Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Int J Clin Pract ; 68(9): 1152-60, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24703228

ABSTRACT

BACKGROUND: The reduced availability of data from non-Western countries limits our ability to understand attention-deficit/hyperactivity disorder (ADHD) treatment outcomes, specifically, adherence and persistence of ADHD in children and adolescents. This analysis assessed predictors of treatment outcomes in a non-Western cohort of patients with ADHD treated with atomoxetine or methylphenidate. METHODS: Data from a 12-month, prospective, observational study in outpatients aged 6-17 years treated with atomoxetine (N = 234) or methylphenidate (N = 221) were analysed post hoc to determine potential predictors of treatment outcomes. Participating countries included the Russian Federation, China, Taiwan, Egypt, United Arab Emirates and Lebanon. Factors associated with remission were analysed with stepwise multiple logistic regression and classification and regression trees (CART). Cox proportional hazards models with propensity score adjustment assessed differences in atomoxetine persistence among initial-dose cohorts. RESULTS: In patients treated with atomoxetine who had available dosing information (N = 134), Cox proportional hazards revealed lower (< 0.5 mg/kg) initial dose was significantly associated with shorter medication persistence (p < 0.01). multiple logistic regression analysis revealed greater rates of remission for atomoxetine-treated patients were associated with age (older), country (United Arab Emirates) and gender (female) (all p < 0.05). CART analysis confirmed older age and lack of specific phobias were associated with greater remission rates. For methylphenidate, greater baseline weight (highly correlated with the age factor found for atomoxetine) and prior atomoxetine use were associated with greater remission rates. CONCLUSIONS: These findings may help clinicians assess factors upon initiation of ADHD treatment to improve course prediction, proper dosing and treatment adherence and persistence. TRIAL REGISTRATION: Observational study, therefore no registration.


Subject(s)
Atomoxetine Hydrochloride/therapeutic use , Attention Deficit Disorder with Hyperactivity/drug therapy , Methylphenidate/therapeutic use , Adolescent , China , Egypt , Female , Humans , Lebanon , Male , Propylamines/therapeutic use , Prospective Studies , Russia , Taiwan , Treatment Outcome , United Arab Emirates
2.
Value Health ; 17(7): A585-6, 2014 Nov.
Article in English | MEDLINE | ID: mdl-27201987
4.
Diabetes Obes Metab ; 14(7): 626-33, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22321776

ABSTRACT

AIM: This observational study evaluated the clinical effectiveness of exenatide BID (exenatide) vs. insulin glargine (glargine) in patients with type 2 diabetes mellitus in ambulatory clinical practice. METHODS: Retrospective analyses were conducted using an electronic medical record (EMR) database among adult patients with type 2 diabetes mellitus initiating exenatide or glargine between 1 November 2006 and 30 April 2009. The cohorts were propensity-score matched to control baseline demographics, clinical measures, health status and medication use. The changes from baseline to a 12-month follow-up period for A1C (primary outcome), weight, body mass index (BMI), blood pressure and lipid levels were compared between the matched cohorts using paired tests. RESULTS: Propensity-score matching between the exenatide (n = 4494) and glargine (n = 5424) cohorts led to 2683 matched pairs with comparable characteristics, including age, gender and baseline clinical values. The exenatide cohort achieved a greater mean reduction in A1C (-0.6% vs. -0.4%, p < 0.01), weight (-2.6 kg vs. -0.2 kg, p < 0.01), BMI (-0.8 kg/m(2) vs. -0.04 kg/m(2) , p < 0.01) and systolic blood pressure (SBP) (-1.8 mmHg vs. -0.1 mmHg, p < 0.01) in the follow-up period. The changes in diastolic blood pressure and lipid levels were not significantly different between cohorts. CONCLUSIONS: Compared to glargine, exenatide-treated patients experienced significant reductions in A1C, weight, BMI and SBP. Acknowledging the limitations of observational research, exenatide showed greater clinical effectiveness than glargine from a large EMR database in the ambulatory care setting.


Subject(s)
Ambulatory Care , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Hypoglycemic Agents/therapeutic use , Insulin, Long-Acting/therapeutic use , Peptides/therapeutic use , Venoms/therapeutic use , Weight Loss/drug effects , Body Mass Index , Body Weight/drug effects , Cohort Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Exenatide , Female , Glycated Hemoglobin/metabolism , Humans , Insulin Glargine , Lipids/blood , Male , Middle Aged , Retrospective Studies , Treatment Outcome
5.
Eur Psychiatry ; 26(5): 313-9, 2011.
Article in English | MEDLINE | ID: mdl-20621454

ABSTRACT

OBJECTIVE: Adding another antipsychotic to a treatment regimen was previously used in evaluating the medication's efficacy. Supplementation of depot antipsychotics with oral antipsychotics is particularly meaningful because depot formulations are typically chosen for patients struggling with adherence to oral antipsychotics. This post-hoc analysis assessed supplementation of olanzapine long-acting injection (olanzapine-LAI) with oral olanzapine. SUBJECTS AND METHODS: We used 12 months of data from an open-label, single-arm extension study of patients with schizophrenia or schizoaffective disorder (N=931) treated with olanzapine-LAI. The prevalence, duration, time to first supplementation, and best predictors of oral supplementation were assessed. RESULTS: Oral supplementation occurred in 21% of patients for a median of 31 days with mean modal dose of 10.8 mg/day. Mean time to first supplementation was shorter for patients who were at least moderately ill at baseline compared to less ill patients (47 vs. 97 days, p<0.001). Best predictors of oral supplementation included a more severe illness profile at baseline, lower olanzapine-LAI dose prior to oral supplementation, supervised living arrangements, and being African-American. CONCLUSION: Supplementation of olanzapine-LAI appears to be infrequent, of relatively short duration, and reserved for more severely ill patients who may require a targeted rescue medication due to signs of impending relapse.


Subject(s)
Antipsychotic Agents/administration & dosage , Benzodiazepines/administration & dosage , Schizophrenia/drug therapy , Administration, Oral , Adult , Antipsychotic Agents/therapeutic use , Benzodiazepines/therapeutic use , Drug Administration Schedule , Female , Humans , Injections , Male , Middle Aged , Olanzapine , Severity of Illness Index
6.
J Child Adolesc Psychopharmacol ; 17(1): 51-62, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17343553

ABSTRACT

OBJECTIVE: The purpose of this research was to provide an initial examination of the effects of atomoxetine and stimulants on emotional expression using a newly developed scale for assessing emotional expression in children with attention-deficit/hyperactivity disorder (ADHD). METHOD: The parent-rated Expression and Emotion Scale for Children (EESC) was collected during two studies. During a cross-sectional validation study, the EESC was completed to assess the child's current treatment and retrospectively for previous medication. In a randomized, placebo-controlled trial of atomoxetine, the EESC was collected at baseline and endpoint. RESULTS: In the validation study, no statistically significant differences in EESC scores were found between groups taking atomoxetine (n = 74) and stimulants (n = 105). Patients who switched from a stimulant to atomoxetine (n = 40) had greater improvement in emotional expression than those switched to another stimulant (n = 21) (p = 0.008). In the clinical trial, no difference in rates of worsening of emotional expression were observed (atomoxetine 8.8%, placebo 12.3%; p = 0.440). CONCLUSION: No treatment differences in emotional expression were observed based on current medications. However, stimulant patients needing to switch medications may have greater improvements in emotional expression by switching to atomoxetine.


Subject(s)
Adrenergic Uptake Inhibitors/therapeutic use , Attention Deficit Disorder with Hyperactivity/drug therapy , Emotions/drug effects , Personality Assessment , Propylamines/therapeutic use , Adrenergic Uptake Inhibitors/adverse effects , Atomoxetine Hydrochloride , Attention Deficit Disorder with Hyperactivity/psychology , Central Nervous System Stimulants/adverse effects , Central Nervous System Stimulants/therapeutic use , Child , Cross-Sectional Studies , Double-Blind Method , Drug Administration Schedule , Female , Humans , Male , Propylamines/adverse effects
7.
Int J Methods Psychiatr Res ; 14(4): 186-201, 2005.
Article in English | MEDLINE | ID: mdl-16395872

ABSTRACT

The development of rating scales for attention-deficit/hyperactivity disorder (ADHD) has traditionally focused on parent-or teacher-rated scales. However, clinician-based instruments are valuable tools for assessing ADHD symptom severity. The ADHD Rating Scale IV (ADHD RS), clinician administered and scored, has been validated as a useful instrument to assess ADHD symptoms among American children and adolescents. In this study, we assessed the psychometric properties of the scale in a recent clinical trial conducted mainly in Europe with over 600 children and adolescents diagnosed with ADHD. The trial was conducted in 11 European countries plus Australia, Israel, and South Africa. Results based on data in the study indicate that this version of the scale has acceptable psychometric properties including inter-rater reliability, test-retest reliability, internal consistency, factor structure, convergent and divergent validity, discriminant validity, and responsiveness. There were low-to-moderate ceiling and floor effects. The psychometric properties were comparable with other validated scales for assessing ADHD symptom severity. These results were consistent across the 14 countries participating in this trial. Overall, the data from this study support the use of the ADHD RS as a clinician-rated instrument for assessing the severity of ADHD symptoms in children and adolescents in Europe.


Subject(s)
Attention Deficit Disorder with Hyperactivity/diagnosis , Attention Deficit Disorder with Hyperactivity/psychology , International Cooperation , Psychometrics/methods , Adolescent , Child , Factor Analysis, Statistical , Female , Humans , Male , Psychiatric Status Rating Scales , Reproducibility of Results , Sensitivity and Specificity
8.
Drug Alcohol Depend ; 67(2): 149-56, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12095664

ABSTRACT

This study compared the subjective, physiological, and psychomotor effects of atomoxetine and methylphenidate with placebo in healthy volunteers. Sixteen non-dependent light drug users participated in six experimental sessions, receiving placebo, atomoxetine (20, 45 and 90 mg) and methylphenidate (20 and 40 mg) using a double-blind, Latin square design. Subjective drug effects were assessed using Visual Analog Scales (VAS), the Addiction Research Center Inventory (ARCI) and Adjective Rating Scales (ARS). Psychomotor performance was evaluated using the Digit Symbol Substitution Test (DSST). Physiological measures were also collected throughout the sessions. Assessments were conducted before drug administration and 30, 60, 90, 120, 150, 180 and 240 min following dosing. Forty milligrams methylphenidate produced significant increases on the stimulant portions of the VAS and ARS and the benzedrine, amphetamine, morphine-benzedrine and lysergic acid diethylamine (LSD) subscales of the ARCI relative to placebo. Ninety mg atomoxetine was reported to be unpleasurable relative to placebo as indicated by significant increases on the 'bad' and 'sick' portions of the VAS, and on the LSD subscale of the ARCI. Compared with placebo, both methylphenidate doses significantly increased systolic blood pressure (BP) and heart rate (HR). For atomoxetine, 90 mg increased diastolic BP, 45 and 90 mg increased systolic BP, and all three doses increased HR relative to placebo. Neither compound produced significant differences from placebo on DSST performance. These results suggest that atomoxetine does not induce subjective effects similar to methylphenidate and suggest that it is unlikely that atomoxetine will have abuse liability.


Subject(s)
Methylphenidate/pharmacology , Propylamines/pharmacology , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Substance-Related Disorders , Adolescent , Adult , Analysis of Variance , Atomoxetine Hydrochloride , Blood Pressure/drug effects , Blood Pressure/physiology , Double-Blind Method , Female , Humans , Least-Squares Analysis , Male , Outcome Assessment, Health Care , Substance-Related Disorders/psychology
9.
Pediatrics ; 108(5): E83, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11694667

ABSTRACT

OBJECTIVE: Atomoxetine is an investigational, nonstimulant pharmacotherapy being studied as potential treatment for attention-deficit/hyperactivity disorder (ADHD). It is thought to act via blockade of the presynaptic norepinephrine transporter in the brain. We assessed the efficacy of 3 doses of atomoxetine compared with placebo in children and adolescents with ADHD. METHODS: A total of 297 children and adolescents who were 8 to 18 years of age and had ADHD as defined by the Diagnostic and Statistical Manual of Mental Disorders, 4th edition, were randomized to placebo or atomoxetine dosed on a weight-adjusted basis at 0.5 mg/kg/day, 1.2 mg/kg/day, or 1.8 mg/kg/day for an 8-week period. ADHD symptoms, affective symptoms, and social and family functioning were assessed using parent and investigator rating scales. RESULTS: Approximately 71% of children enrolled were male, approximately 67% met criteria for mixed subtype (both inattentive and hyperactive/impulsive symptoms), and the only common psychiatric comorbidity was oppositional defiant disorder (approximately 38% of the sample). At baseline, symptom severity was rated as moderate to severe for most children. At endpoint, atomoxetine 1.2 mg/kg/day and 1.8 mg/kg/day were consistently associated with superior outcomes in ADHD symptoms compared with placebo and were not different from each other. The dose of 0.5 mg/kg/day was associated with intermediate efficacy between placebo and the 2 higher doses, suggesting a graded dose-response. Social and family functioning also were improved in the atomoxetine groups compared with placebo with statistically significant improvements in measures of children's ability to meet psychosocial role expectations and parental impact. Discontinuations as a result of adverse events were <5% for all groups. CONCLUSION: Among children and adolescents aged 8 to 18, atomoxetine was superior to placebo in reducing ADHD symptoms and in improving social and family functioning symptoms. Atomoxetine was associated with a graded dose-response, and 1.2 mg/kg/day seems to be as effective as 1.8 mg/kg/day and is likely to be the appropriate initial target dose for most patients. Treatment with atomoxetine was safe and well tolerated.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Drugs, Investigational/therapeutic use , Propylamines/therapeutic use , Adolescent , Age Factors , Algorithms , Analysis of Variance , Atomoxetine Hydrochloride , Attention Deficit Disorder with Hyperactivity/complications , Child , Depression/diagnosis , Depression/etiology , Dose-Response Relationship, Drug , Female , Humans , Male , Propylamines/administration & dosage
10.
J Child Adolesc Psychopharmacol ; 11(3): 251-65, 2001.
Article in English | MEDLINE | ID: mdl-11642475

ABSTRACT

OBJECTIVE: The goal of this study was to evaluate the tolerability and effectiveness of the experimental, noradrenergic specific reuptake inhibitor atomoxetine in the treatment of children with attention deficit hyperactivity disorder (ADHD). METHODS: This was an open, prospective, dose-ranging study of atomoxetine monotherapy in the treatment of 30 children with ADHD between the ages of 7 and 14 years. Atomoxetine was started at 10-20 mg/day and titrated weekly up to 90 mg over 11 weeks, depending on response and adverse effects. Twenty-two children completed the full 11 weeks. We assessed efficacy with weekly clinician and parent ratings of ADHD and oppositional symptoms and monitored adverse effects, laboratory findings, and cardiovascular parameters. RESULTS: Treatment with atomoxetine (mean final, total daily dose of 1.9 mg/kg/day) was very well tolerated without meaningful adverse effects. Atomoxetine significantly reduced core symptoms of ADHD (ADHD-Rating Scale-IV; 38.6% decrease vs. baseline, p < 0.001) with significant improvement (p < 0.05) in all but 1 of the 18 individual items in the ADHD-Rating Scale-IV. More than 75% of subjects who completed 10 weeks of treatment showed > 25% decrease in ADHD symptoms. CONCLUSIONS: These findings extend to children the positive results previously reported in adults diagnosed with ADHD who were treated with atomoxetine. These results support additional controlled trials of atomoxetine in cases of pediatric ADHD.


Subject(s)
Antidepressive Agents/administration & dosage , Attention Deficit Disorder with Hyperactivity/drug therapy , Propylamines/administration & dosage , Adolescent , Antidepressive Agents/blood , Antidepressive Agents/therapeutic use , Atomoxetine Hydrochloride , Attention Deficit Disorder with Hyperactivity/blood , Attention Deficit Disorder with Hyperactivity/physiopathology , Child , Dose-Response Relationship, Drug , Drug Administration Schedule , Electrocardiography , Female , Hemodynamics/drug effects , Hemodynamics/physiology , Humans , Male , Patient Compliance , Propylamines/blood , Propylamines/therapeutic use , Prospective Studies , Psychological Tests , Severity of Illness Index
11.
J Clin Psychopharmacol ; 21(6): 561-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11763002

ABSTRACT

The 1-week single-blind placebo lead-in has long been a standard in double-blind psychopharmacology clinical trials. Although a lead-in period is often necessary (e.g., to receive laboratory results before randomization), some authors have demonstrated that the standard single-blind placebo lead-in's performance was similar to having a lead-in in which placebo was not administered. The single-blind placebo lead-in did not decrease postrandomization placebo response, nor did it increase drug-placebo differences. To eliminate a higher percentage of placebo responders before randomization and to reduce potential biases in baseline ratings, the authors designed and implemented two depression studies with a double-blind variable placebo lead-in period. In these designs, both the patients and personnel at the investigative sites were blinded to the length of the placebo lead-in period and the start of the active treatment period. Approximately 28% of the patients in the double-blind placebo lead-in studies met criteria to be placebo lead-in responders, as compared with fewer than 10% from two single-blind placebo lead-in studies conducted in a similar time frame. Although all patients continued in the study (including placebo lead-in responders), the primary efficacy analysis prospectively excluded double-blind placebo lead-in responders. Analysis of postrandomization changes revealed that double-blind placebo lead-in responders, even when continuing to receive placebo treatment, maintained their response. At the study endpoint, these placebo lead-in responders had significantly lower severity scores than their counterparts who were not lead-in responders. The prospective removal of lead-in responders thus resulted in an increase in mean endpoint placebo group severity scores. This resulted in an increased drug-placebo treatment difference in one of the two studies but had no effect on the treatment difference in the other study.


Subject(s)
Antidepressive Agents/therapeutic use , Clinical Trials as Topic/methods , Clinical Trials as Topic/statistics & numerical data , Depressive Disorder, Major/drug therapy , Double-Blind Method , Humans , Placebos , Randomized Controlled Trials as Topic/methods , Randomized Controlled Trials as Topic/statistics & numerical data , Single-Blind Method
12.
J Biopharm Stat ; 10(3): 319-33, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10959914

ABSTRACT

At early stages of drug development, the maximum tolerated dose (MTD) must be determined in order to have a range of doses that can be safely administered in humans. This is necessary before the compound can be tested in phase II clinical trials to find the optimal dose in terms of clinical outcome. Although heavily criticized, traditional dose escalation methods are still widely used to estimate the MTD. The recently developed Continual Reassessment Method (CRM) and Logistic Dose Ranging Strategy (LDRS) offer advantages compared to the traditional approach, including improved estimation of the MTD. However, the CRM utilizes a prior distribution that must be specified before having any data from the current trial. The LDRS requires the use of hypothesized data through a seed data set established before the start of the trial. When a wide dose range needs to be tested, the assumed information (prior distribution or seed data set) can have a great impact on dosages used during the trial and on the final estimates of the MTD. This paper combines the LDRS and the traditional dose escalation procedure to suggest a practical two-stage method that provides reliable estimates through relatively easy computation, and yet requires almost no prior information.


Subject(s)
Clinical Trials, Phase I as Topic/methods , Dose-Response Relationship, Drug , Logistic Models , Maximum Tolerated Dose , Bayes Theorem , Clinical Trials, Phase I as Topic/statistics & numerical data , Computer Simulation
13.
Stat Med ; 19(16): 2169-84, 2000 Aug 30.
Article in English | MEDLINE | ID: mdl-10931518

ABSTRACT

Currently available antidepressants have a delayed onset of action and there is considerable interest in developing new products which exhibit a shorter time to response. Previous authors have suggested using a cure model to characterize the time to response for the combination of the patients who respond and the patients who do not respond. In this paper, we use the cure model and propose a Cramer-von Mises statistic to compare the time to response distributions for patients who respond to therapy. The asymptotic null distribution of the statistic is derived. A bootstap procedure is also proposed for sample sizes typical in antidepressant clinical trials. Results of the simulation study suggest that for common designs and sample sizes used in such trials, the statistic has reasonable size and power properties as long as censoring is moderate.


Subject(s)
Adrenergic beta-Antagonists/administration & dosage , Antidepressive Agents, Second-Generation/administration & dosage , Antidepressive Agents/administration & dosage , Clinical Trials as Topic/statistics & numerical data , Depressive Disorder/drug therapy , Fluoxetine/administration & dosage , Models, Statistical , Pindolol/administration & dosage , Algorithms , Humans , Sample Size , Statistical Distributions , Survival Analysis , Time Factors , Treatment Outcome
14.
J Psychiatr Res ; 34(1): 3-10, 2000.
Article in English | MEDLINE | ID: mdl-10696827

ABSTRACT

In clinical studies of antidepressants, the Hamilton Depression Rating Scale (HAMD) total score has been the gold standard instrument for establishing and comparing the efficacy of new treatments. However, the HAMD is a multidimensional measure, which may reduce its ability to detect differences between treatments, in particular, changes in core symptoms of depression. Two meta-analyses were conducted to compare the responsiveness of the HAMD total score with several published unidimensional subscale scores based upon core symptoms of depression. The first compared the above instrument's ability to detect differences between fluoxetine and placebo across eight studies involving over 1600 patients. The second analysis involved four studies and over 1200 patients randomized to tricyclic antidepressants and placebo. In both meta-analyses, the unidimensional core subscales outperformed the HAMD total score at detecting treatment differences. The implications of this on sample sizes and power for clinical studies will be discussed. In fact, studies based on the observed effect sizes from the core subscales would require approximately one-third less patients than studies based on the HAMD total score. Effect sizes from each individual HAMD item will also be presented to help explain the differences in responsiveness between the scales.


Subject(s)
Depression/diagnosis , Psychological Tests , Antidepressive Agents/therapeutic use , Clomipramine/therapeutic use , Depression/drug therapy , Double-Blind Method , Fluoxetine/therapeutic use , Humans , Imipramine/therapeutic use , Sensitivity and Specificity , Severity of Illness Index
15.
Psychopharmacol Bull ; 34(1): 19-24, 1998.
Article in English | MEDLINE | ID: mdl-9564194

ABSTRACT

The implementation of a multisite, randomized, clinical psychopharmacologic trial involves a substantial investment of time and effort on the part of all participants. Because of their complexity, such clinical trials present unique methodological and design challenges. Indeed, it is not uncommon for such studies to conclude with uninterpretable results, due in part to such methodological pitfalls. It has been suggested that clarification of such methodologic dilemmas is one of the most important challenges facing the future of industry-sponsored psychopharmacologic drug development. Among the many factors that may contribute to problematic clinical trial results, error in measuring the phenomena being studied is of particular concern. In this article, we describe the outcome of an intensive series of interrater reliability training sessions for the 17-item Hamilton Depression Rating Scale conducted at the start of a Phase II multisite clinical drug trial. The data underscore the magnitude of error present in such a test setting and provide preliminary evidence for the potential effect of this problem on the detection of clinical change.


Subject(s)
Depressive Disorder/drug therapy , Multicenter Studies as Topic , Psychopharmacology , Research Design , Adult , Aged , Double-Blind Method , Female , Humans , Male , Middle Aged , Observer Variation , Psychiatric Status Rating Scales
16.
Lancet ; 349(9065): 1594-7, 1997 May 31.
Article in English | MEDLINE | ID: mdl-9174562

ABSTRACT

BACKGROUND: Major depression affects more than 5% of the population and is a serious health and economic problem. Antidepressants have a slow onset of action and are effective in less than two-thirds of patients. The biochemical effects of selective serotonin reuptake inhibitors may be limited by the negative feedback from serotonin autoreceptors. Pindolol is an antagonist of both serotonin autoreceptors and beta-adrenoceptors. We studied the effect of the addition of pindolol to fluoxetine antidepressant treatment. METHODS: Of 132 eligible patients with major depression, 111 were randomly assigned to treatment with fluoxetine (20 mg daily) and either placebo or pindolol (7.5 mg daily). Patients were assessed twice a week for the first 3 weeks of active treatment and then once a week until the end of the study (42 days). Hamilton and Montgomery-Asberg depression-rating scales were used to assess depression severity. FINDINGS: The proportion of patients who responded to treatment with fluoxetine and pindolol was greater than that with fluoxetine and placebo (41/55 [75%] vs 33/56 [59%], [90% CI 1.1-30.1], p = 0.04). The proportion of patients who achieved a sustained response was also greater in the fluoxetine and pindolol group than in the fluoxetine and placebo group (38/55 [69%] vs 27/56 [48%] [5.9-35.9], p = 0.03). The number of days to reach a sustained response was lower in the fluoxetine and pindolol group than in the fluoxetine and placebo group (median 19 vs 29 days, p = 0.01), however, there was no difference in the time-to-onset of clinical improvement when stringent conditions were used (15 vs 18 days, p = 0.20). INTERPRETATION: The addition of pindolol to fluoxetine antidepressant treatment increases the effectiveness of fluoxetine therapy. Further work is needed to resolve whether the time to clinical improvement benefits from this combination and whether the increase in efficacy occurs with other antidepressants.


Subject(s)
Depression/drug therapy , Fluoxetine/therapeutic use , Pindolol/therapeutic use , Selective Serotonin Reuptake Inhibitors/therapeutic use , Adult , Antidepressive Agents, Second-Generation/administration & dosage , Antidepressive Agents, Second-Generation/therapeutic use , Double-Blind Method , Drug Therapy, Combination , Female , Fluoxetine/administration & dosage , Humans , Male , Middle Aged , Pindolol/administration & dosage , Psychiatric Status Rating Scales , Selective Serotonin Reuptake Inhibitors/administration & dosage , Time Factors , Treatment Outcome
17.
Psychopharmacol Bull ; 31(2): 227-37, 1995.
Article in English | MEDLINE | ID: mdl-7491373

ABSTRACT

Fluoxetine is an efficacious, nonsedative antidepressant, but its selective efficacy on symptoms of insomnia has not been thoroughly explored. In this analysis, the effects of fluoxetine versus placebo on symptoms of insomnia were evaluated in three clinical subgroups: patients with baseline sleep disturbance, melancholia, or reduced rapid eye movement (REM) latency. Eighty-nine outpatients with major depression completed 2 nights of polysomnography (PSG) and were randomized to fluoxetine or placebo. Within each subgroup of patients, fluoxetine was statistically significantly more effective than placebo in improving non-sleep Hamilton Rating Scale for Depression (HAM-D) items (HAM-D-17 total minus scores from Items 4, 5, and 6). Numerical improvement in HAM-D sleep total (sum of HAM-D Items 4, 5, and 6) was also seen for fluoxetine versus placebo. Fluoxetine did not exacerbate sleep disturbance either at Week 1 or at endpoint. These findings suggest that fluoxetine is an effective antidepressant in patients with baseline sleep disturbance, melancholia, and reduced REM latency.


Subject(s)
Fluoxetine/therapeutic use , Sleep Initiation and Maintenance Disorders/drug therapy , Adolescent , Adult , Depression/drug therapy , Depressive Disorder/drug therapy , Female , Humans , Male , Middle Aged , Placebo Effect , Psychiatric Status Rating Scales , Sleep Wake Disorders/drug therapy , Sleep, REM/drug effects , Treatment Outcome
18.
J Biopharm Stat ; 4(2): 147-64, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7951271

ABSTRACT

The continual reassessment method (1) (CRM) for phase I cancer trials provides improved estimation of the maximum tolerated dose (MTD), and fewer patients receive ineffective dose levels compared to the traditionally used design. However, the CRM has not gained acceptance in practice owing to concerns with administering dose levels that are too toxic. In this article, several conservative modifications of the CRM are introduced. The result is a procedure that improves estimation of the MTD and decreases the use of ineffective doses, without significantly increasing the use of toxic dose levels. The CRM with modification outperforms the traditional method in a simulation study.


Subject(s)
Antineoplastic Agents/therapeutic use , Clinical Trials, Phase I as Topic/methods , Neoplasms/drug therapy , Research Design/statistics & numerical data , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Humans , Software
19.
Psychiatry Res ; 52(3): 327-39, 1994 Jun.
Article in English | MEDLINE | ID: mdl-7991725

ABSTRACT

Fluoxetine and placebo were compared in 89 outpatients with major depression with (n = 45) or without (n = 44) a reduced or shortened rapid eye movement latency (SREML) (< or = 65 minutes) to determine whether rapid eye movement latency (REML) predicted placebo and/or antidepressant response. Men and women were stratified based on polysomnographic recordings and then randomly assigned to receive double-blind fluoxetine (20 mg/day) or placebo for 8 weeks after a 2-week, single-blind, placebo lead-in period. Fluoxetine-treated patients demonstrated a significantly greater reduction in the Hamilton Rating Scale for Depression total score and a significantly greater response rate than placebo-treated patients in both the SREML and the combined strata. Treatment differences in the non-SREML stratum were not statistically significant. Results supported REML as a predictor of placebo nonresponse but did not predict a differential fluoxetine response in patients with SREML compared with patients without SREML.


Subject(s)
Depressive Disorder/drug therapy , Fluoxetine/therapeutic use , Reaction Time/drug effects , Sleep, REM/drug effects , Adolescent , Adult , Ambulatory Care , Depressive Disorder/psychology , Double-Blind Method , Female , Fluoxetine/adverse effects , Humans , Male , Middle Aged , Personality Inventory , Polysomnography , Treatment Outcome
20.
J Affect Disord ; 30(3): 163-73, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8006243

ABSTRACT

Fluoxetine (20 mg/day) and placebo were compared in 89 outpatient men and women with major depression with (n = 52) or without (n = 37) DSM-III-R melancholia in an 8-week double-blind study to determine predictors of treatment response. Fluoxetine was statistically superior to placebo both within the melancholic subtype and in the total patient group (all measures). Response rate and mean decrease in 17-item Hamilton Depression Rating Scale total score approached statistical significance in favor of fluoxetine-treated melancholic patients compared with fluoxetine-treated non-melancholic patients. There were no statistically significant differences between fluoxetine-treated and placebo-treated non-melancholic patients. Results support DSM-III-R melancholia as a predictor of antidepressant response.


Subject(s)
Depressive Disorder/drug therapy , Fluoxetine/therapeutic use , Adolescent , Adult , Aged , Depressive Disorder/psychology , Double-Blind Method , Female , Fluoxetine/adverse effects , Humans , Male , Middle Aged , Psychiatric Status Rating Scales , Reaction Time/drug effects , Sleep, REM/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...