Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Clin Invest ; 67(1): 93-102, 1981 Jan.
Article in English | MEDLINE | ID: mdl-7451659

ABSTRACT

Human as well as murine granulocytes have been shown to kill the larval stages of helminth parasites; the mechanism of this cell-mediated cytotoxicity is, however, poorly understood. The present study was designed to assess the role of peroxidative processes in killing of schistosomula of Schistosoma mansoni by human granulocytes in vitro. The rate of H(2)O(2) production by human neutrophils, eosinophils, and basophils was measured upon incubation with schistosomula alone or in the presence of specific antibody or complement. Opsonized parasites (antibody and/or complement) increased the rate of H(2)O(2) production by neutrophils, eosinophils, and basophils by respective percentages of 500, 500, and 371. The rate of H(2)O(2) release was directly related to the number of granulocytes and to the proportion of cells attached to the surface of the schistosomula. Increased hydrogen peroxide release occurred by 10 min of incubation and was demonstrable up to 16 h after addition of leukocytes to schistosomula. The primary source of this oxygen product was found to be the granulocytes adherent to the schistosomula and not those that remained unattached. Hydrogen peroxide production by neutrophils and eosinophils was quantitatively similar (schistosomula coated with antibody plus complement stimulated 5 x 10(6) neutrophils and eosinophils to release H(2)O(2) at respective rates of 0.35 and 0.40 nmol/min). Granulocyte-mediated parasite killing correlated with rate of H(2)O(2) generation; both processes were inhibited by catalase. To define further the role of oxidative metabolites, neutrophils and eosinophils of two subjects with chronic granulomatous disease were used; marked reduction of granulocyte-mediated parasite mortality was observed. Peroxidase was required for H(2)O(2)-mediated killing. Addition of the peroxidase inhibitors azide (1 mM), cyanide (1 mM), or aminotriazole (1 cM) to neutrophilschistosomula mixtures significantly reduced parasite cytotoxicity (P < 0.01); similar reduction was observed when eosinophils were used (P < 0.01). Fixation of halide (iodide) to trichloroacetic acid-precipitable protein (2.4-6.0 nmol/h per 10(7) neutrophils) was demonstrated in the presence of granulocytes, opsonins, and parasites; this process was completely inhibited by 1 mM azide. These data indicate that contact between the surfaces of human granulocytes and schistosomula results in release of cellular hydrogen peroxide and iodination. The generation of H(2)O(2) and its interaction with peroxidase appear to be crucial in effecting in vitro granulocyte-mediated parasite cytotoxicity.


Subject(s)
Granulocytes/physiology , Hydrogen Peroxide/metabolism , Schistosoma mansoni/physiology , Animals , Basophils/physiology , Eosinophils/physiology , Granulomatous Disease, Chronic/blood , Humans , Neutrophils/physiology , Opsonin Proteins , Schistosoma mansoni/immunology
2.
Cancer Res ; 41(6): 2305-7, 1981 Jun.
Article in English | MEDLINE | ID: mdl-7016312

ABSTRACT

The mutagenicity of niridazole for Salmonella typhimurium depends upon the enzymic reduction of the nitro function. The response of niridazole nitroreductase-deficient bacteria to niridazole is reduced to 4.4 and 0.19% that exhibited by the enzyme-proficient parent strain when the deficiency is the result of a base substitution and frame-shift mutation, respectively. The results are taken to indicate that the residual activity (4.4%) seen in the strain with a base substitution mutation reflects the activity of an enzyme with an amino acid substitution, while the basal level (0.19%) of activity indicates the action of a different nitroreductase with a low specificity for niridazole.


Subject(s)
Niridazole/pharmacology , Oxidoreductases/metabolism , Salmonella typhimurium/drug effects , Genotype , Mutagens , Mutation/drug effects , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics
3.
Cancer Res ; 40(12): 4599-605, 1980 Dec.
Article in English | MEDLINE | ID: mdl-6893679

ABSTRACT

Niridazole is a nitrothiazole anthelmintic agent used to treat schistosomiasis. Its antibacterial activity was found to require the presence of the nitro group; a synthetic desnitro analog was completely inactive. Niridazole was mutagenic for Salmonella tester strains TA1538, TA98, and TA100, suggesting that it was both a frame-shift- and a base substitution-type mutagen. It was effective under both aerobic and anaerobic conditions, while similar testing of the desnitro niridazole produced consistently negative results. Addition of rat liver S-9 fraction under either aerobic or anaerobic conditions did not enhance mutagenicity. However, since bacterial killing limited the dose of niridazole to 0.33 microgram/plate in standard tester strains (1/20 Km for the mammalian liver enzymes), further studies were performed using niridazole-resistant, histidine-dependent mutants derived from strains TA98 and TA100. These mutants were found to be nitroreductase deficient and to resist the mutagenic effects of niridazole, in the presence or absence of S-9, up to concentrations of 10 microgram/plate. In addition, even at niridazole concentrations of up to 100 microgram/plate, rat liver S-9 was ineffective in enhancing the mutagenicity of niridazole. These results suggest that the mutagenicity of niridazole is dependent on its aromatic nitro group and a specific bacterial nitroreductase.


Subject(s)
Mutagens , Niridazole , Oxidoreductases/metabolism , Aerobiosis , Anti-Bacterial Agents , Biotransformation , Microsomes, Liver/metabolism , Niridazole/metabolism , Nitro Compounds/metabolism , Nitroreductases , Structure-Activity Relationship
4.
J Clin Oncol ; 15(1): 94-102, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8996129

ABSTRACT

PURPOSE: To compare the efficacy, characteristics of onset/recovery, and safety of ketamine/atropine/midazolam with meperidine/midazolam used as premedication for painful procedures in children with cancer. METHODS: A randomized, double-blind crossover trial for two successive painful procedures (bone marrow aspiration or biopsy, lumbar puncture, or combined procedures) was performed at a referral-based pediatric hematology-oncology clinic and associated inpatient service of a university teaching hospital. Twenty-two children, aged 24 to 178 months, were enrolled and 18 (81.8%) completed the double-blind, crossover trial. Each child received intravenous premedication with either meperidine 2 mg/kg and midazolam 0.1 mg/kg (MM) or atropine 0.01 mg/kg, midazolam 0.05 mg/kg, and ketamine 1.5 mg/kg (KM) on one occasion followed by the alternative regimen on a second occasion. The initial premedication regimen was chosen by random assignment. RESULTS: Efficacy was assessed by a trained observer using the Observational Scale of Behavioral Distress-Revised (OSBD-R). Operator, nurse, parent, and patient opinions of efficacy were recorded on a visual analog scale (VAS). Side effects were monitored by pulse oximetry, nasal end-tidal capnography, and serial blood pressure measurements. Use of KM resulted in significantly less procedural distress than MM (1.37 +/- 2.20 v 7.04 +/- 8.06 OSBD-R units; P < .05). Both operators and nurses rated KM more effective than MM. KM use was associated with earlier readiness for the procedure (19.2 v 24.0 minutes) and more rapid recovery (39.3 v 74.6 minutes for removal of monitoring devices and 58.5 v 87.1 minutes for discharge). Procedures undertaken after ketamine sedation were associated with fewer side effects than observed with MM sedation (hypoxia, 17.7% v 82.4%; hypotension, 16.6% v 55.6%; reduced respiratory rate, 0% v 38.9%). The incidence of emergence reactions or behavioral abnormalities within 24 hours postprocedure was similar in both treatment groups. At 7 days postprocedure, no child had persistent behavioral abnormalities and all children had amnesia for the procedure. Parents and children expressed a preference for KM over MM in 12 of 18 cases (P < .05). CONCLUSION: A premedication regimen of KM produced superior sedation with a faster onset and recovery and fewer side effects than a MM combination.


Subject(s)
Anesthetics , Biopsy , Bone Marrow Examination , Hypnotics and Sedatives , Ketamine , Meperidine , Midazolam , Spinal Puncture , Child , Child Behavior/drug effects , Child, Preschool , Cross-Over Studies , Double-Blind Method , Female , Humans , Male
5.
Clin Pharmacol Ther ; 44(2): 232-8, 1988 Aug.
Article in English | MEDLINE | ID: mdl-3293877

ABSTRACT

We have evaluated the utility of optimal sampling strategy coupled with adaptive study design in the determination of individual patient and population pharmacokinetic parameter values. In 9 patients with cystic fibrosis receiving a short (1 minute) infusion of ceftazidime pharmacokinetic parameter values were determined with a nonlinear least-squares estimator analyzing a traditional, geometrically spaced set of 12 postinfusion serum samples drawn over 8 hours. These values were compared with values generated from four sample subsets of the 12 obtained at optimal times and analyzed by nonlinear least-squares estimator, as well as a maximum a posteriori probability Bayesian estimator with prior distributions placed on beta and clearance. The four sampling times were determined according to an adaptive design optimization technique that employs sequential updating of population prior distributions on parameter values. Compared with the 12-point determination, the four optimal points analyzed with the maximum a posteriori probability Bayesian estimator faithfully reproduced both microscopic and hybrid pharmacokinetic parameter values for individual patients and, consequently, also produced accurate measures of population central tendency and dispersion. This has important implications in being able to more efficiently derive target patient population pharmacokinetic information for new drugs. This should also allow generation of better concentration-effect relationships in populations of interest.


Subject(s)
Blood Specimen Collection , Pharmacokinetics , Adolescent , Adult , Bayes Theorem , Ceftazidime/blood , Ceftazidime/pharmacokinetics , Child , Chromatography, High Pressure Liquid , Cystic Fibrosis/blood , Cystic Fibrosis/metabolism , Female , Humans , Male , Models, Biological , Research Design , Retrospective Studies
6.
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
7.
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
8.
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
9.
Am J Med ; 82(4A): 174-9, 1987 Apr 27.
Article in English | MEDLINE | ID: mdl-3555032

ABSTRACT

Ciprofloxacin has potent in vitro activity against Pseudomonas aeruginosa and Pseudomonas cepacia strains isolated from cystic fibrosis patients. Our previous single-dose pharmacokinetic and pharmacodynamic studies identified important differences between cystic fibrosis patients and age- and sex-matched controls. Based on these data, 30 acutely ill cystic fibrosis patients (aged 18 to 44 years) received 750 mg of ciprofloxacin orally every eight hours for 21 days. Multiple timed serum, urine, and sputum samples for pharmacokinetic analysis were obtained on Days 3, 12, 14, and 21 of the study. Estimates of steady-state pharmacokinetic parameters averaged (+/- SD): t1/2 beta, 3.8 (1) hours; Vd/F, 4.4 (2) liters/kg; Cl/F, 772.9 (301) ml/minute/1.73 m2; Fe, 46 percent; peak, 5.4 (2) mg/liter; and trough, 1.8 (0.8) mg/liter. Serum ciprofloxacin concentrations and pharmacokinetic estimates remained unchanged throughout the study. Sputum ciprofloxacin concentrations exceeded those observed in serum. Sputum cultures revealed 43 P. aeruginosa (MIC90 = 2 micrograms/ml) and 15 P. cepacia (MIC90 = 16 micrograms/ml) strains. Sputum ciprofloxacin concentrations exceeded the MIC90 for P. aeruginosa approximately fivefold, yet only eight isolates were fully suppressed. Posttreatment sputum cultures revealed 35 P. aeruginosa (MIC90 = 16 micrograms/ml) and 15 P. cepacia (MIC90 = 16 micrograms/ml). All patients showed clinical improvement based upon the results of pulmonary function tests and an acute clinical efficacy score (median pre 49/post 60). No patients experienced drug-related toxicity. Ciprofloxacin monotherapy is effective for the acute treatment of cystic fibrosis patients. The development of pathogen resistance during oral therapy may limit its utility in ambulatory patients.


Subject(s)
Ciprofloxacin/therapeutic use , Cystic Fibrosis/drug therapy , Lung Diseases/drug therapy , Pseudomonas Infections/drug therapy , Adolescent , Adult , Ciprofloxacin/administration & dosage , Ciprofloxacin/metabolism , Clinical Trials as Topic , Cystic Fibrosis/complications , Female , Humans , Kinetics , Male , Pseudomonas Infections/complications
10.
Am J Med ; 79(2A): 37-46, 1985 Aug 09.
Article in English | MEDLINE | ID: mdl-3895917

ABSTRACT

The in vitro activity of ceftazidime against Pseudomonas aeruginosa and P. cepacia isolates from patients with cystic fibrosis was compared with that of other antipseudomonal drugs. Ceftazidime was as potent as imipenem against P. aeruginosa and the only drug effective against P. cepacia. An evaluation of the elimination kinetics of ceftazidime in 20 cystic fibrosis patients revealed an elimination half-life of 1.76 hours, an apparent distribution volume of 0.27 liters/kg, and a serum clearance rate of 133.9 ml/minute/1.73m2. Urinary recovery of ceftazidime was 87 percent within the first 24 hours after administration of the drug, with 65 percent recovered in the first two-hour fraction. Probenecid administration had no effect on the elimination kinetics of ceftazidime. Forty-three patients who had either shown no response to conventional therapy or had sputum Pseudomonas isolates that were susceptible only to ceftazidime received 75 courses of therapy. In 67 percent of these patients, the clinical response, when evaluated using an objective clinical efficacy scoring system, was considered favorable. Clinical failures were not associated with the development of drug resistance. Thus, ceftazidime can be recommended for the treatment of acute pulmonary exacerbations in patients with cystic fibrosis.


Subject(s)
Ceftazidime/therapeutic use , Cystic Fibrosis/drug therapy , Pseudomonas/drug effects , Adolescent , Adult , Ceftazidime/metabolism , Ceftazidime/pharmacology , Child , Cystic Fibrosis/microbiology , Drug Resistance, Microbial , Female , Humans , Kinetics , Male , Microbial Sensitivity Tests , Probenecid/pharmacology , Sputum/metabolism
11.
Pediatrics ; 91(2): 369-78, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8424013

ABSTRACT

Children hospitalized in a pediatric intensive care unit are frequently distressed. The purpose of this study was to identify the patterns of use of sedative agents in pediatric critical care patients. A questionnaire survey was mailed to 45 directors of Pediatric Critical Care Fellowship Training Programs listed in Critical Care Medicine, January 1989. The response rate was 75.6% (34 questionnaires). The most commonly identified goals of sedation were reduced patient discomfort or distress and fewer unplanned extubations. The agents most frequently employed for this purpose were opioids (morphine or fentanyl), chloral hydrate, or benzodiazepines. Although conventional doses are used, opioids and benzodiazepines are often given hourly or by continuous infusion. Satisfaction with the efficacy and safety of commonly used opioids was greater (most common response "very satisfied") than for the benzodiazepines ("somewhat satisfied"). The physician's or nurse's clinical impression was reported to be the "most important" criterion for deciding when a patient required a dose of sedative; objective criteria were selected as less important. The majority of patients (65.7%) in the surveyed units were ideally "sedated to the point of no distress with as-needed medication." The majority of respondents (76.4%) identified efficacy as the major problem with sedation. Drug withdrawal was considered to be the major problem with sedative use by only a minority of respondents (6.9%). Although withdrawal is seen in 61.8% of units, it is generally treated when recognized, rather than prevented by routine tapering of sedation. Optimal sedation of pediatric intensive care unit patients is considered problematic, despite the use of frequent doses of many sedatives. Systematic investigation of pharmacodynamic response to these agents in the pediatric critical care population is indicated.


Subject(s)
Conscious Sedation/statistics & numerical data , Fellowships and Scholarships/statistics & numerical data , Intensive Care Units, Pediatric/statistics & numerical data , Pediatrics/methods , Practice Patterns, Physicians'/statistics & numerical data , Stress, Psychological/drug therapy , Attitude of Health Personnel , Canada , Conscious Sedation/adverse effects , Conscious Sedation/methods , Evaluation Studies as Topic , Hospital Bed Capacity , Hospitals/classification , Humans , Intensive Care Units, Pediatric/organization & administration , Organizational Objectives , Pediatrics/education , Pediatrics/organization & administration , Physician Executives/psychology , Respiration, Artificial/adverse effects , Stress, Psychological/diagnosis , Stress, Psychological/etiology , Surveys and Questionnaires , United States
12.
Clin Pharmacokinet ; 35(1): 37-47, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9673833

ABSTRACT

Midazolam is a parenteral benzodiazepine with sedative, amnesic, anxiolytic, muscle relaxant and anticonvulsant properties. The drug exerts its clinical effect by binding to a receptor complex which facilitates the action of the inhibitory neurotransmitter gamma-aminobutyric acid (GABA). Midazolam has a faster onset and shorter duration of action than other benzodiazepines such as diazepam and lorazepam. The most serious adverse events associated with midazolam in children include hypoventilation, decreased oxygen saturation, apnoea and hypotension. It is water soluble in the commercially prepared formulation but becomes lipid soluble at physiological pH and can then cross the blood brain barrier. It is metabolised in the liver by the cytochrome P450 system, and its chief metabolite is 1-hydroxymethyl midazolam. The latter is conjugated to the glucuronide form, and it has only minimal biological activity. Midazolam is excreted primarily by the kidney. Its half-life in children over 12 months is reported to be 0.8 to 1.8 hours, with a clearance of 4.7 to 19.7 ml/min/kg. Doses given to children must be calculated on a mg/kg basis. For children 6 months to 5 years of age the initial dose is 0.05 to 0.1 mg/kg. A total dose up to 0.6 mg/kg titrated slowly may be necessary to achieve the desired endpoint. For children 6 to 12 years of age the initial dose is 0.025 to 0.05 mg/kg with a total dose up to 0.4 mg/kg to achieve the desired end-point.


Subject(s)
Midazolam/pharmacokinetics , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/pharmacokinetics , Anti-Anxiety Agents/pharmacology , Child , Drug Interactions , Drug Tolerance , Emergency Service, Hospital , Humans , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/pharmacokinetics , Hypnotics and Sedatives/pharmacology , Infant , Intensive Care Units , Midazolam/administration & dosage , Midazolam/pharmacology
13.
Clin Pharmacokinet ; 14(4): 189-216, 1988 Apr.
Article in English | MEDLINE | ID: mdl-3292100

ABSTRACT

Rational pharmacotherapy is dependent upon an understanding of the clinical pharmacokinetic and pharmacodynamic properties of the drugs employed. Although the available data on drug biodisposition and action in the neonate have increased considerably in the last few years, pharmacokinetic-pharmacodynamic interactions for many drugs remain poorly understood. The ontogeny of drug absorption, distribution, metabolism, and elimination are addressed in this review. Drug absorption from any site depends upon both the physicochemical properties of the drug and a variety of patient factors. Absorption of orally administered drugs may be affected by changes in gastric acidity and emptying time as well as by bile salt pool size, bacterial colonisation, and extraintestinal disease states such as congestive heart failure. Factors affecting drug absorption following intramuscular, percutaneous, and rectal administration are also discussed. Drug distribution in the neonate is influenced by a variety of important and predictable age-dependent factors. The developmental aspects of protein binding and body water compartments are described. Additionally, hepatic drug metabolism assumes an important role in understanding the pharmacokinetic and pharmacodynamic properties of many compounds. Certain biotransformation pathways, including hydroxylation by the P450 mono-oxygenase system and glucuronidation, demonstrate only limited activity at birth, while other pathways, such as sulphate or glycine conjugation, appear very efficient at birth. Elimination of drugs excreted unchanged in the urine is dramatically reduced in the newborn, compared with older infants and children, due to immaturity of both glomerular filtration and tubular secretory processes. The glomerular filtration rate remains markedly reduced prior to 34 weeks gestational age, increasing as a function of post-conceptual age until adult values are achieved by approximately 2.5 to 5 months of age. Tubular secretory capacity is also limited at birth, approaching adult values by approximately 7 months of age. Published reports describing the pharmacokinetics and pharmacodynamics of commonly used drugs in the neonatal period, as well as differences in drug biodisposition among premature infants, full term neonates, and older infants and children, are reviewed. Our recommendations for neonatal drug therapy are based upon a critical interpretation of these data, an understanding of fetal development and maturational processes, and an understanding of how disease states may affect drug biodisposition in the neonate.


Subject(s)
Infant, Newborn/metabolism , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Absorption , Drug Administration Routes , Half-Life , Humans , Liver/metabolism , Pharmaceutical Preparations/administration & dosage , Protein Binding , Tissue Distribution
14.
Am J Cardiol ; 57(2): 44A-53A, 1986 Jan 24.
Article in English | MEDLINE | ID: mdl-3511657

ABSTRACT

Careful management of fluid and electrolytes has long been an intrinsic part of pediatric practice. However, the augmentation of these manipulations through the rational use of diuretic agents requires considerable skill. In pediatric medicine, the regulation of pharmacokinetic processes and their interface with pharmacodynamic processes show dramatic age-related changes. These ontogenetic processes and their modification by various disease states must be considered carefully before selection and application of diuretic agents. The available data concerning the ontogeny of renal function and the attempts to apply diuretic therapy to pediatric disease are reviewed. It is concluded that results obtained to date suffer from the absence of a rigorous attempt to answer the fundamental therapeutic questions: What drug? What dose? What duration of therapy? A rational "target-effect" strategy is proposed for the application of diuretic agents to pediatric medicine.


Subject(s)
Diuretics/therapeutic use , Acid-Base Equilibrium , Bumetanide/metabolism , Bumetanide/therapeutic use , Calcium/urine , Child , Child, Preschool , Chlorides/metabolism , Diuretics/adverse effects , Diuretics/metabolism , Diuretics/pharmacology , Ethacrynic Acid/metabolism , Ethacrynic Acid/therapeutic use , Furosemide/metabolism , Furosemide/therapeutic use , Glomerular Filtration Rate , Glomerulonephritis/drug therapy , Humans , Hydrocephalus/drug therapy , Infant , Infant, Newborn , Infant, Premature , Kidney/blood supply , Kidney/drug effects , Kidney/embryology , Kidney/physiology , Kinetics , Metabolic Clearance Rate , Nephrotic Syndrome/drug therapy , Prostaglandins/biosynthesis , Pulmonary Edema/drug therapy , Regional Blood Flow , Water-Electrolyte Balance
15.
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
16.
Aliment Pharmacol Ther ; 16(5): 899-907, 2002 May.
Article in English | MEDLINE | ID: mdl-11966498

ABSTRACT

BACKGROUND: The use of over-the-counter antacids has increased in children under the age of 12 years, and has been followed by an apparent increase in the use of over-the-counter histamine-2 receptor antagonists. However, the pharmacokinetic and pharmacodynamic effects of over-the-counter histamine-2 receptor antagonists in the paediatric population are largely unknown. AIM: To evaluate the pharmacokinetics and pharmacodynamics of a single dose of the over-the-counter histamine-2 receptor antagonist, ranitidine, 75 mg, in children with symptoms of gastro-oesophageal reflux disease. METHODS: Children aged between 4 and 11 years with symptoms of heartburn suspected to be due to gastro-oesophageal reflux disease were recruited at six clinical centres. Following a single dose of either oral ranitidine, 75 mg (n=19), or placebo (n=10), recording of intragastric pH and serial blood sampling were carried out for 6 h. RESULTS: The estimated pharmacokinetic parameters of ranitidine, 75 mg, were as follows: the median Cmax value of 477 ng/mL occurred within a median of 2.5 h after dosing, and the median half-life was 2.0 h. The intragastric pH began to rise approximately 30 min after dosing with ranitidine to a peak of pH; 4. The pH in the ranitidine group remained higher than that in the placebo group throughout the 6-h evaluation period. Adverse events were generally mild. CONCLUSIONS: Ranitidine, 75 mg, significantly increased the intragastric pH in children aged 4-11 years. The pharmacokinetic and pharmacodynamic profiles were similar to those in adults. Ranitidine, 75 mg, appears to be effective for the control of intragastric acidity for 5-6 h in children aged 4-11 years.


Subject(s)
Anti-Ulcer Agents , Gastroesophageal Reflux/drug therapy , Ranitidine , Anti-Ulcer Agents/pharmacokinetics , Anti-Ulcer Agents/pharmacology , Anti-Ulcer Agents/therapeutic use , Area Under Curve , Child , Child, Preschool , Double-Blind Method , Female , Half-Life , Humans , Hydrogen-Ion Concentration , Male , Ranitidine/pharmacokinetics , Ranitidine/pharmacology , Ranitidine/therapeutic use
17.
Pediatr Infect Dis J ; 17(11): 1070-5; discussion 1099-100, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9850001

ABSTRACT

Selection of appropriate antibiotic treatment for children with acute otitis media (AOM) is challenging. Although the diagnosis is relatively easy for experienced clinicians, the distinction between AOM and otitis media with effusion is often more subtle. In general therapy is empiric and the pathogen causing disease in a given patient remains unknown. However, this situation is made even more difficult by the dynamic nature of the pathogenesis of AOM. Both the proportion of patients infected with one of the three primary pathogens, Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis, and the antimicrobial susceptibility patterns of these pathogens are changing. Currently there are 16 antibiotics labeled for use in AOM. Only 2 are reliably effective against penicillin-resistant pneumococcus: high dose amoxicillin (80 to 100 mg/kg/day) and im ceftriaxone. Among the others all are beta-lactamase-stable and have proven clinical effectiveness in AOM patients infected with H. influenzae or M. catarrhalis. Even with the high spontaneous resolution rate reported for AOM, antimicrobial therapy remains the standard of care in the United States. Recognition of the fundamental determinants of effective therapy should permit rational antibiotic selection for each patient.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/therapeutic use , Otitis Media/drug therapy , Acute Disease , Amoxicillin/therapeutic use , Bacterial Infections/diagnosis , Bacterial Infections/drug therapy , Ceftriaxone/therapeutic use , Child , Child, Preschool , Decision Making , Drug Resistance, Microbial , Humans , Infant , Otitis Media/diagnosis , Otitis Media/microbiology
18.
Pediatr Infect Dis J ; 20(3): 337-42, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11303847

ABSTRACT

BACKGROUND: Because determining the pharmacokinetics of drugs used in pediatric patients allows for appropriate dosing and optimal clinical response, we have reviewed the pharmacokinetic data on the use of cefepime in the pediatric population. METHODS: Three studies encompassing 88 patients ages 2 months to 16 years examined the pharmacokinetics of cefepime given as a single iv dose, as multiple iv doses and by im administration. In all studies serial blood and urine or cerebrospinal fluid (CSF) samples were collected after a single dose and/or at steady state, defined as after at least 2 days of dosing. Pharmacokinetic parameters were generated from concentration-vs.-time curves and were analyzed using noncompartmental methods. RESULTS: In all studies cefepime exhibited a linear pharmacokinetic profile and concentrations declined proportionally over time. Minimal accumulation was observed after multiple dosing. Pharmacokinetic parameters were similar in all studies and appeared to be dose-independent. Mean (range) parameters observed in this review were: t 1/2 = 1.7 h (1.26 to 1.93); volume of distribution at steady state, 0.37 liter/kg (0.33 to 0.40); total body clearance, 3.1 ml/min/kg (1.43 to 4.01); renal total body clearance, 2.3 ml/min/kg (1.86 to 3.05); absolute bioavailability of cefepime after the im dose, 82.3%; and urinary recovery, 72% (57 to 85%). Penetration into CSF appeared to be good, with CSF concentrations averaging 3.3 to 5.7 microg/ml 0.5 and 8 h after administration of the dose, respectively. CONCLUSION: Cefepime displayed a linear pharmacokinetic profile, was well-absorbed via im injection and had adequate penetration into the CSF of patients with bacterial meningitis, compared with other beta-lactams.


Subject(s)
Cephalosporins/pharmacokinetics , Meningitis, Bacterial/drug therapy , Adolescent , Cefepime , Cephalosporins/administration & dosage , Cephalosporins/cerebrospinal fluid , Cephalosporins/therapeutic use , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Humans , Infant , Injections, Intramuscular , Injections, Intravenous , Male , Safety , Time Factors , Treatment Outcome
19.
Pediatr Infect Dis J ; 10(5): 375-80, 1991 May.
Article in English | MEDLINE | ID: mdl-1906160

ABSTRACT

A total of 137 children with acute otitis media with effusion were randomly allocated to treatment with cefprozil (30 mg/kg/day divided into two equal doses), an investigational cephalosporin or amoxicillin clavulanate potassium (40 mg/kg/day divided into three equal doses) for 10 days. The most common pathogens obtained from middle ear cavities by tympanocentesis were Streptococcus pneumoniae (33%), Haemophilus influenzae (19.6%) and Moraxella catarrhalis (8.3%). Patients were scheduled for follow-up visits at midtreatment, at end of therapy and at 30 days. Of the 137 children 122 were evaluable. Five of 60 patients (8.3%) treated with cefprozil and 14 of 62 patients (22.5%) treated with amoxicillin clavulanate potassium were considered therapeutic failures because of persistence of symptoms and/or isolation of the original pathogen or superinfection (P = 0.05). Rates of relapse, reinfection and persistent middle ear effusion as documented by tympanogram were comparable in both groups. When persistent middle ear effusion was analyzed by pneumatic otoscopy, 64 of 103 affected ears (62.1%) treated with cefprozil and 80 of 105 affected ears (76.1%) treated with amoxicillin clavulanate potassium were abnormal (P = 0.04). Loose stools were more common in children treated with amoxicillin clavulanate potassium than in children treated with cefprozil (P = 0.0004). Based on the efficacy results from this study, the lower gastrointestinal side effects and the convenience of twice-a-day dosing, we believe that cefprozil in a dosage of 30 mg/kg/day divided every 12 hours represents a potential alternative for the treatment of acute otitis media with effusion in children.


Subject(s)
Amoxicillin/therapeutic use , Cephalosporins/therapeutic use , Clavulanic Acids/therapeutic use , Otitis Media with Effusion/drug therapy , Acute Disease , Adolescent , Amoxicillin/adverse effects , Amoxicillin-Potassium Clavulanate Combination , Cephalosporins/adverse effects , Child , Child, Preschool , Clavulanic Acids/adverse effects , Drug Administration Schedule , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/therapeutic use , Haemophilus influenzae/isolation & purification , Humans , Infant , Moraxella catarrhalis/isolation & purification , Otitis Media with Effusion/microbiology , Recurrence , Streptococcus pneumoniae/isolation & purification , Cefprozil
20.
Pediatr Infect Dis J ; 14(7 Suppl): S115-20, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7567311

ABSTRACT

A randomized, controlled, single blind clinical trial was conducted in children with acute otitis media to evaluate the safety and efficacy of a 10-day course of therapy with ceftibuten 9 mg/kg taken as a single daily dose, up to a maximum daily dose of 400 mg, compared with cefaclor 40 mg/kg/day in three divided doses, up to a maximum of 1 g/day. Patients were evaluated any time from 1 to 3 days after completion of therapy (posttreatment follow-up). A total of 154 patients (106 ceftibuten, 48 cefaclor) were evaluable for efficacy. Clinical success as determined by resolution (cure) or improvement of signs and symptoms of infection were seen in 89 and 88% of patients treated with ceftibuten and cefaclor, respectively, at the posttreatment follow-up visit. At the extended follow-up visit (any time from 2 to 4 weeks after completion of therapy), clinical success was sustained in 88 and 82% of the ceftibuten-treated and cefaclor-treated patients, respectively. A total of 391 patients (264 ceftibuten, 127 cefaclor) were included in the safety analysis. Treatment-related adverse experiences occurred in 8% of ceftibuten-treated patients and 14% of cefaclor-treated patients. All were mild or moderate and the majority were gastrointestinal. There were no deaths or serious adverse events. The results of this study suggest that ceftibuten is an effective and well-tolerated alternative to other antibiotic therapies for the treatment of children with acute otitis media.


Subject(s)
Cefaclor/therapeutic use , Cephalosporins/therapeutic use , Otitis Media with Effusion/drug therapy , Acute Disease , Adolescent , Bacterial Infections/drug therapy , Cefaclor/administration & dosage , Cefaclor/adverse effects , Ceftibuten , Cephalosporins/administration & dosage , Cephalosporins/adverse effects , Child , Child, Preschool , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Infant , Male , Otitis Media with Effusion/microbiology , Prospective Studies , Single-Blind Method , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL