Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Antimicrob Agents Chemother ; 41(8): 1783-7, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9257761

ABSTRACT

The pharmacokinetic characteristics of cefepime were determined after first dose (n = 35) and again under steady-state conditions (n = 31) with a group of 37 infants and children. In eight subjects, a cefepime dose given by intramuscular injection was substituted for an intravenous dose, and disposition characteristics were studied again. Study subjects ranged in age from 2.1 months to 16.4 years, and all had normal renal function. Each patient received 50 mg of cefepime/kg of body weight intravenously every 8 h, up to a total maximum individual dose of 2 g. With the exception of one study patient who received a single cefepime dose for surgical prophylaxis, the patients received cefepime for 2 to 13 days. Elimination half-life (t1/2), steady-state volume of distribution, total body clearance, and renal clearance after first dose administration averaged 1.7 h, 0.35 liter/kg, and 3.1 and 1.9 ml/min/kg, respectively. Although cefepime t1/2 and mean residence time (MRT) were slightly longer for subjects <6 months of age than for older subjects, no differences in cefepime disposition characteristics between first dose and steady-state evaluations were observed. t1/2 (1.8 versus 1.9 h) and MRT (2.3 versus 3.2 h) were slightly prolonged after intramuscular administration, reflecting the influence of absorption from the intramuscular injection site on cefepime elimination. Bioavailability after intramuscular administration averaged 82% (range, 61 to 124%). Fifty-seven percent of the first dose and 88.9% of the last dose were recovered as unchanged drug in urine over the 8- and 24-h sampling periods, respectively. These pharmacokinetic data support a single cefepime dosing strategy for patients > or =2 months of age. The integration of the cefepime pharmacokinetic data generated in our study with the MICs for important pathogens responsible for infections in infants and children supports the administration of a dose of 50 mg of cefepime/kg every 12 h for patients > or =2 months of age to treat infections caused by pathogens for which cefepime MICs are < or =8 mg/liter.


Subject(s)
Cephalosporins/pharmacokinetics , Adolescent , Age Factors , Cefepime , Cephalosporins/administration & dosage , Cephalosporins/blood , Child , Child, Preschool , Female , Humans , Infant , Injections, Intramuscular , Injections, Intravenous , Male
2.
J Antimicrob Chemother ; 39(6): 789-96, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9222049

ABSTRACT

The pharmacokinetics of teicoplanin were assessed after a single dose and under multidose conditions in 12 infants and children. Study patients ranged in age from 2.4 to 11 years. Each patient received teicoplanin 6 mg/kg body weight given intravenously over 20-30 min, once daily for five consecutive days. Multiple timed blood and urine samples were obtained over the 6 day sampling period and were analysed for teicoplanin by both microbiological assay and HPLC. Three-compartment pharmacokinetic analysis was used to describe the drug's disposition characteristics. Peak and 24 h trough serum teicoplanin concentrations averaged 39.3 and 1.8 mg/L after the first dose with little accumulation observed after 5 days of therapy. Teicoplanin disposition was variable; V(d)ss ranged from 0.31 to 0.68 L/kg, t(1/2)gamma from 6.5 to 18.1 h and CI from 29 to 51 mL/h/kg. A substantial amount of the administered drug distributed rapidly to the largest, third compartment, with egress approximately four-fold slower than ingress. The majority of the drug was excreted unchanged in the urine. Teicoplanin administration was well tolerated by all study subjects. Using the teicoplanin pharmacokinetic data derived in our study, a dose of teicoplanin 8 mg/kg body weight administered every 12 h should achieve target serum trough concentrations averaging 11 mg/L in children. Higher doses, e.g. 15 mg teicoplanin/kg administered every 12 h, may be needed for the treatment of deep-seated staphylococcal infections and/or endocarditis.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Teicoplanin/pharmacokinetics , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/urine , Child , Child, Preschool , Female , Humans , Injections, Intravenous , Male , Teicoplanin/blood , Teicoplanin/urine
3.
Clin Pharmacol Ther ; 60(4): 414-23, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8873689

ABSTRACT

OBJECTIVES: Account for the interindividual variability in the pharmacokinetics and pharmacodynamics of bumetanide after intravenous administration of single doses to critically ill infants. METHODS: This prospective open-label study was carried out in the pediatric intensive care unit of a university-based children's hospital. Fifty-three volume-overloaded critically ill infants (age range, 4 days to 6 months) were divided into two groups: those with heart disease (31 infants) and those with lung disease (22 infants). Each patient received a single intravenous bolus dose of bumetanide. Doses, selected in sequential order, ranged from 0.005 to 0.100 mg/kg. Age was used as a continuous variable to determine its effects on the variability in the pharmacokinetics and pharmacodynamics of bumetanide. Hierarchical multiple regression analyses were used to assess the effects of age, disease, and other drugs on the variability in the effects of bumetanide. RESULTS: Total clearance, renal clearance, and nonrenal clearance of bumetanide all increased with age (p < 0.05), but the ratio of renal clearance to total clearance remained constant at about 0.4. Half-life and mean residence time decreased markedly in the first month of life (p < 0.05). Bumetanide excretion rate normalized for dose also increased with increasing age. Patients with lung disease exhibited a significantly greater clearance and shorter half-life (p < 0.05) than those with heart disease, whereas volume of distribution was similar in both groups. The primary determinant of bumetanide excretion rate was the administered dose (73%). Dose-response curves for urine flow rate and electrolyte excretion were similar between disease groups. The time course of the effect of bumetanide excretion rate on pharmacodynamics responses was similar between disease groups, as was the duration of the diuretic effect. CONCLUSIONS: The pharmacokinetics of bumetanide were influenced significantly by age and disease. Differences in pharmacokinetics between patients with lung and heart disease were primarily due to differences in total clearance. The administered dose of bumetanide and age were positive determinants of bumetanide excretion rate and pharmacodynamic responses. Pharmacodynamic responses as a function of bumetanide excretion rate were not significantly different between disease groups.


Subject(s)
Bumetanide/pharmacology , Bumetanide/pharmacokinetics , Diuretics/pharmacology , Diuretics/pharmacokinetics , Heart Diseases/metabolism , Infant, Newborn, Diseases/metabolism , Lung Diseases/metabolism , Aging/metabolism , Critical Illness , Female , Humans , Infant , Infant, Newborn , Injections, Intravenous , Male , Prospective Studies , Regression Analysis
4.
Clin Pharmacol Ther ; 60(4): 405-13, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8873688

ABSTRACT

OBJECTIVE: Define the pharmacokinetics of bumetanide after single intravenous doses in volume-overloaded critically ill infants. METHODS: A prospective, open-label study was carried out in a group of 58 infants aged 0 to 6 months who required diuretic therapy. Each patient received a single dose of intravenous bumetanide. Doses selected in sequential order ranged from 0.005 to 0.10 mg/kg. Hematologic and serum chemistry studies were performed before and at 6 and 24 hours after bumetanide administration. Determinations of urine volume and chemistries were performed before (collected from -2 to -4 hours to time 0) and at 1, 2, 3, 4, 6, and 12 hours after bumetanide dosing. Serum samples collected at time 0 and at 5, 15, 30, 60, 120, 180, 240, 360, and 480 minutes and urine collected at time 0 and at 0 to 1, 1 to 2, 2 to 3, 3 to 4, 4 to 6, and 6 to 12 hours were analyzed for bumetanide concentration. Data were evaluated by standard noncompartmental pharmacokinetic techniques. RESULTS: Peak serum bumetanide concentrations occurred at 5 minutes after bumetanide administration. Area under the curve and peak serum bumetanide concentrations showed linear increases over the twentyfold dose range; whereas beta volume of distribution, volume of distribution at steady state, clearance, renal clearance, half-life, and mean residence time values were independent of dose. Peak urinary excretion rates of bumetanide increased linearly with increasing doses. The mean percent of bumetanide recovered in the urine from 0 to 12 hours was 40% +/- 15% of the administered dose. CONCLUSIONS: Distribution and elimination kinetics of bumetanide were similar in all patients. Elimination kinetics were first order over the dose range of 0.005 to 0.10 mg/kg. Pharmacokinetic parameter estimates (beta volume of distribution, volume of distribution at steady state, clearance, renal clearance, half-life, and mean residence time) were independent of the dose of bumetanide administered. Single doses of bumetanide up to 0.10 mg/kg appear to be well tolerated in acutely ill volume-overloaded infants aged 0 to 6 months.


Subject(s)
Bumetanide/pharmacokinetics , Diuretics/pharmacokinetics , Infant, Newborn, Diseases/physiopathology , Area Under Curve , Bumetanide/blood , Bumetanide/urine , Chromatography, High Pressure Liquid , Critical Illness , Diuretics/blood , Diuretics/urine , Female , Humans , Infant , Infant, Newborn , Injections, Intravenous , Male , Prospective Studies
5.
Clin Pharmacol Ther ; 60(4): 424-34, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8873690

ABSTRACT

OBJECTIVES: Determine the diuretic effects of single intravenous doses of bumetanide in volume-overloaded critically ill infants. METHODS: A prospective, open-label study was carried out in 56 infants aged 0 to 6 months who required diuretic therapy. Each patient received a single intravenous dose of bumetanide. Doses selected in sequential order ranged from 0.005 to 0.10 mg/kg. Determinations of urine volume, electrolytes, creatinine levels, and osmolality were performed before (collected from -2 to -4 hours to time 0) and at 1, 2, 3, 4, 6, and 12 hours after bumetanide dosing. Serum samples collected at time 0 and at 5, 15, 30, 60, 120, 180, 240, 360, and 480 minutes and urine aliquots collected at time 0, 0 to 1, 1 to 2, 2 to 3, 3 to 4, 4 to 6, and 6 to 12 hours were analyzed for bumetanide concentration. Individual changes in urine flow rate and electrolyte excretion were plotted against corresponding bumetanide excretion rates, taken as the effective dose of the drug. RESULTS: Peak bumetanide excretion rates increased linearly with increasing doses of drug. Time course patterns for urine flow rate and electrolyte excretion were similar for all dosage groups. Urine flow rate and electrolyte excretion increased linearly up to a bumetanide excretion rate of approximately 7 micrograms/kg/hr and either plateaued (urine flow rate) or declined at a bumetanide excretion rate of > 10 micrograms/kg/hr. Diuretic efficiency of bumetanide was maximal at doses of 0.005 to 0.010 mg/kg but decreased at higher doses. CONCLUSIONS: Maximal diuretic responses occurred at a bumetanide excretion rate of about 7 micrograms/kg/hr, corresponding to doses of 0.035 to 0.040 mg/kg. Higher doses produced a proportionately higher bumetanide excretion rate but no increased diuretic effect. Lower doses of bumetanide had the greatest diuretic efficiency, suggesting that continuous infusion of low doses of bumetanide or intermittent low-dose boluses may produce optimal diuretic responses in critically ill infants.


Subject(s)
Bumetanide/administration & dosage , Diuretics/administration & dosage , Urination/drug effects , Bumetanide/blood , Bumetanide/urine , Critical Illness , Diuretics/blood , Diuretics/urine , Dose-Response Relationship, Drug , Electrolytes/urine , Female , Humans , Infant , Infant, Newborn , Injections, Intravenous , Male , Prospective Studies
6.
Crit Care Med ; 24(9): 1473-81, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8797618

ABSTRACT

OBJECTIVE: To assess the pharmacokinetics and pharmacodynamics of propofol sedation of critically ill, mechanically ventilated infants and children. DESIGN: A prospective clinical study. SETTING: A pediatric intensive care unit (ICU) in a university hospital. PATIENTS: Clinically stable, mechanically ventilated pediatric patients were enrolled into our study after residual sedative effects from previous sedative therapy dissipated and the need for continued sedation therapy was defined. Patients were generally enrolled just before extubation. INTERVENTIONS: A stepwise propofol dose escalation scheme was used to determine the steady-state propofol dose necessary to achieve optimal sedation, as defined by the COMFORT scale, a validated scoring system which reliably and reproducibly quantifies a pediatric patient's level of distress. When in need of continued sedation, study patients received an initial propofol loading dose of 2.5 mg/kg and were immediately started on a continuous propofol infusion of 2.5 mg/kg/hr. The propofol infusion rate was adjusted and repeat loading doses were administered, if needed, using a coordinated dosing scheme to maintain optimal sedation for a 4-hr steady-state period. After 4 hrs of optimal sedation, the propofol infusion was discontinued and simultaneous blood sampling and COMFORT scores were obtained until the patient recovered. Additional blood samples were obtained up to 24 hrs after stopping the infusion and analyzed for propofol concentration by high-performance liquid chromatography. MEASUREMENTS AND MAIN RESULTS: Twenty-nine patients were enrolled into this study. One patient was withdrawn from this study because of an acute decrease in blood pressure occurring with the first propofol loading dose; 28 patients completed the study. All patients were sedated immediately after the first 2.5-mg/kg propofol loading dose. Eight patients were adequately sedated with the starting propofol dose regimen, whereas five patients required downward dose adjustment and 11 patients required dosage increases to achieve optimal sedation. Four patients failed to achieve adequate sedation after five dose escalations and the drug was stopped. Recovery from sedation (COMFORT score of > or = 27) after stopping the propofol infusion was rapid, averaging 15.5 mins in 23 of 24 evaluable patients. In 13 patients who were extubated after stopping the propofol infusion, the time to extubation was also rapid, averaging 44.5 mins. Determination of the blood propofol concentration at the time of recovery from propofol sedation was possible in 15 patients. The blood propofol concentration was variable, ranging between 0.262 to 2.638 mg/L but < or = 1 mg/L in 13 of 15 patients. Similarly, tremendous variation was observed in propofol pharmacokinetics. Propofol disposition was best characterized by a three-compartment model with initial rapid distribution into a small central compartment, V1, and two larger compartments, V2 and V3, which are two-and 20-fold greater in volume, respectively, than V1. Redistribution from V2 and V3 into V1 was much slower than ingress, underscoring the importance of the propofol concentration in V1 as reflective of the drug's sedative effect. Propofol was well tolerated. Two patients experienced an acute decrease in blood pressure which resolved without treatment. CONCLUSIONS: We conclude that a descending propofol dosing strategy, which maintains the propofol concentration constant in the central compartment (V1) while drug accumulates in V2 and V3 to intercompartmental steady-state, is necessary for effective propofol sedation in the pediatric ICU. Our proposed dosing scheme to achieve and maintain the blood propofol concentration of 1 mg/L would appear effective for sedation of most clinically stable, mechanically ventilated pediatric patients.


Subject(s)
Conscious Sedation , Critical Care , Propofol/administration & dosage , Adolescent , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Humans , Infant , Infusions, Intravenous , Male , Propofol/blood , Propofol/pharmacokinetics , Respiration, Artificial
7.
Comput Biomed Res ; 28(4): 319-34, 1995 Aug.
Article in English | MEDLINE | ID: mdl-8549123

ABSTRACT

We report on the construction of neural networks for determining whether pediatric patients requiring transport to a tertiary care center should be moved by air or by ground. The networks were based on the functional-link net architecture. In two experiments, feedforward supervised-learning neural nets were trained with examples of an expert's decisions and then were used in a consulting mode to provide advice on cases not previously encountered. Training and validation were performed by a combination of the k-fold cross-validation and leaving-one-out sampling methods. Use of the functional-link net rather than the customary backpropagation net enabled us to carry out the training with fairly large amounts of data in realistically short time periods. In the first experiment, capillary refill, skin color, and stridor were consistently the input variables that were most strongly associated with the decision output. In both experiments, the networks were validated by comparing their performance retrospectively against the determination of an expert pediatric transport physician. The network was trained based on the expert's opinion about the correct mode of transport for each case with error rates of less than 10(-5).


Subject(s)
Decision Making, Computer-Assisted , Neural Networks, Computer , Transportation of Patients/methods , Artificial Intelligence , Child , Humans
8.
J Clin Pharmacol ; 35(7): 658-65, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7560245

ABSTRACT

The pharmacokinetic characteristics of ticarcillin and clavulanic acid were determined after the first dose (n = 22) and again under steady-state conditions (n = 16) in a group of infants and children. Study subjects ranged in age from 1 month to 9.3 years; all but 3 study patients were 6 months of age or older. Each patient received 50 mg of ticarcillin and 1.7 mg of clavulanic acid (30:1 ratio) per kg of body weight given intravenously every 4 hours. Elimination half-life, steady-state volume of distribution, and body clearance averaged 1.1 hours, 0.22 L/kg, and 2.7 mL/min/kg, respectively, for ticarcillin, and 0.9 hours, 0.4 L/kg, and 6.2 mL/min/kg, respectively, for clavulanic acid. A total of 71% of the ticarcillin and 50% of the clavulanic acid dose were excreted unchanged in the urine over the 4-hour sampling period. Corresponding renal clearances averaged 2.1 and 3.2 mL/min/kg for ticarcillin and clavulanic acid, respectively. No differences were observed between first dose and steady-state evaluations in the pharmacokinetic behavior of either agent. In contrast, the pharmacokinetic behavior of clavulanic acid was significantly different from that observed for ticarcillin. These pharmacokinetic data combined with known in vitro susceptibilities of important clinical pathogens support a dose of 80 mg of ticarcillin and 2.7 mg/kg clavulanic acid per kg body weight given as a fixed dose combination every 8 hours for the treatment of most systemic infections that occur outside the central nervous system.


Subject(s)
Drug Therapy, Combination/pharmacokinetics , Child , Child, Preschool , Clavulanic Acids/administration & dosage , Clavulanic Acids/pharmacokinetics , Drug Administration Schedule , Drug Therapy, Combination/administration & dosage , Half-Life , Humans , Infant , Infant, Newborn , Injections, Intravenous , Metabolic Clearance Rate , Ticarcillin/administration & dosage , Ticarcillin/pharmacokinetics , beta-Lactamase Inhibitors
9.
MD Comput ; 12(3): 200-5, 1995.
Article in English | MEDLINE | ID: mdl-7596250

ABSTRACT

We developed a research database for a five-year prospective investigation of the medical, social, and developmental correlates of chronic lung disease during the first three years of life. We used the Ingres database management system and the Statit statistical software package. The database includes records containing 1300 variables each, the results of 35 psychological tests, each repeated five times (providing longitudinal data on the child, the parents, and behavioral interactions), both raw and calculated variables, and both missing and deferred values. The four-layer menu-driven user interface incorporates automatic activation of complex functions to handle data verification, missing and deferred values, static and dynamic backup, determination of calculated values, display of database status, reports, bulk data extraction, and statistical analysis.


Subject(s)
Data Interpretation, Statistical , Database Management Systems , Medical Records Systems, Computerized , Software , Bronchopulmonary Dysplasia/diagnosis , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Longitudinal Studies , Male , Prospective Studies
10.
Antimicrob Agents Chemother ; 38(12): 2817-26, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7695268

ABSTRACT

The pharmacokinetics of piperacillin and tazobactam were assessed after single-dose administration to 47 infants and children. Study subjects ranging in age from 2 months to 12 years were randomized to receive one of two different doses of a piperacillin-tazobactam combination (8:1): a low dose (n = 23) of 50 and 6.25 mg of piperacillin and tazobactam per kg of body weight, respectively, or a high dose (n = 24) of 100 and 12.5 mg, respectively. The pharmacokinetic behavior of tazobactam was very similar to that observed for piperacillin, supporting the use of these two agents in a fixed-dose combination. No differences in the pharmacokinetics of piperacillin or tazobactam were observed between the two doses administered. The elimination parameters half-life and total body clearance decreased and increased, respectively, with increasing age, whereas volume parameters (volume of distribution and steady-state volume of distribution) remained relatively constant for both compounds. The primary metabolite of tazobactam, metabolite M1, was measurable in the plasma of 18 of the 47 study subjects; 17 of these 18 subjects received the high doses. More than 70% of the administered piperacillin and tazobactam doses were excreted unchanged in the urine over a 6-h collection period. These data combined with the known in vitro susceptibilities of a broad range of pediatric bacterial pathogens indicate that a dose of 100 mg of piperacillin and 12.5 of mg tazobactam per kg of body weight administered as a fixed-dose combination every 6 to 8 h would be appropriate to initiate clinical efficacy studies in infants and children for the treatment of systemic infections arising outside of the central nervous system.


Subject(s)
Penicillanic Acid/analogs & derivatives , Piperacillin/pharmacokinetics , beta-Lactamase Inhibitors , Child , Child, Preschool , Humans , Infant , Metabolic Clearance Rate , Microbial Sensitivity Tests , Penicillanic Acid/pharmacokinetics , Piperacillin/pharmacology , Tazobactam
11.
J Pediatr ; 124(5 Pt 1): 765-71, 1994 May.
Article in English | MEDLINE | ID: mdl-7513757

ABSTRACT

This study sought to determine whether very low birth weight (VLBW) infants (< 1500 gm) with fetal cocaine exposure differed from non-cocaine-exposed VLBW infants in incidence of neonatal medical complications and in later developmental outcome. Forty-one cocaine-exposed, VLBW infants, followed in a longitudinal study, were compared with 41 non-cocaine-exposed, VLBW infants of comparable race, social class, age, and incidence of bronchopulmonary dysplasia. Cocaine-exposed infants were identified on the basis of combined findings of maternal and/or infant urine immunoassay and on the basis of maternal self-report. At birth, groups did not differ on medical risk factors except that cocaine-exposed infants had a higher incidence of mild (grades I to II) intraventricular hemorrhage. Cocaine-using women were also more likely to use other drugs, especially alcohol, marijuana, and tobacco. At follow-up, at mean corrected ages of 16.5 +/- 8 months for 30 cocaine-exposed infants and 18.5 +/- 7 months for 37 non-cocaine-exposed infants, standardized assessments of cognitive (Mental Development Index) and motor (Psychomotor Development Index) development were administered. Cocaine-exposed infants had lower mean cognitive (83 +/- 27 vs 91 +/- 19), and motor (85 +/- 25 vs 96 +/- 18) scores; the incidence of developmental delay was significantly higher even after control for the effects of intraventricular hemorrhage and chronologic age. Cocaine-exposed VLBW infants were also more likely to be living with relatives or in foster homes. We conclude that these VLBW, cocaine-exposed infants were at increased risk of intraventricular hemorrhage, were more likely to be placed outside maternal care, and had higher incidences of cognitive and motor delays at follow-up.


Subject(s)
Cerebral Hemorrhage/chemically induced , Cocaine/adverse effects , Developmental Disabilities/chemically induced , Infant, Low Birth Weight , Prenatal Exposure Delayed Effects , Adult , Female , Humans , Infant, Newborn , Male , Pregnancy , Pregnancy Complications , Prospective Studies , Regression Analysis , Substance-Related Disorders/complications
12.
Crit Care Med ; 22(1): 163-70, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8124960

ABSTRACT

OBJECTIVE: To derive a target range of optimal sedation for the COMFORT Scale and to prospectively test that target range against intensivist assessment of adequacy of sedation. DESIGN: Serial prospective agreement cohort studies. SETTING: Twelve-bed pediatric intensive care unit in an urban academic teaching hospital. PATIENTS: Eighty-five mechanically ventilated children (aged 0 to 102 months). INTERVENTIONS: Three serial prospective studies comparing simultaneous, independent ratings conducted by trained observers using an objective scale and intensive care physicians using global assessment. The initial study was designed to derive the target range. The second study was conducted to verify that target range in a second population. The third study was added to evaluate relative variability in methods used in the second study. MEASUREMENTS AND MAIN RESULTS: Adequacy of sedation using visual analog scale and descriptive ratings or the COMFORT Scale (a previously validated behaviorally anchored scale to rate eight behavioral or physiologic dimensions of distress). The first study comprised 100 observations. Groups of patients described by the intensivist as inadequately sedated, optimally sedated, and excessively sedated had different mean COMFORT scores (30.5 +/- 0.7 vs. 22.9 +/- 5.8 vs. 14.3 +/- 0.7, respectively, p < .05). The target range of optimal sedation was defined as COMFORT scores of 17 to 26. The second study verified the target range prospectively in a second group of 96 observations. The COMFORT score was strongly associated with the sedation adequacy rating by the intensivist (p < .001; r2 = .662). COMFORT scores accurately predicted the patient assignment to adequacy of sedation categories by the intensivist in 66.1% of observations. Discrepancy between physicians occurred in 38.5% of 16 paired physician ratings in the second study. In the third study, 120 observations comparing paired COMFORT scores to paired physician ratings of the same subjects demonstrated significantly less variability in COMFORT assessment of adequacy of sedation. COMFORT scores were similarly unbiased (1.1% vs. 0.22%) but more precise (8.0% vs. 16.7%) than intensivist ratings (p < .025). CONCLUSION: Adequacy of sedation is measured more consistently by observers using the COMFORT Scale than by intensivist global assessment.


Subject(s)
Critical Care/methods , Respiration, Artificial , Child , Child, Preschool , Conscious Sedation/methods , Female , Humans , Infant , Infant, Newborn , Intensive Care Units, Pediatric , Male , Prospective Studies
13.
J Speech Hear Res ; 36(3): 568-79, 1993 Jun.
Article in English | MEDLINE | ID: mdl-7687313

ABSTRACT

The present investigation addresses two primary hypotheses: (a) that a subset of children with developmental language disorders exhibits significantly more disfluencies than other children with language disorders and (b) that differences between the disfluent and nondisfluent groups observed in fluency may be related to differences in language deficits. Spontaneous language samples from 60 preschool children with developmental language disorders were analyzed for frequency and type of disfluencies. Comparisons of the frequency of disfluencies across subjects revealed that a subset of 10 subjects exhibited significantly more disfluencies than the other subjects with language disorders. Demographic, intelligence, and language variables were compared across the two groups to determine whether such factors could account for the differences in fluency. The subjects with greater percentages of disfluencies were found to be significantly older and demonstrated significantly higher scores on two standard measures of vocabulary. These findings were interpreted in light of two models of disfluencies: the neuropsycholinguistic (Perkins, Kent, & Curlee, 1991) and Demands and Capacities (Adams, 1990; Starkweather, 1987). This suggests that some children with language disorders are at risk for fluency breakdown because of dysynchronies in the development of lexical and syntactic aspects of language or as a result of mismathces between speaking demands and capacities.


Subject(s)
Developmental Disabilities/diagnosis , Language Development , Language Disorders/diagnosis , Age Factors , Child Language , Child, Preschool , Female , Humans , Intelligence , Language Tests , Male , Speech , Stuttering/diagnosis , Verbal Behavior
14.
Pediatrics ; 90(3): 380-4, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1518692

ABSTRACT

Recurrent episodes of hypoxemia may affect the growth, cardiac function, neurologic outcome, and survival of infants with bronchopulmonary dysplasia (BPD). As oral feeding might stress these infants by compromising pulmonary function even after hospital discharge, we measured oxygen saturation (SaO2) via pulse oximetry before, during the initial 10 minutes of, and immediately after oral feeding in 11 patients with BPD, 12 very low birth weight infants, and 23 healthy full-term infants. All infants with BPD had been previously discharged from the hospital after weaning from supplemental oxygen. Studies were done at a mean postconceptional age of 43 weeks while the infants were fed at home by one of their parents. Levels of SaO2 for the three groups were comparable before and during feeds. After feeding, the infants with BPD had significantly lower mean levels of SaO2 (84 +/- 8% [SD] vs 93 +/- 4% and 93 +/- 3%, respectively; P less than .01). They also spent more time after feeding with an SaO2 less than 90% (64 +/- 34% of time vs 27 +/- 33% for the very low birth weight and 22 +/- 20% for the term group; P less than .01) and greater time with an SaO2 less than 80% (37 +/- 28% vs 4 +/- 10% and 4 +/- 8%, respectively; P less than .01). Desaturation in infants with BPD was related to larger volume and faster oral intake during feeding. Thus, the data indicate that desaturation after feeding remains a recurrent problem for survivors of BPD after discharge.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Bronchopulmonary Dysplasia/blood , Eating , Infant, Low Birth Weight , Infant, Premature, Diseases/blood , Infant, Premature , Oxygen/blood , Body Weight , Follow-Up Studies , Gestational Age , Humans , Infant Food , Infant, Newborn , Oximetry , Time Factors
15.
Brain Cogn ; 19(2): 208-52, 1992 Jul.
Article in English | MEDLINE | ID: mdl-1642861

ABSTRACT

Children and adolescents with unilateral left- or right-hemisphere lesions were administered a standardized test of mathematics ability and a battery of experimental tests that examined the components of numerical and arithmetic processing. All lesioned groups showed at least marginally lower scores on the standardized test than the controls. More importantly, lesion-related deficits in performance were observed, especially for younger left-lesioned subjects (ages 7-12), on the verbal counting, digit matching, speeded addition, and written subtraction tasks; deficits among younger right-lesioned subjects were similar in nature, yet less pronounced than in the left-hemisphere group. Older left-lesioned subjects showed differences from their controls only on complex verbal counting and speeded addition. Correlations among the various measures indicated two further points. First, earlier onset of left-hemisphere lesion is associated with more serious disruption of mathematical processing. Second, these disruptions are not well assessed by a typical standardized test of mathematical performance, but are clearly in evidence with more precise, focused tasks.


Subject(s)
Brain Diseases/physiopathology , Cognition Disorders/physiopathology , Functional Laterality/physiology , Mathematics , Adolescent , Age Factors , Brain/physiology , Brain/physiopathology , Brain Diseases/diagnosis , Child , Cognition/physiology , Cognition Disorders/diagnosis , Female , Humans , Male , Problem Solving
16.
DICP ; 25(4): 344-7, 1991 Apr.
Article in English | MEDLINE | ID: mdl-1926899

ABSTRACT

The single-dose pharmacokinetics of ceftizoxime sodium were studied in 52 neonates and infants between 0.1 and 189 days of age. Subjects received ceftizoxime 25 or 50 mg/kg iv over 15-30 minutes. The drug was administered q8-12h for five days to permit tolerance evaluation on repetitive dosing. No differences were observed in ceftizoxime pharmacokinetic parameter estimates relative to dose. However, marked differences were observed in ceftizoxime pharmacokinetic characteristics relative to infant age; ceftizoxime half-life and mean residence time decreased, whereas body clearance increased with infant age. Ceftizoxime volume of distribution remained relatively constant over infant age. No adverse effects associated with ceftizoxime administration were observed. These data suggest that ceftizoxime 50 mg/kg q12h be used for infants less than or equal to 2 weeks of age (less than or equal to 40 weeks postconceptional age) and that 50 mg/kg q8h be administered for older infants.


Subject(s)
Ceftizoxime/pharmacokinetics , Aging/metabolism , Ceftizoxime/administration & dosage , Chromatography, High Pressure Liquid , Gestational Age , Humans , Infant , Infant, Newborn , Models, Biological
17.
Antimicrob Agents Chemother ; 34(6): 1172-7, 1990 Jun.
Article in English | MEDLINE | ID: mdl-2393278

ABSTRACT

The first-dose and multidose pharmacokinetics of imipenem and cilastatin were evaluated in 41 premature infants during their first week of life. Premature infants (gestational age, less than or equal to 37 weeks) were assigned to receive 10-, 15-, 20-, or 25-mg/kg doses of imipenem-cilastatin (1:1) as a single- or multiple-dose regimen. A total of 39 infants received a single dose, whereas 18 infants received multiple doses. No differences were observed in pharmacokinetic parameter estimates for either agent relative to the dose administered or infant body weight; thus, the data were pooled. Elimination half-life, steady-state volume of distribution, and body clearance averaged 2.5 h, 0.5 liter/kg, and 2.5 ml/min per kg, respectively, for imipenem and 9.1 h, 0.4 liter/kg, and 0.5 ml/min per kg, respectively, for cilastatin. Similar values for these parameter estimates were observed after multidose administration, although substantial accumulation of cilastatin in serum was observed. A total of 21% of the imipenem and 43% of the cilastatin were excreted unchanged in the urine over a 12-h collection period. Corresponding renal clearances averaged 0.4 and 0.2 ml/min per kg for imipenem and cilastatin, respectively. Substantial differences were observed in the route by which imipenem was cleared from the body compared with data from adult volunteers. These data suggest that infants should receive an imipenem dose of 20 mg/kg administered every 12 h for the treatment of bacterial infections outside the central nervous system.


Subject(s)
Cilastatin/pharmacokinetics , Imipenem/pharmacokinetics , Infant, Premature/metabolism , Humans , Infant, Newborn
18.
Comput Biomed Res ; 22(6): 516-31, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2591206

ABSTRACT

Conventional relational database management systems fail to address three features of statistical data management in a biomedical/clinical database, namely, that (1) statistical and medical data (SMD) require a great deal of space and need to be stored in a reduced form with minimal duplication; indeed, SMD have many derived/calculated and summary statistics that make the number of attributes in a relation (i.e., a set of records) grow rapidly and dynamically; (2) most SMD have hierarchical structures that are difficult to manage using the relational data model since SMD are stored in separate relations for duplication and space considerations; and (3) the management of SMD is made easier if it is possible to reorganize relations or group data, a capability lacking in conventional relational database management systems. In this paper, we (1) introduce five extended relational operators, (lattice) NEST, (lattice) UNNEST, MERGE, SPREAD, and GEN, to reorganize relations; (2) integrate the extended operators with conventional relational algebra and introduce the concept of the lattice relational model; and (3) give applications of the extended relational operators and the lattice relational model in solving the problems of statistical data manipulation in medical databases.


Subject(s)
Database Management Systems , Mathematical Computing , Medical Informatics Computing , Software , Hospital Records , Humans
19.
Cancer Res ; 49(13): 3520-4, 1989 Jul 01.
Article in English | MEDLINE | ID: mdl-2731174

ABSTRACT

The purine analogue O6-methylguanine is an effective biochemical modulator of the DNA repair protein O6-alkylguanine-DNA alkyltransferase. Inactivation of the alkyltransferase by O6-methylguanine sensitizes tumor cells to nitrosoureas in vitro. The pharmacokinetics of O6-methylguanine were evaluated in female Sprague-Dawley rats following administration of a single 40 mg/kg i.v. bolus dose. Two-compartment pharmacokinetic analysis revealed a terminal elimination half-life of 2.3 +/- 0.68 h, a total body clearance of 6.0 +/- 0.53 ml/min/kg, and a volume of distribution at steady state of 948 +/- 186 ml/kg. To inactivate the alkyltransferase, 80 mg/kg O6-methylguanine was given at 0 and 2 h. Alkyltransferase decreased in bone marrow, kidney, lung, spleen, and intestine by 20-90%. Regeneration of alkyltransferase activity was observed 22-70 h after the first bolus dose of O6-methylguanine. A continuous infusion protocol, which achieved a steady state serum concentration of 10.3 +/- 1.5 micrograms O6-methylguanine/ml at 15 h, resulted in a similar degree of inactivation of tissue alkyltransferase to that observed following bolus drug infusion. O6-Methylguanine tissue concentrations, including that determined in brain, were 1.7- to 4-fold higher than that in serum, indicating that O6-methylguanine is concentrated in most if not all tissues. These studies establish pharmacokinetic parameters of O6-methylguanine in rats and suggest that effective biochemical modulation of alkyltransferase can be achieved in vivo. Further studies are indicated to assess the extent to which biochemical modulation of alkyltransferase reduces tumor nitrosourea resistance in vivo.


Subject(s)
DNA/metabolism , Guanine/analogs & derivatives , Methyltransferases/metabolism , Animals , Dose-Response Relationship, Drug , Drug Administration Schedule , Guanine/administration & dosage , Guanine/pharmacokinetics , Injections, Intravenous , Metabolic Clearance Rate , Methyltransferases/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase , Rats , Tissue Distribution
20.
Leuk Res ; 13(9): 771-9, 1989.
Article in English | MEDLINE | ID: mdl-2796383

ABSTRACT

We determined lymphocyte aryl hydrocarbon hydroxylase (AHH) inducibility for members of 13 families with one or more children with acute lymphoblastic leukemia (ALL) and 12 control families. Pedigree analysis suggested that aromatic hydrocarbon responsiveness (i.e. inducibility) is a codominant trait. Heterozygotes were found to be moderately responsive with IR values intermediate between homozygous minimally responsive and homozygous highly responsive individuals. Homozygous recessive and heterozygous genotypes accounted for 54% and 36% of ALL children respectively. The risk of ALL among minimally aromatic hydrocarbon responsive children was twice that of highly responsive children.


Subject(s)
Aryl Hydrocarbon Hydroxylases/biosynthesis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Aryl Hydrocarbon Hydroxylases/genetics , Enzyme Induction , Humans , Pedigree , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...