feed icon rss

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    Elsevier BV ; 2019
    In:  Journal of Pharmaceutical and Biomedical Analysis Vol. 174 ( 2019-09), p. 256-262
    In: Journal of Pharmaceutical and Biomedical Analysis, Elsevier BV, Vol. 174 ( 2019-09), p. 256-262
    Type of Medium: Online Resource
    ISSN: 0731-7085
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2019
    detail.hit.zdb_id: 1491820-1
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Antimicrobial Agents and Chemotherapy, American Society for Microbiology, Vol. 62, No. 5 ( 2018-05)
    Abstract: Newborns with congenital cytomegalovirus (CMV) infection are at high risk for developing permanent sequelae. Intravenous ganciclovir therapy is frequently used for the treatment of congenital CMV infection. A target area under the concentration-time curve from 0 to 24 h (AUC 0–24 ) of 40 to 50 μg · h/ml is recommended. The standard dose has resulted in a large variability in ganciclovir exposure in newborns, indicating the unmet need of dosage individualization for this vulnerable population, but the implementation of this strategy remains challenging in clinical practice. We aim to evaluate the clinical utility of model-based dosage individualization of ganciclovir in newborns using an opportunistic sampling approach. The predictive performance of a published ganciclovir population pharmacokinetic model was evaluated using an independent patient cohort. The individual dose was adjusted based on the target AUC 0–24 to ensure its efficacy. A total of 26 newborns with congenital CMV infection were included in the present study. Only 11 (42.3%) patients achieved the target AUC 0–24 after being given the standard dose. For all the subtherapeutic patients (achieving 〈 80% of the target AUC) ( n = 5), a model-based dosage adjustment was performed using the Bayesian estimation method combined with the opportunistic sampling strategy. The adjusted doses were increased by 28.6% to 60.0% in these five patients, and all adapted AUC 0–24 values achieved the target (range, 48.6 to 66.1 μg · h/ml). The clinical utility of model-based dosing individualization of ganciclovir was demonstrated in newborns with congenital CMV infection. The population pharmacokinetic model combined with the opportunistic sampling strategy provides a clinically feasible method to adapt the ganciclovir dose in neonatal clinical practice. (This study has been registered at ClinicalTrials.gov under registration no. NCT03113344.)
    Type of Medium: Online Resource
    ISSN: 0066-4804 , 1098-6596
    RVK:
    Language: English
    Publisher: American Society for Microbiology
    Publication Date: 2018
    detail.hit.zdb_id: 1496156-8
    SSG: 12
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Archives of Disease in Childhood, BMJ, Vol. 101, No. 1 ( 2016-01), p. e1.37-e1
    Abstract: Pharmacokinetic modeling approach is often applied to evaluate antimicrobials in neonates. However, the clinical application of the model-based personalized antimicrobial therapy is still limited. Our objective is to evaluate the clinical utility and safety of the model-based patient-tailored dosing of vancomycin in neonates. Methods A model-based vancomycin dosing calculator, which was developed using the pharmacokinetic information from our previously published population pharmacokinetic model, has been integrated into the routine clinical care in 3 NICUs (Cochin, Robert Debré and Clocheville Hospitals) between June 2012 and November 2014. The target attainment rate was selected as the endpoint for evaluating the clinical utility. The percentage of patients achieving the target concentration of vancomycin was calculated using the first TDM samples taken 6–24 hours after starting vancomycin treatment. The safety evaluation was focused on nephrotoxicity, which was evaluated based on changes in serum creatinine concentration from a baseline value obtained within the 48 hours of starting vancomycin treatment. Results A total of 190 neonates were included. The mean of the 190 first TDM vancomycin concentrations was 20.0 mg/L (10th-90th percentiles: 13.1–27.6). After receiving patient-tailored doses, the target attainment rate was 71.6% (n=136) within the range of 15–25 mg/L (90.5%, n=172, within the range of 10–30 mg/L). None of the included patients has developed vancomycin-related nephrotoxicity. Conclusion The present work provides an evidence-based study to demonstrate the clinical utility and safety of a model-based patient-tailored dose regimen of vancomycin in neonates. The results from population pharmacokinetic study were successfully integrated in neonatal clinical practice to individualise vancomycin therapy.
    Type of Medium: Online Resource
    ISSN: 0003-9888 , 1468-2044
    Language: English
    Publisher: BMJ
    Publication Date: 2016
    detail.hit.zdb_id: 1481191-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Archives of Disease in Childhood, BMJ, Vol. 101, No. 1 ( 2016-01), p. e1.41-e1
    Abstract: Children with haematological malignancy represent an identified subgroup of the paediatric population with specific pharmacokinetic parameters. In these patients, inadequate empirical antibacterial therapy may result in infection-related morbidity and increased mortality, making optimization of the dosing regimen essential. As paediatric data are limited, our aim was to evaluate the population pharmacokinetics of teicoplanin in order to define the appropriate dosing regimen in this high-risk population. Methods The current dose of teicoplanin was evaluated in children with haematological malignancy. Population pharmacokinetics of teicoplanin was analysed using NONMEM software. The dosing regimen was optimised based on the final model. Results Eighty-five children (age range: 0.5 to 16.9 years) were included. Therapeutic drug monitoring and opportunistic samples (n=143) were available for analysis. With the current recommended dose of 10 mg/kg/day, 41 children (48%) had sub-therapeutic steady-state trough concentrations (Css,min 〈 10 mg/liter). A two-compartment pharmacokinetic model with first-order elimination was developed. Systematic covariate analysis identified that bodyweight (size) and creatinine clearance significantly influenced teicoplanin clearance. The model was validated internally. Its predictive performance was further confirmed in an external validation. In order to reach the target AUC of 750 mg·h/L, 18 mg/kg was required for infants, 14 mg/kg for children and 12 mg/kg for adolescents. A patient-tailored dose regimen was further developed and reduced variability in AUC and Css,min values compared to the mg/kg-basis dose, making the modelling approach an important tool for dosing individualization. Conclusions This first population pharmacokinetic study of teicoplanin in children with haematological malignancy provided evidence-based support to individualize teicoplanin therapy in this vulnerable population.
    Type of Medium: Online Resource
    ISSN: 0003-9888 , 1468-2044
    Language: English
    Publisher: BMJ
    Publication Date: 2016
    detail.hit.zdb_id: 1481191-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Georg Thieme Verlag KG ; 2018
    In:  American Journal of Perinatology Vol. 35, No. 06 ( 2018-05), p. 589-598
    In: American Journal of Perinatology, Georg Thieme Verlag KG, Vol. 35, No. 06 ( 2018-05), p. 589-598
    Abstract: Background Multicenter neonatal clinical trials aim to provide evidence-based drug evaluation, but recruiting neonates requires collaboration, standard procedures, and trained neonatologists. Methods A questionnaire based on a previous Delphi study was sent to European neonatal intensive care units (NICUs) to collect their research experience and identify areas for improvement. Results Of 247 NICUs,79 (32%) responded: 69 were level III units and 10 were level II units. In level III centers, 62% had medical staff dedicated to research and 65% conducted regular in-house audits. Similarities were observed in the median number of trials per year (level II: 2; level III: 5), Good Clinical Practice training (level II: 78%; level III: 66%), and standard operating procedures (level II: 63%; level III: 71%). Most NICUs had access to scientific advice for trial design, conduct, data management, and regulatory aspects. Involvement of patient advocacy groups was more common in level II units (level II: 75%; level III: 59%). A “quality” score of 34 “quality” research items was calculated for all centers (mean: 23.2 ± 6.2; range: 6–34). Conclusion Research experience and processes vary across Europe. Harmonizing research practices and setting standards will allow building a European neonatal network for effective, safe, and quality neonatal drug development.
    Type of Medium: Online Resource
    ISSN: 0735-1631 , 1098-8785
    Language: English
    Publisher: Georg Thieme Verlag KG
    Publication Date: 2018
    detail.hit.zdb_id: 2042426-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Informa UK Limited ; 2015
    In:  Expert Review of Clinical Pharmacology Vol. 8, No. 5 ( 2015-09-03), p. 635-648
    In: Expert Review of Clinical Pharmacology, Informa UK Limited, Vol. 8, No. 5 ( 2015-09-03), p. 635-648
    Type of Medium: Online Resource
    ISSN: 1751-2433 , 1751-2441
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2015
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 130, No. Suppl_1 ( 2017-12-07), p. 975-975
    Abstract: Background: The pharmacokinetic profile of most drugs is dependent on patient's covariates and may be influenced by the disease. Cefotaxime is frequently prescribed in paediatric patients with Sickle Cell Disease (SCD), characterized by vaso-occlusive complications, chronic haemolytic anaemia and defective immunological function predisposing to severe infection. Considering the very particular inflammatory context and the complexity of the disease, one may suspect than SCD could influence the pharmacokinetic profile of many drugs, which has already been reported for the morphine in SCD patients. Data on the impact of SCD on Cefotaxime disposition are missing, while the prescription of an optimal treatment against potentially fatal infections is essential. In the present study, our aims were to determine Cefotaxime pharmacokinetics when prescribed in SCD children for suspected or proven bacterial infection, identify significant covariates and perform Monte-Carlo simulations to optimize drug dosage. Results: Cefotaxime serum concentrations were measured in 80 paediatric SCD patients receiving Cefotaxime. A total of 110 concentrations were available for pharmacokinetic analysis. The population pharmacokinetic model developed from the SCD samples analyze made it possible to predict the plasma concentrations obtained according to the prescribed dosages. We found that the prescription of a standard dose of Cefotaxime (50 mg/kg every 8 hours) or a high dose of Cefotaxime (50 mg/kg every 6 hours) were both insufficient to reach the therapeutic target (ie a time above the minimum inhibitory concentration of the germ (T & gt;MIC) of 80%, which is recommended for severe bacterial infection), particularly in young children (Figure 1) . With the standard dose of Cefotaxime, only 38% of patients under 12 years old reached the therapeutic target, which increase to 82% with a high dose, meaning that 18% of the patients are still not efficiently treated in case of bacteria infection. In case of germs (Streptococcus pneumoniae or Salmonella) with intermediate sensitivity (MCI=1g/l), the amount of underdosed patients could reach 81% with the prescription of a standard dose (Figure 1). Moreover, Cefotaxime clearance increased by 22% in patients with acute chest syndrome, highlighting the possible impact of severe vaso-occlusion and inflammation on drug metabolism. Conclusion: Our data suggest that SCD patients, particularly young children, may have a particular Cefotaxime pharmacokinetic profile, possibly due to severe inflammatory condition. Considering the predisposition to severe infection in these patients, the prescribed dose of Cefotaxime should not be lower than 50 mg/kg/6h. In case of acute chest syndrome, this dose could even be insufficient to treat a bacterial infection (including due to sensitive germ with MIC= 0.5g/L or below) and should be adapted depending on patients' characteristics and clinical presentation. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2017
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Antimicrobial Agents and Chemotherapy, American Society for Microbiology, Vol. 62, No. 9 ( 2018-09)
    Abstract: Azithromycin is extensively used in children with community-acquired pneumonia (CAP). Currently, the intravenous azithromycin is used off-label in children partly due to lacking of pharmacokinetic data. Our objective was to evaluate the population pharmacokinetics (PPK) and optimize dose strategy in order to improve treatment in this distinctive population. This was a prospective, multicenter, open-labeled pharmacokinetic study. Blood samples were collected from hospitalized pediatric patients and concentrations were determined by liquid chromatography-tandem mass spectrometry (LC-MS/MS). PPK analysis was conducted using NONMEM software. The pharmacokinetic data from 95 pediatric patients (age range, 2.1 to 11.7 years) were available for analysis. The PPK was best fitted by a two-compartment model with linear elimination. Covariate analysis verified that body weight and alanine aminotransferase (ALT) had significant effects on azithromycin pharmacokinetics, yielding a 24% decrease of clearance in patients with ALT of 〉 40. Monte Carlo simulation showed that for children with normal liver function, a loading-dose strategy (a loading dose of 15 mg/kg of body weight followed by maintenance doses of 10 mg/kg) would achieve the ratio of the area under free drug plasma concentration-time curve over 24 h ( f AUC) to MIC 90 ( f AUC/MIC) target of 3 h in 53.2% of hypothetical patients, using a normative MIC susceptibility breakpoint of 2 mg/liter. For children with ALT of 〉 40, the proposed dose needed to decrease by 15% to achieve comparable exposure. The corresponding risk of overdose for the recommended dosing regimen was less than 5.8%. In conclusion, the PPK of azithromycin was evaluated in children with CAP and an optimal dosing regimen was constructed based on developmental pharmacokinetic-pharmacodynamic modeling and simulation.
    Type of Medium: Online Resource
    ISSN: 0066-4804 , 1098-6596
    RVK:
    Language: English
    Publisher: American Society for Microbiology
    Publication Date: 2018
    detail.hit.zdb_id: 1496156-8
    SSG: 12
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Society for Microbiology ; 2018
    In:  Antimicrobial Agents and Chemotherapy Vol. 62, No. 4 ( 2018-04)
    In: Antimicrobial Agents and Chemotherapy, American Society for Microbiology, Vol. 62, No. 4 ( 2018-04)
    Abstract: Cefotaxime is the first-line treatment for meningitis in neonates and young infants. However, limited data on cefotaxime cerebrospinal fluid (CSF) concentrations in neonates and young infants were available. The aim of the present study was to evaluate the penetration of cefotaxime into CSF in neonates and young infants. Blood and CSF samples were collected from neonates and young infants treated with cefotaxime using an opportunistic pharmacokinetic sampling strategy, and concentrations were quantified by high-performance liquid chromatography-tandem mass spectrometry. The analysis was performed using NONMEM and R software. Thirty neonates and young infants (postmenstrual age range, 25.4 to 47.4 weeks) were included. A total of 67 plasma samples and 30 CSF samples were available for analysis. Cefotaxime plasma and CSF concentrations ranged from 2.30 to 175.42 mg/liter and from 0.39 to 25.38 mg/liter, respectively. The median ratio of the CSF concentration to the plasma concentration was 0.28 (range, 0.06 to 0.76). Monte Carlo simulation demonstrated that 88.4% and 63.9% of hypothetical neonates treated with 50 mg/kg of body weight three times a day (TID) would reach the pharmacodynamic target (the percentage of the dosing interval that the free antimicrobial drug concentration remains above the MIC, 70%) using the standard EUCAST MIC susceptibility breakpoints of 2 mg/liter and 4 mg/liter, respectively. The penetration of cefotaxime into the CSF of neonates and young infants was evaluated using an opportunistic sampling approach. A dosage regimen of 50 mg/kg TID could cover the most causative pathogens with MICs of 〈 2 mg/liter. Individual dosage adaptation was required for more resistant bacterial strains, such as Staphylococcus aureus .
    Type of Medium: Online Resource
    ISSN: 0066-4804 , 1098-6596
    RVK:
    Language: English
    Publisher: American Society for Microbiology
    Publication Date: 2018
    detail.hit.zdb_id: 1496156-8
    SSG: 12
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    American Society for Microbiology ; 2016
    In:  Antimicrobial Agents and Chemotherapy Vol. 60, No. 11 ( 2016-11), p. 6626-6634
    In: Antimicrobial Agents and Chemotherapy, American Society for Microbiology, Vol. 60, No. 11 ( 2016-11), p. 6626-6634
    Abstract: Cefotaxime is one of the most frequently prescribed antibiotics for the treatment of Gram-negative bacterial sepsis in neonates. However, the dosing regimens routinely used in clinical practice vary considerably. The objective of the present study was to conduct a population pharmacokinetic study of cefotaxime in neonates and young infants in order to evaluate and optimize the dosing regimen. An opportunistic sampling strategy combined with population pharmacokinetic analysis using NONMEM software was performed. Cefotaxime concentrations were measured by high-performance liquid chromatography-tandem mass spectrometry. Developmental pharmacokinetics-pharmacodynamics, the microbiological pathogens, and safety aspects were taken into account to optimize the dose. The pharmacokinetic data from 100 neonates (gestational age [GA] range, 23 to 42 weeks) were modeled with an allometric two-compartment model with first-order elimination. The median values for clearance and the volume of distribution at steady state were 0.12 liter/h/kg of body weight and 0.64 liter/kg, respectively. The covariate analysis showed that current weight, GA, and postnatal age (PNA) had significant impacts on cefotaxime pharmacokinetics. Monte Carlo simulations demonstrated that the current dose recommendations underdosed older newborns. A model-based dosing regimen of 50 mg/kg twice a day to four times a day, according to GA and PNA, was established. The associated risk of overdose for the proposed dosing regimen was 0.01%. We determined the population pharmacokinetics of cefotaxime and established a model-based dosing regimen to optimize treatment for neonates and young infants.
    Type of Medium: Online Resource
    ISSN: 0066-4804 , 1098-6596
    RVK:
    Language: English
    Publisher: American Society for Microbiology
    Publication Date: 2016
    detail.hit.zdb_id: 1496156-8
    SSG: 12
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages