in the nirmatrelvir group, with 0 deaths, as compared with 7.01% (27 of 385 patients) in the placebo group, with 7 deaths. Efficacy was maintained in the final analysis involving the 1379 patients in the modified intention-to-treat population, with a difference of -5.81 percentage points (95% CI, -7.78 to -3.84; P<0.001; relative risk reduction, 88.9%). All 13 deaths occurred in the placebo group. The viral load was lower with nirmatrelvir plus ritonavir than with placebo at day 5 of treatment, with an adjusted mean difference of -0.868 log10 copies per milliliter when treatment was initiated within 3 days after the onset of symptoms. The incidence of adverse events that emerged during the treatment period was similar in the two groups (any adverse event, 22.6% with nirmatrelvir plus ritonavir vs. 23.9% with placebo; serious adverse events, 1.6% vs. 6.6%; and adverse events leading to discontinuation of the drugs or placebo, 2.1% vs. 4.2%). Dysgeusia (5.6% vs. 0.3%) and diarrhea (3.1% vs. 1.6%) occurred more frequently with nirmatrelvir plus ritonavir than with placebo. CONCLUSIONS: Treatment of symptomatic Covid-19 with nirmatrelvir plus ritonavir resulted in a risk of progression to severe Covid-19 that was 89% lower than the risk with placebo, without evident safety concerns. (Supported by Pfizer; ClinicalTrials.gov number, NCT04960202.).


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Lactams , Leucine , Nitriles , Proline , Ritonavir , Administration, Oral , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Disease Progression , Double-Blind Method , Hospitalization , Humans , Lactams/administration & dosage , Lactams/adverse effects , Lactams/therapeutic use , Leucine/administration & dosage , Leucine/adverse effects , Leucine/therapeutic use , Nitriles/administration & dosage , Nitriles/adverse effects , Nitriles/therapeutic use , Proline/administration & dosage , Proline/adverse effects , Proline/therapeutic use , Ritonavir/administration & dosage , Ritonavir/adverse effects , Ritonavir/therapeutic use , SARS-CoV-2 , Treatment Outcome , Vaccination , Viral Load/drug effects , Viral Protease Inhibitors/administration & dosage , Viral Protease Inhibitors/adverse effects , Viral Protease Inhibitors/therapeutic use
3.
Clin Trials ; 17(5): 535-544, 2020 10.
Article in English | MEDLINE | ID: mdl-32643966

ABSTRACT

BACKGROUND: The ICH E9(R1) addendum states that the strategy to account for intercurrent events should be included when defining an estimand, the treatment effect to be estimated based on the study objective. The estimator used to assess the treatment effect needs to be aligned with the estimand that accounted for intercurrent events. Regardless of the strategy, missing data resulting from patient premature withdrawal could undermine the robustness of the study results. Informative censoring due to dropouts in an events-based study is one such example. Sensitivity analyses using imputation methods are useful to examine the uncertainty due to informative censoring and address the robustness and strength of the study results. METHODS: We assessed the effect of premature patient withdrawal in the PRECISION study, a randomized non-inferiority clinical trial of patients with chronic arthritic pain that compared the cardiovascular safety of three nonsteroidal anti-inflammatory drugs-based treatment policies or paradigms. The protocol-defined use of concomitant or rescue medications was permitted since changes in pain medications due to insufficient analgesia were expected in patients in this long-term study. Anticipating that premature study discontinuations could potentially lead to informative censoring, a supplementary analysis was pre-specified in which censored outcomes due to the premature study discontinuation were imputed based on adverse events that were clinically associated with the primary endpoint (cardiovascular outcome based on the Antiplatelet Trialists Collaboration composite endpoint). Furthermore, tipping point analyses were conducted to test the robustness of the primary analysis results by assuming data censored not at random. The level of increase at which the primary study conclusion would change was estimated. RESULTS: For the analysis of time to first primary endpoint event through 30 months, 4065 out of the 24,081 enrolled patients were lost to follow-up, withdrew consent, or were no longer willing to participate in the study. These withdrawals occurred gradually and resulted in a cumulative total of 5893 censored patient-years of observation (10.2%). The rate of discontinuation and the baseline characteristics of the discontinued patients were similar across the three treatment groups. The non-inferiority conclusion from the primary analysis was confirmed in the supplementary analysis incorporating relevant adverse events. Furthermore, tipping point analyses demonstrated that in order to lose non-inferiority in the primary analysis, the risk of primary endpoint events during the censored observation time would have to increase by more than 2.7-fold in the celecoxib group while remaining constant in the other nonsteroidal anti-inflammatory drugs groups, demonstrating that the scenarios where the study results are invalid appear not plausible. CONCLUSIONS: Supplementary and sensitivity analyses presented to address informative censoring in PRECISION helped to further interpret and strengthen the study results.


Subject(s)
Arthritis/drug therapy , Data Interpretation, Statistical , Patient Dropouts/statistics & numerical data , Randomized Controlled Trials as Topic/methods , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cardiovascular Diseases/epidemiology , Censorship, Research , Endpoint Determination/methods , Endpoint Determination/statistics & numerical data , Female , Humans , Intention to Treat Analysis , Male , Middle Aged , Proportional Hazards Models , Randomized Controlled Trials as Topic/statistics & numerical data
4.
Kidney Int Rep ; 4(9): 1235-1247, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31517143

ABSTRACT

INTRODUCTION: This double-blind, randomized controlled trial compared the safety and efficacy of subcutaneous epoetin alfa-epbx, an epoetin alfa biosimilar, with the reference product, epoetin alfa, in hemodialysis patients with end-stage kidney disease (ESKD) and anemia who were receiving epoetin alfa maintenance treatment. METHODS: Eligible patients (n = 320) were randomized (1:1) to subcutaneous epoetin alfa-epbx or epoetin alfa in a titration phase; patients who demonstrated stable subcutaneous dosing (n = 246) were re-randomized to receive subcutaneous epoetin alfa-epbx or epoetin alfa 1 to 3 times per week in a 16-week maintenance phase. Co-primary endpoints were least-squares mean difference between treatments in mean weekly hemoglobin concentration and mean weekly epoetin dose per kilogram body weight (BW) during the last 4 weeks of treatment in the maintenance phase. RESULTS: The least-squares mean difference (95% confidence interval [CI]) between treatments in weekly hemoglobin was 0.04 g/dl (-0.17 to 0.24 g/dl) and weekly epoetin dose/kg BW was -2.34 U/kg per week (-14.51 to 9.82 U/kg per week). The 95% CIs were contained within the prespecified equivalence margins of ±0.5 g/dl (weekly hemoglobin) and ±45 U/kg per week (weekly epoetin dose/kg BW). In the epoetin alfa-epbx and epoetin alfa groups, respectively, 4.0% and 4.1% of patients required blood transfusions, 69.7% and 70.5% reported adverse events, 18.9% and 27.0% reported serious adverse events, and 3 and 2 deaths were reported. Five patients were confirmed positive for anti-recombinant human erythropoietin antibody, 2 of whom tested positive at baseline. All patients tested negative for neutralizing antibodies. CONCLUSIONS: This comparative clinical trial demonstrated equivalence in efficacy and similar safety of subcutaneously administered epoetin alfa-epbx to epoetin alfa.

5.
Clin J Am Soc Nephrol ; 13(8): 1204-1214, 2018 08 07.
Article in English | MEDLINE | ID: mdl-29921734

ABSTRACT

BACKGROUND AND OBJECTIVES: This study was conducted to compare the safety and efficacy of intravenous epoetin alfa-epbx, an epoetin alfa biosimilar, to epoetin alfa in patients on hemodialysis with ESKD and anemia. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: In this 24-week, multicenter, double-blind comparative efficacy and safety study, 612 patients on hemodialysis with ESKD and anemia who had stable hemoglobin and were receiving stable doses of intravenous epoetin alfa were randomized (1:1) to intravenous epoetin alfa or epoetin alfa-epbx. Dosing was adjusted according to the epoetin alfa prescribing information. The coprimary efficacy end points were the least squares mean difference between the two treatments in mean weekly hemoglobin level and mean weekly epoetin dose per kilogram of body weight during the last 4 weeks of treatment. RESULTS: The least squares mean difference between epoetin alfa-epbx and epoetin alfa in weekly hemoglobin was -0.12 g/dl and the 95% confidence interval (-0.25 to 0.01) was contained within the prespecified equivalence margin (-0.5 to 0.5 g/dl). The least squares mean difference between epoetin alfa-epbx and epoetin alfa in weekly epoetin dose per kilogram of body weight was 0.37 U/kg per week, and the 95% confidence interval (-10.40 to 11.13) was contained within the prespecified equivalence margin (-45 to 45 U/kg per week). Incidences of adverse events (77.1% versus 75.3%), serious adverse events (24.9% versus 27.0%), and deaths (n=5 versus 6) were similar between the epoetin alfa-epbx and epoetin alfa groups, respectively. Five patients tested positive for anti-recombinant human erythropoietin antibodies at baseline, and two additional patients (n=1 per group) developed anti-recombinant human erythropoietin antibodies while on study treatment. All patients tested negative for neutralizing antibodies, and no patient in either group experienced an event of pure red cell aplasia. CONCLUSIONS: This 24-week, comparative, clinical trial in patients on hemodialysis with ESKD and anemia demonstrated there is no clinically meaningful difference in efficacy or safety between epoetin alfa-epbx and epoetin alfa.


Subject(s)
Anemia/drug therapy , Biosimilar Pharmaceuticals/administration & dosage , Epoetin Alfa/analogs & derivatives , Epoetin Alfa/administration & dosage , Hematinics/administration & dosage , Administration, Intravenous , Anemia/etiology , Double-Blind Method , Female , Humans , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/therapy , Male , Middle Aged , Renal Dialysis
6.
Clin Ther ; 38(8): 1778-88, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27473384

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the pharmacokinetic (PK) and pharmacodynamic (PD) equivalence of single 100 U/kg subcutaneous doses of Epoetin Hospira and Epogen in healthy male subjects as part of an overall program to demonstrate biosimilarity of Epoetin Hospira to the reference product Epogen. METHODS: This single-center, open-label, randomized, 2-period, crossover study was conducted in 81 healthy male subjects. Subjects were randomized to Sequence 1, in which they received 100 U/kg of Epoetin Hospira or to sequence 2, in which they received 100 U/kg Epogen subcutaneously in the first study period and the alternative treatment in the second study period. Blood was collected for determination of baseline-adjusted epoetin concentrations (BAECs) for pharmacokinetics and for determination of reticulocyte percentage of total erythrocytes for pharmacodynamics throughout both study periods. The primary PK end points were the geometric mean ratios (GMRs) of the 2 treatments for AUC0-t and Cmax based on the BAEC, and the primary PD end points were the GMRs of the 2 treatments for AUC0-t and Cmax for reticulocyte percentage. FINDINGS: The GMRs of Epoetin Hospira to Epogen for the BAEC-derived PK parameters were 1.05 for AUC0-t with a 90% CI of 1.01 to 1.11, and 1.09 for Cmax with a 90% CI of 1.01 to 1.18, with both 90% CIs contained within the prespecified equivalence margin of 0.80 to 1.25. The GMRs (Epoetin Hospira/Epogen) for the reticulocyte percentage-derived PD parameters were 1.01 for AUC0-t with a 95% CI of 0.98 to 1.05, and 1.02 for Cmax with a 95% CI of 0.98 to 1.06, with both 95% CIs contained within the prespecified equivalence margin of 0.80 to 1.25. Overall, the adverse events were of similar frequency (11.7% and 13.9% for Epoetin Hospira and Epogen, respectively) and severity between treatments. One subject had a positive anti- recombinant human erythropoietin antibody result by radioimmunoprecipitation assay before dosing and throughout the conduct of the study with negative neutralizing antibodies and with no evidence of clinical deterioration or impact on the pharmacokinetics, pharmacodynamics, or safety. IMPLICATIONS: The results of this study established the PK and PD equivalence of single 100 U/kg subcutaneous doses of the proposed biosimilar Epoetin Hospira to the reference product Epogen in healthy male subjects and support the overall demonstration of biosimilarity of Epoetin Hospira and Epogen.


Subject(s)
Epoetin Alfa/administration & dosage , Recombinant Proteins/administration & dosage , Adolescent , Adult , Antibodies, Neutralizing/immunology , Cross-Over Studies , Epoetin Alfa/pharmacokinetics , Epoetin Alfa/pharmacology , Humans , Male , Middle Aged , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Young Adult
7.
J Pediatr ; 164(2): 276-82.e1-3, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24238862

ABSTRACT

OBJECTIVE: To investigate the safety, efficacy, and pharmacokinetic profile of dexmedetomidine in preterm and full-term neonates ≥ 28 to ≤ 44 weeks gestational age. STUDY DESIGN: Forty-two intubated, mechanically ventilated patients (n = 42) were grouped by gestational age into group I (n = 18), ≥ 28 to <36 weeks, and group II (n = 24), ≥ 36 to ≤ 44 weeks. Within each age group, there were 3 escalating dose levels, including a loading dose (LD, µg/kg) followed by a maintenance dose (MD, µg · kg(-1) · h(-1)) for 6-24 hours: level 1, 0.05 LD/MD; level 2, 0.1 LD/MD; and level 3, 0.2 LD/MD. The primary endpoint was the number of patients requiring sedation as determined by the Neonatal Pain, Agitation, Sedation Scale. RESULTS: During dexmedetomidine infusion, 5% of Neonatal Pain, Agitation, Sedation Scale scores were >3, indicating agitation/pain, with 4 patients (10%) requiring more sedation and 17 (40%) requiring more analgesia. Though there was significant variability in pharmacokinetic variables, group I appeared to have lower weight-adjusted plasma clearance (0.3 vs 0.9 L · h(-1) · kg(-1)) and increased elimination half-life (7.6 vs 3.2 hours) compared with group II. Fifty-six adverse events (AEs) were reported in 26 patients (62%); only 3 AEs (5%) were related to dexmedetomidine. There were no serious AEs and no AEs or hemodynamic changes requiring dexmedetomidine discontinuation. CONCLUSION: Dexmedetomidine is effective for sedating preterm and full-term neonates and is well-tolerated without significant AEs. Preterm neonates had decreased plasma clearance and longer elimination half-life.


Subject(s)
Dexmedetomidine/pharmacokinetics , Infant, Premature, Diseases/drug therapy , Infant, Premature , Dexmedetomidine/administration & dosage , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Gestational Age , Half-Life , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacokinetics , Infant, Newborn , Infant, Premature, Diseases/blood , Infusions, Intravenous , Male , Pain Measurement , Prospective Studies , Respiration, Artificial , Treatment Outcome
8.
Crit Care Med ; 38(12): 2311-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20838332

ABSTRACT

OBJECTIVES: To determine the relationship between the number of delirium days experienced by intensive care patients and mortality, ventilation time, and intensive care unit stay. DESIGN: Prospective cohort analysis. SETTING: Patients from 68 intensive care units in five countries. PATIENTS: Three hundred fifty-four medical and surgical intensive care patients enrolled in the SEDCOM (Safety and Efficacy of Dexmedetomidine Compared with Midazolam) trial received a sedative study drug and completed at least one delirium assessment. INTERVENTIONS: Sedative drug interruption and/or titration to maintain light sedation with daily arousal and delirium assessments up to 30 days of mechanical ventilation. MEASUREMENTS AND MAIN RESULTS: The primary outcome was all-cause 30-day mortality. Multivariable analysis using Cox regression incorporating delirium duration as a time-dependent variable and adjusting for eight relevant baseline covariates was conducted to quantify the relationship between number of delirium days and the three main outcomes. Overall, delirium was diagnosed in 228 of 354 patients (64.4%). Mortality was significantly lower in patients without delirium compared to those with delirium (15 of 126 [11.9%] vs. 69 of 228 [30.3%]; p<.001). Similarly, the median time to extubation and intensive care unit discharge were significantly shorter among nondelirious patients (3.6 vs. 10.7 days [p<.001] and 4 vs. 16 days [p<.001], respectively). In multivariable analysis, the duration of delirium exhibited a nonlinear relationship with mortality (p=.02), with the strongest association observed in the early days of delirium. In comparison to 0 days of delirium, an independent dose-response increase in mortality was observed, which increased from 1 day of delirium (hazard ratio, 1.70; 95% confidence interval, 1.27-2.29; p<.001), 2 days of delirium (hazard ratio, 2.69; confidence interval, 1.58-4.57; p<.001), and ≥3 days of delirium (hazard ratio, 3.37; confidence interval, 1.92-7.23; p<.001). Similar independent relationships were observed between delirium duration and ventilation time and intensive care length of stay. CONCLUSIONS: In ventilated and lightly sedated intensive care unit patients, the duration of delirium was the strongest independent predictor of death, ventilation time, and intensive care unit stay after adjusting for relevant covariates.


Subject(s)
Cause of Death , Delirium/mortality , Hospital Mortality/trends , Hypnotics and Sedatives/therapeutic use , Respiration, Artificial/methods , Aged , Cohort Studies , Conscious Sedation/adverse effects , Conscious Sedation/methods , Critical Care/methods , Critical Illness , Delirium/diagnosis , Delirium/therapy , Dexmedetomidine/administration & dosage , Dexmedetomidine/adverse effects , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Humans , Hypnotics and Sedatives/adverse effects , Intensive Care Units , Kaplan-Meier Estimate , Length of Stay , Male , Midazolam/administration & dosage , Midazolam/adverse effects , Middle Aged , Multivariate Analysis , Predictive Value of Tests , Proportional Hazards Models , Prospective Studies , Respiration, Artificial/adverse effects , Risk Assessment
9.
Am J Ther ; 17(6): 586-95, 2010.
Article in English | MEDLINE | ID: mdl-20535016

ABSTRACT

GABA-mediated sedatives have respiratory depressant properties that may be detrimental in patients with difficult airways. In this randomized, double-blind, multicenter, Phase IIIb Food and Drug Administration study, safety and efficacy of dexmedetomidine compared with placebo were evaluated as the primary sedative for awake fiberoptic intubation (AFOI). Patients were randomized to receive dexmedetomidine or saline. Patients were sedated with dexmedetomidine or rescue midazolam to achieve targeted sedation (Ramsay Sedation Scale ≥ 2) before topicalization and throughout AFOI. Primary efficacy endpoint was percentage of patients requiring rescue midazolam; secondary efficacy endpoints were total dose of rescue midazolam, percentage requiring additional rescue nonmidazolam medications, anesthesiologist's assessment of ease of subject care, and patient recall and satisfaction 24 hours postoperatively. Less rescue midazolam was required to maintain Ramsay Sedation Scale ≥2 (47.3% vs. 86.0%, P < 0.001), and supplemental midazolam dose was lower (1.07 ± 1.5 mg vs. 2.85 ± 3.0 mg, P < 0.001) with dexmedetomidine compared with placebo. More Mallampati Class IV patients treated with dexmedetomidine were successfully intubated without midazolam than with placebo (66.7% vs. 8.3%, P = 0.009). Dexmedetomidine decreased blood pressure and heart rate compared with placebo patients sedated with midazolam. Patients and anesthesiologists showed favorable satisfaction responses in both groups. Adverse events and patient recall were similar in both groups. Dexmedetomidine is effective as the primary sedative in patients undergoing AFOI. Some patients may require small supplemental doses of midazolam, in addition to dexmedetomidine, to achieve sufficient sedation for AFOI. Dexmedetomidine provides another AFOI option for sedation of patients with difficult airways.


Subject(s)
Dexmedetomidine/therapeutic use , Fiber Optic Technology , Hypnotics and Sedatives/therapeutic use , Blood Pressure/drug effects , Conscious Sedation , Dexmedetomidine/administration & dosage , Dexmedetomidine/adverse effects , Double-Blind Method , Female , Heart Rate/drug effects , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Intubation, Intratracheal , Male , Midazolam/administration & dosage , Midazolam/therapeutic use , Middle Aged , Patient Satisfaction , Prospective Studies
10.
Anesth Analg ; 110(1): 47-56, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19713256

ABSTRACT

BACKGROUND: Dexmedetomidine (DEX) is increasingly being used as a sedative for monitored anesthesia care (MAC) because of its analgesic properties, "cooperative sedation," and lack of respiratory depression. In this randomized, multicenter, double-blind, Phase III Food and Drug Administration study, we evaluated the safety and efficacy of two doses of DEX for sedation of patients undergoing a broad range of surgical or diagnostic procedures requiring MAC. METHODS: Three hundred twenty-six patients were randomized 2:2:1 to DEX 0.5 microg/kg, DEX 1 microg/kg, or saline placebo initial loading dose, followed by a maintenance infusion of 0.2-1.0 microg x kg(-1) x h(-1) of DEX (or equivalent volume of saline) titrated to a targeted level of sedation (< or = 4 on the Observer's Assessment of Alertness/Sedation Scale [OAA/S]). Study drug was started at least 15 min before placement of regional or local anesthetic block. Midazolam was given for OAA/S > 4 and fentanyl for pain. The primary end-point was the percentage of patients not requiring rescue midazolam. RESULTS: Significantly fewer patients in the 0.5- and 1-microg/kg DEX groups required supplemental midazolam compared with placebo (59.7% [80/134], 45.7% [59/129] vs 96.8% [61/63], respectively; P < 0.001) and at lower doses to achieve an OAA/S < or = 4 before and during surgery compared with the saline group (1.4 and 0.9 mg vs 4.1 mg, respectively; P < 0.001, each group compared with placebo). Both DEX groups required significantly less fentanyl (84.8 and 83.6 microg vs 144.4 microg, respectively; P < 0.001, for both DEX groups versus placebo) for all surgical subtypes. Anesthesiologists indicated significantly increased ease of achieving and maintaining targeted sedation in both DEX groups compared with placebo with midazolam (P < 0.001). Patient satisfaction was significantly higher with DEX (P < or = 0.009, both groups versus placebo). Common adverse events with DEX were protocol-defined bradycardia and hypotension that were predominately mild to moderate in severity. The incidence of clinically significant respiratory depression (defined as a respiratory rate of < 8 or an oxygen saturation of < 90%) was lower in DEX-treated patients (P = 0.018, for both groups versus placebo). CONCLUSIONS: DEX is an effective baseline sedative for patients undergoing MAC for a broad range of surgical procedures providing better patient satisfaction, less opioid requirements, and less respiratory depression than placebo rescued with midazolam and fentanyl.


Subject(s)
Anesthesia , Dexmedetomidine , Hypnotics and Sedatives , Adolescent , Adult , Aged , Aged, 80 and over , Anesthesia, Intravenous , Anesthetics, Intravenous , Dexmedetomidine/administration & dosage , Dexmedetomidine/adverse effects , Double-Blind Method , Female , Fentanyl , Hemodynamics/drug effects , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Male , Middle Aged , Monitoring, Intraoperative , Patient Satisfaction , Prospective Studies , Young Adult
11.
Crit Care Med ; 38(2): 497-503, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19789442

ABSTRACT

OBJECTIVE: To compare the intensive care unit costs and determine factors influencing these costs in mechanically ventilated patients randomized to dexmedetomidine or midazolam by continuous infusion. DESIGN: Cost minimization analysis of a double-blind, multicenter clinical trial randomizing patients 2:1 to receive dexmedetomidine or midazolam from the institutional perspective. SETTING: Sixty-eight intensive care units in the United States, Australia, New Zealand, Brazil, and Argentina. PATIENTS: A total of 366 intubated intensive care unit patients anticipated to require sedation for >24 hrs. MEASUREMENTS AND MAIN RESULTS: Intensive care unit resource use was compared within the two treatment arms, using the U.S. representative costs for these resources. The analyses characterized patient costs from start of study drug until intensive care unit discharge including costs associated with the intensive care unit stay, costs during mechanical ventilation, study drug acquisition cost, and costs of treating adverse drug reactions probably or possibly related to study drugs. Blinded to treatment group, costs were calculated using Medicare reimbursement schedules, average IMS drug costs, expert opinion, and peer-reviewed literature. Censored lengths of intensive care unit stay and mechanical ventilation were imputed, using a nonparametric adjustment algorithm. Crude and multivariate median regressions were performed to relate intensive care unit cost and treatment. Including drug acquisition cost, sedation with dexmedetomidine was associated with a median total intensive care unit cost savings of $9679 (confidence interval, $2314-$17,045) compared with midazolam. The primary cost drivers were reduced costs of intensive care unit stay (median savings, $6584, 95% confidence interval, $727-$12,440) and reduced costs of mechanical ventilation (median savings, $2958, 95% confidence interval, $698-$5219). CONCLUSIONS: Continuous sedation with dexmedetomidine results in significantly lower total intensive care unit costs compared with midazolam infusion for intensive care unit sedation, primarily due to decreased intensive care unit stay costs and reduced mechanical ventilation costs.


Subject(s)
Critical Care/economics , Dexmedetomidine/economics , Hypnotics and Sedatives/economics , Midazolam/economics , Cost Savings/economics , Cost-Benefit Analysis , Critical Care/methods , Dexmedetomidine/adverse effects , Dexmedetomidine/therapeutic use , Double-Blind Method , Female , Humans , Hypnotics and Sedatives/adverse effects , Hypnotics and Sedatives/therapeutic use , Intensive Care Units/economics , Long-Term Care/economics , Long-Term Care/methods , Male , Midazolam/adverse effects , Midazolam/therapeutic use , Respiration, Artificial/economics , Respiration, Artificial/methods , Treatment Outcome , United States
12.
JAMA ; 301(5): 489-99, 2009 Feb 04.
Article in English | MEDLINE | ID: mdl-19188334

ABSTRACT

CONTEXT: Gamma-aminobutyric acid receptor agonist medications are the most commonly used sedatives for intensive care unit (ICU) patients, yet preliminary evidence indicates that the alpha(2) agonist dexmedetomidine may have distinct advantages. OBJECTIVE: To compare the efficacy and safety of prolonged sedation with dexmedetomidine vs midazolam for mechanically ventilated patients. DESIGN, SETTING, AND PATIENTS: Prospective, double-blind, randomized trial conducted in 68 centers in 5 countries between March 2005 and August 2007 among 375 medical/surgical ICU patients with expected mechanical ventilation for more than 24 hours. Sedation level and delirium were assessed using the Richmond Agitation-Sedation Scale (RASS) and the Confusion Assessment Method for the ICU. INTERVENTIONS: Dexmedetomidine (0.2-1.4 microg/kg per hour [n = 244]) or midazolam (0.02-0.1 mg/kg per hour [n = 122]) titrated to achieve light sedation (RASS scores between -2 and +1) from enrollment until extubation or 30 days. MAIN OUTCOME MEASURES: Percentage of time within target RASS range. Secondary end points included prevalence and duration of delirium, use of fentanyl and open-label midazolam, and nursing assessments. Additional outcomes included duration of mechanical ventilation, ICU length of stay, and adverse events. RESULTS: There was no difference in percentage of time within the target RASS range (77.3% for dexmedetomidine group vs 75.1% for midazolam group; difference, 2.2% [95% confidence interval {CI}, -3.2% to 7.5%]; P = .18). The prevalence of delirium during treatment was 54% (n = 132/244) in dexmedetomidine-treated patients vs 76.6% (n = 93/122) in midazolam-treated patients (difference, 22.6% [95% CI, 14% to 33%]; P < .001). Median time to extubation was 1.9 days shorter in dexmedetomidine-treated patients (3.7 days [95% CI, 3.1 to 4.0] vs 5.6 days [95% CI, 4.6 to 5.9]; P = .01), and ICU length of stay was similar (5.9 days [95% CI, 5.7 to 7.0] vs 7.6 days [95% CI, 6.7 to 8.6]; P = .24). Dexmedetomidine-treated patients were more likely to develop bradycardia (42.2% [103/244] vs 18.9% [23/122]; P < .001), with a nonsignificant increase in the proportion requiring treatment (4.9% [12/244] vs 0.8% [1/122]; P = .07), but had a lower likelihood of tachycardia (25.4% [62/244] vs 44.3% [54/122]; P < .001) or hypertension requiring treatment (18.9% [46/244] vs 29.5% [36/122]; P = .02). CONCLUSIONS: There was no difference between dexmedetomidine and midazolam in time at targeted sedation level in mechanically ventilated ICU patients. At comparable sedation levels, dexmedetomidine-treated patients spent less time on the ventilator, experienced less delirium, and developed less tachycardia and hypertension. The most notable adverse effect of dexmedetomidine was bradycardia. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00216190 Published online February 2, 2009 (doi:10.1001/jama.2009.56).


Subject(s)
Adrenergic alpha-Agonists , Conscious Sedation , Critical Illness , Dexmedetomidine , GABA Modulators , Hypnotics and Sedatives , Midazolam , Respiration, Artificial , APACHE , Adrenergic alpha-Agonists/administration & dosage , Adrenergic alpha-Agonists/adverse effects , Aged , Conscious Sedation/methods , Delirium/epidemiology , Dexmedetomidine/administration & dosage , Dexmedetomidine/adverse effects , Double-Blind Method , Female , GABA Modulators/administration & dosage , GABA Modulators/adverse effects , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/adverse effects , Intensive Care Units , Kaplan-Meier Estimate , Length of Stay , Male , Midazolam/administration & dosage , Midazolam/adverse effects , Middle Aged , Prospective Studies
13.
Arthritis Rheum ; 48(12): 3328-37, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14673984

ABSTRACT

OBJECTIVE: To evaluate the efficacy and safety of tacrolimus as monotherapy in controlling the signs and symptoms of patients with rheumatoid arthritis (RA). METHODS: This was a 6-month, phase III, double-blind, multicenter study. Patients with active RA who had discontinued all disease-modifying antirheumatic drugs (DMARDs) for an appropriate washout period (at least 1 month) and who, after the washout period, had a stable joint count (at least 10 tender/painful joints and 7 swollen joints) were stratified according to DMARD intolerance or DMARD resistance, and randomized to receive a single daily oral dose of placebo, tacrolimus 2 mg, or tacrolimus 3 mg. RESULTS: A total of 464 patients received at least 1 dose of study drug. Baseline characteristics were similar among the 3 treatment groups. American College of Rheumatology 20% improvement (ACR20) success (defined as completion of 6 months of treatment and an ACR20 response at the month 6 visit) for the placebo, tacrolimus 2 mg, and tacrolimus 3 mg groups was 10.2%, 18.8% (P < 0.05 versus placebo), and 26.8% (P < 0.0005 versus placebo), respectively. At the end of treatment, the ACR20 and ACR50 response rates in the 3-mg group were 32.0% (P < 0.005 versus placebo) and 11.8% (P < 0.05 versus placebo), respectively. DMARD-intolerant patients had better ACR response rates than did DMARD-resistant patients. Although serum creatinine levels increased by >/=40% from baseline at some time during the trial in 20% and 29% of patients receiving tacrolimus 2 mg/day and 3 mg/day, respectively, the serum creatinine level remained within the normal range throughout the trial in approximately 90% of patients. CONCLUSION: Tacrolimus, at dosages of both 2 mg/day and 3 mg/day, is efficacious and safe as monotherapy for patients with active RA, but treatment with the 3-mg dose of tacrolimus resulted in generally better ACR response rates.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Immunosuppressive Agents/administration & dosage , Tacrolimus/administration & dosage , Administration, Oral , Double-Blind Method , Drug Resistance , Female , Humans , Immunosuppressive Agents/adverse effects , Male , Middle Aged , Tacrolimus/adverse effects , Treatment Outcome
14.
Arthritis Rheum ; 48(10): 2763-8, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14558080

ABSTRACT

OBJECTIVE: To assess the safety of tacrolimus used in combination with oral methotrexate (MTX) to control the signs and symptoms of rheumatoid arthritis (RA) in patients whose disease remains active despite treatment with MTX. METHODS: This was a multicenter open-label study conducted at 13 US sites. Eighty patients who at baseline had active RA (mean tender/painful joint count 29.4, mean swollen joint count 17.4, mean erythrocyte sedimentation rate 25.1 mm/hour) despite treatment for >/=1 month with a stable, maximally tolerated dosage of oral MTX (/=30% maximum increase in the Cr level from baseline during the study, with the Cr level in 3 patients (3.8%) exceeding the range considered normal for their age and sex. The maximum Cr level during the study was 1.8 mg/dl. The ACR20 clinical response rate at the end of treatment was 52.5% (95% confidence interval 41.6-63.4%). CONCLUSION: In patients whose active RA persists despite treatment with MTX, tacrolimus in combination with MTX is safe and well-tolerated and provides clinical benefit.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Immunosuppressive Agents/administration & dosage , Methotrexate/administration & dosage , Tacrolimus/administration & dosage , Administration, Oral , Creatinine/blood , Drug Therapy, Combination , Female , Humans , Hyperglycemia/diagnosis , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/blood , Kidney/drug effects , Male , Methotrexate/adverse effects , Middle Aged , Tacrolimus/adverse effects , Tacrolimus/blood , Treatment Outcome
15.
Am J Clin Nutr ; 75(6): 1012-6, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12036807

ABSTRACT

BACKGROUND: Frame size is a description of the supportive structure of the skeleton that is used to adjust for skeletal mass and size in measures of body composition and weight. OBJECTIVE: Data from the Fels Longitudinal Study were used to investigate the relation between bicristal, elbow, knee, biacromial, and wrist breadths and measures of total body fat (TBF), fat-free mass (FFM), bone mineral content (BMC), and bone mineral density (BMD) from dual-energy X-ray absorptiometry. DESIGN: The sample consisted of cross-sectional data from 224 white men and 277 white women aged 23-65 y. Multiple regressions were conducted with stature-adjusted measures of body composition and bone mineral status as dependent variables and age and frame size as independent variables. RESULTS: Frame-size measures were significantly and positively associated with all body-composition and bone mineral measures in bivariate analyses. In both men and women, the significant models explained more of the variance in measures of TBF (R2 = 0.51 and 0.66, respectively) and FFM (R2 = 0.35 and 0.39, respectively) than in measures of BMC (R2 = 0.18 and 0.23, respectively) and BMD (R2 = 0.08 and 0.18, respectively). Bicristal, knee, and wrist breadths were associated with TBF, and biacromial, knee, and wrist breadths were positively associated with FFM. Biacromial breadth was positively associated with BMC and BMD. CONCLUSIONS: Frame size was more closely associated with TBF and FFM than with BMC and BMD. The association between frame size and body composition seems to be more structural than substantive. The relations between frame size and BMC and BMD are weak and apparently not related to body composition.


Subject(s)
Body Composition/physiology , Body Constitution/physiology , Bone Density/physiology , Absorptiometry, Photon , Adipose Tissue/anatomy & histology , Adipose Tissue/physiology , Adult , Aged , Anthropometry , Cross-Sectional Studies , Female , Humans , Longitudinal Studies , Male , Middle Aged , Regression Analysis , Sex Characteristics
16.
Am J Hum Biol ; 10(5): 589-598, 1998.
Article in English | MEDLINE | ID: mdl-28561536

ABSTRACT

This study determines: (1) patterns of change from childhood to young adulthood in body mass index (BMI), systolic blood pressure (SBP), and diastolic blood pressure (DBP), (2) effects of elevated BMI values on changes in blood pressures (BP), (3) extent of tracking for SBP, DBP, and BMI, and (4) prediction of future risk for elevated BP from earlier values. Annual serial BP and BMI data were available for 198 white females, ages 8-22 years, enrolled in the Fels Longitudinal Study. Patterns of change in BMI were described by a random effects model with a time series model for the correlated residuals. Serial BMI measures were differentiated from age-specific means to measure relative individual BMI levels. Serial BP were analyzed using a similar model to the BMI where relative individual BMI levels were included as an explanatory variable. There was a general increasing trend for SBP, DBP, and BMI from 8-22 years, but the rates of increase declined with age. At the same chronological age, early menarche females had a significantly greater BMI mean value than late menarche females. An average increase of 1 kg/m2 in deviation from BMI population means resulted in an average increase of 1.2 mmHg in SBP and 0.6 mmHg in DBP. Having SBP and DBP levels 1 standard deviation above mean levels, relative to females at mean levels, as early as age 9 represents an odds ratio of 2 for exceeding national 75th percentile levels of SBP and DBP at age 21. Am. J. Hum. Biol. 10: 589-598, 1998. © 1998 Wiley-Liss, Inc.

SELECTION OF CITATIONS
SEARCH DETAIL
...