review article

Precision of Pharmacokinetic Parameters in Children: A Systematic Review

Melany Gaetani1,2,4,5, George Tomlinson6,7, Baseer Yasseen2, Elizabeth Uleryk8, Christopher Parshuram1-6,9,10*

1Department of Critical Care Medicine, The Research Institute, Hospital for Sick Children, Ontario, Canada

2Center for Safety Research, The Research Institute, Hospital for Sick Children, Ontario, Canada

3Child Health Evaluative Sciences, The Research Institute, Hospital for Sick Children, Ontario, Canada

4Department of Paediatrics, University of Toronto, Ontario, Canada

5Interdepartmental Division of Critical Care Medicine, University of Toronto, Ontario, Canada

6Institute of Health Policy Management and Evaluation, University of Toronto, Ontario, Canada

7Department of Medicine, University Health Network and Mt Sinai Hospital, Ontario, Canada

8The hospital for Sick Children, Hospital Library, University of Toronto, Ontario, Canada

9Center for Quality Improvement and Patient Safety, University of Toronto, Ontario, Canada

10Faculty of Medicine, University of Toronto, Ontario, Canada

*Corresponding author: Christopher Parshuram, Department of Critical Care Medicine, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada

Received Date: 30 January, 2020; Accepted Date: 13 February, 2020; Published Date: 18 February, 2020

Citation: Gaetani M, Tomlinson G, Yasseen B, Uleryk E, Parshuram C (2020) Precision of Pharmacokinetic Parameters in Children: A Systematic Review. J Hosp Health Care Admin 4: 131. DOI: 10.29011/2688-6472.000031

Abstract

The purpose of paediatric pharmacokinetic studies is to provide precise estimates of the true value of pharmacokinetic parameters to inform dosing. We evaluated age-specific estimates of pharmacokinetic parameters in peer-reviewed studies. Systematic reviews of 10 drugs included publications describing >1 pharmacokinetic study in patients <18 years. A pharmacokinetic study was defined as peer-reviewed primary data used to estimate one or more of Volume of distribution (Vd) or Clearance (CL). We defined uncertainty as the width of the 95% confidence interval divided by the population estimate of the study mean. Acceptable uncertainty as uncertainty was defined as <20%. Review of 21594 abstracts identified 137 eligible manuscripts containing 203 pharmacokinetic studies that included 176 Vd and 178 CL parameter estimates. Pharmacokinetic parameter estimates were derived from 2 or more age groups in 227 (64%) estimates. Acceptable uncertainty was found in 59 (34%) Vd and 35 (20%) CL, estimates. Heterogeneity was found with wide prediction intervals reported by drug and parameter. There was limited literature to support allometric dosing in the ten commonly used drugs we studied. There are multiple factors that may contribute to variability. We explore the importance of precise pharmacokinetic data, utility of parameter estimate ranges and the feasibility and extrapolation of pharmacokinetic parameter estimates. Factors contributing to observed uncertainty included modest sample sizes, within sample heterogeneity and dosing that accounts for patient factors in addition to age. These present opportunities to improve the quality and utility of pharmacokinetic data that underpins paediatric dosing.

Introduction

The success of drug therapy is contingent upon administration of the optimal dose at the optimal dosing interval. Dose and frequency recommendations are based on pharmacokinetic parameter estimates derived from pharmacokinetic studies from relevant populations [1-3]. Systematic error in dosing may occur if recommendations are based on estimates that are significantly different from the true population mean. In turn, routine sub-therapeutic or supra-therapeutic dosing will lead to biased estimates of drug effectiveness and toxicity, and undermine the value of paediatric drug therapy.

As with other estimates of ‘true’ population values, the precision with which population pharmacokinetic parameters are known can be assessed by the 95% confidence interval of the mean [4]. This interval includes the most plausible values of the mean for the population represented by the sample.

Confidence intervals for population values can be calculated in individual pharmacokinetic studies, or, if studies are sufficiently homogeneous, by combining similar pharmacokinetic studies using meta-analysis. Parameters known with greater precision will have narrower 95% confidence intervals and thus can more precisely inform dosing recommendations. Presently there is no recommended size for a confidence interval of pharmacokinetic parameter. By proxy, industry standards for pharmacotherapeutic precision suggest that variations of less than 10% are acceptable for preparation and administration [5].

The objectives of this study were to estimate average values of pharmacokinetic parameters in children reported in peer-reviewed publications, to assess the degree of heterogeneity between studies of similar populations and report ranges of pharmacokinetic parameters for the 10 most commonly used drugs in the ICU.

Materials and Methods

We performed a systematic review and meta-analysis of paediatric pharmacokinetic studies of ten drugs commonly given to children: acetaminophen, dopamine, fentanyl, furosemide, gentamicin, lorazepam, midazolam, morphine, ranitidine and vancomycin, were selected as they are commonly administered to critically ill children [6]. The main outcome was the precision of the population estimates of the pharmacokinetic parameters for volume of distribution and clearance. Precision was operationalized as the uncertainty of each estimate, based on our previous work [7-9] and USP standards for intravenous preparations [5]. Acceptable uncertainty was defined as where the margins of the 95% confidence interval for the population mean were within 10% of the study mean. Greater uncertainty was quantified by use of >10-50% and >50% categories.

Search Strategy

Searches were constructed and performed by an academic librarian (EU), using MEDLINE (1966 – December 2018) and EMBASE (1980 – December 2018) databases. A three-tier strategy was used. First, alternate generic, brand and alternative names for each drug were identified using MEDLINE (scope notes), EMBASE (CAS registry number), e-CPS, and STAT! Ref. These comprehensive lists of drug name alternatives were used as keywords. Second, the selection was restricted to paediatric pharmacokinetic studies using the “limit” function in search criteria (18 years or less) and keywords including “neonate, infant, child, adolescent, and pediatric”. Third, the literature search was narrowed to pharmacokinetic studies using keywords including “pharmacokinetics, kinetics, volume of distribution, clearance”.

Study identification, eligibility and data abstraction

Eligible publications described at least one pharmacokinetic study in patients aged 18 years and younger. An eligible pharmacokinetic study was defined as the use of primary data from intravenous drug administration, to a group of paediatric patients to estimate one or more of the pharmacokinetic parameters - clearance, and volume of distribution. Pharmacokinetic parameter estimates had to be reported in a way that was amenable to statistical analysis and not normalized to adult weight. One publication could include more than one pharmacokinetic study.

Within each pharmacokinetic study we abstracted 5 main items. First, the age group or age groups of the patients evaluated. We predefined 6 specific age groups; Neonatal unspecified (birth - 28 days post-natal age), premature neonates (<36 weeks gestational age), term neonates (36 weeks gestational age -28 days post-natal age), infant (age >28 days – 2 years), child (age 2 years – 12 years) and adolescent (age >12 years – 18 years) similar to previous recommendations for paediatric drug studies [10]. To describe studies where age was incompletely specified we included a generic group for studies with an unspecified paediatric (age <18 years). Second, pharmacokinetic parameter estimates were abstracted and converted into common units for each of volume of distribution (L/kg) and clearance (L/kg/hr). Third, the standard deviation of the estimate was abstracted or calculated from either raw data or the standard error in the report [4]. Finally, the number of age groups studied and the sample size of each age group was recorded. The sample size of a pharmacokinetic study was defined as the sample size of the parameter with the greatest number of subjects.

An academic librarian (EU) conducted the searches, and exported results into Mendeley (Elsevier, Amsterdam, Netherlands). Two members of the study team (MG, BY), independently reviewed titles and abstracts to identify ineligible studies and then reviewed the remaining publications in full to identify eligible studies and abstract data. Inconsistent data were reviewed by a third person (CP) and differences resolved by consensus. The percentage agreement was calculated for study inclusion and the abstracted parameter estimates.

Data management and analysis

There were two types of studies: (1) those estimating parameters on each individual and reporting the mean and standard deviation across individuals; and (2) those using a single hierarchical model that included all individuals to estimate population parameters and between-individual heterogeneity. These studies both estimate the same underlying quantities, but by different means, so to pool them, we used the following approaches: In the first type of study, the sample mean, standard deviation and number of individuals in the sample were abstracted. In the second type of study, the population estimate and the estimated between-individual standard deviation (as reported directly or estimated from the coefficient of variation) and number of individuals were abstracted.

For each study, using the published sample size and the published or calculated between individual standard deviation of the parameter value, and, assuming a normal distribution for each estimate around its mean with standard error equal to SD/sqrt(n), we calculated the 95% confidence interval for the mean value of each pharmacokinetic parameter, For each parameter, we calculated the uncertainty of the estimate by relating the width of the 95% confidence interval to the point estimate using the following formula:

Uncertainty of estimate = 

Standardizing the imprecision by the estimated population mean reflects the effect of potential variations in the true value of the mean on dosing recommendations. For example, if the true value of the volume of distribution for a drug was 30% more than the population estimate from a given study, then dosing recommendation may result in routine under-dosing by 30% and a conclusion that therapy is less effective than when it is optimally dosed. Acceptable uncertainty was defined as variation of 20%; this corresponds to the ends of the confidence interval being 10% higher and 10% lower than the estimated mean.

Next, we evaluated heterogeneity between study-specific estimates of parameters. Using a random effects meta-analysis model, with weights equal to the reciprocals of the variance of each estimate (i.e., the reciprocal of the standard error squared), we calculated the pooled population estimate and its 95% confidence interval for each parameter, first within each age group, and following exploration of the data, across age groups. Graphical presentation shows point-estimates for the pooled values of PK parameters and the 95% confidence intervals for the mean for meta-analyses of studies of the same drug in similar populations. The I2 value was used to summarize the extent of heterogeneity and a 95% prediction interval was added to the figure to facilitate interpretation of between-study variation in the estimated values of PK parameters. In this study the prediction interval covers the range of likely values for the PK value in a new study with characteristics similar to the pre-existing studies [11]. Research Ethics Board approval was not required.

Results

Review of 21594 publications identified from the MEDLINE and EMBASE searches resulted in review of 641 full publications, and inclusion of 138 eligible publications [12-150], of 11,154 patients that described 203 pharmacokinetic studies, and described estimates for mean values of 178 clearances and 176 volume of distribution parameter estimates (Figure 1).

There were 26 (19%) manuscripts published in the 1980s, 41 (30%) in the 1990s, 17 (12%) were published in the 2000s and 54 (39%) were published after 2010. All 10 preselected drugs had been evaluated. Each drug had a median (IQR) of 6 (5-14) pharmacokinetic studies, ranging from 3 (2%) studies for lorazepam to 49 (36%) studies for vancomycin. Parameter estimates using age-specific data were reported for 5 drugs in premature neonates, 5 drugs in term neonates, 3 drugs in infants, 3 drugs in children and 2 drugs in adolescents. Of 354 parameter estimates 220 (62%) were for gentamicin or vancomycin (Table 1).

Pharmacokinetic Study Characteristics

The ages of children studied and corresponding sample sizes were reported in all 203 (100%) pharmacokinetic studies. Diagnoses and baseline medical conditions among patients from all studies were common and expected given age groups, clinical setting and administered medications. Pharmacokinetic parameter estimates were derived from a single age group in 141 (40%) estimates from patients from 2 age groups in 43 (12%) parameter estimates, and from 3 or more age groups in 40 (11%) estimates. Fifty-five (16%) parameter estimates described their population as being from all ‘paediatric’ age ranges and in 75 (21%) estimates included patients classified only as ‘neonatal’ without specification of gestational age. The median (IQR) sample size was 19 (10-39). Ten (5%) studies had fewer than 5 participants, 112 (55%) studies had 20 or fewer, and 28 (10%) had 100 or more participants (Figure 2).

Pharmacokinetic parameter estimates of population value

Volume of distribution was reported in 176 (87%) studies. There was a median (IQR) uncertainty of 28 (16-48)% and greater than 20% uncertainty was present in 117 (66%) estimates. Clearance was reported in 178 (88%) studies. The median (IQR) uncertainty was 43 (25-72)% and greater than 20% uncertainty was found in 143 (80%) studies. There was significant heterogeneity found across all drugs in both volumes of distribution and clearance estimates (Figure 3).

The precision of PK parameter estimates by drug and age

The largest heterogeneity was found within the pharmacokinetic parameter estimates for dopamine clearance. The smallest heterogeneity was found within estimates for clearance of lorazepam (Figure 4). A large degree of heterogeneity was still found within studies with single age groups by drug (Supplementary Material Figure 1, Table 1).

Discussion

This systematic review was conducted to identify peer-reviewed publications of primary data describing paediatric pharmacokinetic parameters in 10 of the most commonly administered drugs in critically ill children. The main findings relate to the wide range of precision parameter estimates factors that may contribute to the observed precision, the importance of precise pharmacokinetic data and utility of parameter estimate ranges, and the feasibility and extrapolation of pharmacokinetic parameter estimates.

First, we found limited literature to guide allometric dosing - the predominant method used for most drug dosing recommendations - in the ten commonly used drugs we studied. For both volumes of distribution and clearance parameters, there was significant uncertainty within studies and considerable heterogeneity between studies. Prediction intervals for the parameters for the next study of the same drugs in the same age – in purportedly similar patients - were wide. Variations from paediatric dosing recommendations have been reported as ‘routine’ by frontline staff in a large paediatric hospital [151], they may reflect the limited utility, availability and scientific grounding of current paediatric dosing recommendations [152,153]. The majority of patients studied 9908 (89%) and studies 82 (59%) included were of vancomycin and gentamycin. Therapeutic levels are frequently measured for these two drugs reflecting the limited expectations of pharmacokinetic precision in practice. Studies incorporating patient level data in these, and other drugs, highlight the need for ongoing research.

Second, we found the heterogeneity between studies of the same drug in the same age group was similar to the heterogeneity between age groups. One interpretation of this finding is that physiologic changes associated with age [154,155], are less important than the other physiologic changes occurring in the patients studied receiving drugs used in critically ill children. Another interpretation is that relative contributions of inter-individual differences to drug metabolism and excretion [156-158], may be greater than those of either age or underlying disease processes. Precedents of approaches that combine data across age groups include the use of adult pharmacokinetic data in physiologically based pharmacokinetic models to calculate estimates for paediatric parameters. These calculated parameters had precision of +/-50% values [159], similar to the precision we found in primary studies. Other methods integrating other biologically relevant variables into multivariable pharmacokinetic models have demonstrated higher degrees of precision in model development and validation. These suggest that there is more precise prediction of levels for drugs such as midazolam [160], caffeine [161] and meropenem [162], than is currently extrapolated from pharmacokinetic data, and complement estimates derived from our meta-analyses.

Third, 141 parameters estimates were derived from single age groups that have an endorsed biologic basis [163], and quarter of studies had a sample size of ten or less. The addressable origins of variability include the specificity of definitions of populations - age and other conditions, the sample sizes supporting calculation of precise estimates. Robust knowledge of pharmacokinetic parameter estimates may be especially useful in instances where volume of distribution, and clearance would be altered clinically. Though these parameters are difficult to directly correlate to one another, understanding variations in each and the potential for impact on another parameter could be important. For example, if dosing for specific drugs were to be driven by clearance rather than volume of distribution, variance in the parameter estimates for volume of distribution becomes less impactful. These include instances with the use of technology such as renal replacement or extracorporeal membrane oxygenation where paediatric drug dosing guidelines and data may be scarce and pharmacokinetic parameters such as volume of distribution and clearance could be impacted both in general and in relation to each other [164,165].

Limitations

There are several potential limitations to this work. First, data on study quality factors was not analysed or reported in relation to the pharmacokinetic parameters studied. Second, we chose only 10 commonly used drugs in critically ill children. This sample is unlikely to be representative of drugs administered to children in other settings, however we believe it likely to accurately reflect the extent of peer-reviewed pharmacokinetic data for children in other medicines. Third, despite the development of a comprehensive strategy, by an academic librarian, this systematic review may have missed relevant pharmacokinetic studies. We included only English language articles, and chose a manual study selection approach [166], but excluded grey literature – including unpublished studies conducted by pharmaceutical companies, these may include relevant data that has not been subjected to the scrutiny of peer-review [167]. Finally, our analytic approach was conservative. By assuming parameters were normally distributed, we probably under-estimated the confidence intervals for each pharmacokinetic parameter, and thus our results may under-state the current true uncertainty of paediatric pharmacokinetic data.

Conclusions

In this systematic review of 10 commonly used drugs in critically ill children we found that heterogeneity in the estimates of pharmacokinetic parameters, with similar precision within and between age groups. Meta-analysis provides pooled estimates that could be used to provide a rational basis for therapy, however the prediction interval highlights opportunities for ongoing pharmacokinetic research, to improve our understanding of drug handling in the paediatric patient, about the potential value of dose titration and customized therapy.

Acknowledgement

This endeavour was not directly funded and the authors of this paper do not hold any commercial associations that might pose a conflict of interest in connection with the manuscript.

Author Contributions

Melany Gaetani contributed though acquisition, analysis, and interpretation of data as well as drafting of the manuscript and statistical analysis. She had full access to all of the data in the study and shares responsibility for the integrity of the data and the accuracy of the data analysis. George Tomlinson made substantial contributions to the intellectual content through conception, design and statistical analysis.

Baseer Yasseen and Elizabeth Uleryk contributed though acquisition of data. Christopher Parshuram contributed in the conceptualization of the study question, study design, and had full access to all of the data in the study and shares responsibility for the integrity of the data, the accuracy of the data analysis, revision of the manuscript for important intellectual content.

Conflicts of Interest

The authors declare no conflicts of interest.


Figure 1: Systematic review process of peer reviewed publications. The process of systematic review is depicted above. A total of 138 publications were included which identified 203 pharmacokinetic studies across the 2 parameters.  Studies were excluded if not in the English language, not within the paediatric age group (<18 years), if the reported pharmacokinetic parameters were not amenable to data analysis, or the medications was administered intravenously.



Figure 2: Sample size of Pharmacokinetic studies. Sample sizes from the 203 pharmacokinetic studies evaluated. The median (IQR) sample size was 19 (10-39). Ten (5%) studies had fewer than 5 participants, 112 (55%) studies had 20 or fewer, and 28 (10%) had 100 or more participants.



Figure 3: Uncertainty of pharmacokinetic parameter estimates by drug and parameter. Uncertainty of clearance and volume of distribution estimates from 138 peer-reviewed studies. The dark dots represent studies where the uncertainty was <20% by parameter and by drug. The red dots represent those studies where the uncertainty of the population estimate was >20%. The percentage beside each drug is the proportion of studies that fell within the predefined acceptable uncertainty range of <20%.



Figure 4: Pooled pharmacokinetic parameter estimates by drug. Pooled estimates of each pharmacokinetic parameter by drug are demonstrated. The blue bars represent the estimated mean and confidence interval for the mean of parameter by drug. The pink overlying bars represent prediction intervals demonstrating the heterogeneity found within each estimate. The number beside each parameter estimate by drug represents the number of pharmacokinetic parameter estimates that were included in each prediction interval. 

Drug name

Pharmacokinetic Studies

Pharmacokinetic parameter estimates

Number calculated exclusively from children from single age group

Total evaluated

 

All

Single age group

Preterm

Term

Infant

Child

Adol.

 

N(col. %)

N(col. %)

Acetaminophen

6

3

3

2

0

0

0

11

Dopamine

6

3

0

0

0

0

0

7

Fentanyl

9

5

0

0

0

0

0

17

Furosemide

5

4

2

0

0

2

0

9

Gentamicin

56

43

22

10

2

7

4

93

Lorazepam

4

2

0

0

0

0

4

7

Midazolam

19

8

0

0

0

6

0

36

Morphine

24

21

20

7

3

0

0

38

Ranitidine

5

3

0

4

0

0

0

9

Vancomycin

69

35

18

4

11

8

2

127

 

 

 

 

 

 

 

 

 

Total

203

127

65

27

16

23

10

354


Table 1: Pharmacokinetic Parameter studies reviewed by drug. We describe the studies included in this systematic review of peer reviewed articles by drug. The 138 articles identified provided 203 individual pharmacokinetic studies for a total of 354 parameter estimates. 141 (40%) of pharmacokinetic parameter estimates were from a single age group studied. Studies that included unspecified neonatal or paediatric age groups were not considered a single age group. ‘Adol’ represents the adolescent age group.

References

  1. Kearns GL, Abdel-Rahman SM, Alander SW, Blowey DL, Leeder JS, et al. (2003) Developmental pharmacology-drug disposition, action, and therapy in infants and children. N Engl J Med 349: 1157-1167.
  2. Yokoi T (2009) Essentials for starting a pediatric clinical study (1): Pharmacokinetics in children. J Toxicol Sci 34: SP307-312.
  3. Anderson GD (2010) Developmental pharmacokinetics. Semin Pediatr Neurol 17: 208-213.
  4. Feinstein AR, Concato J (1998) The quest for "power": contradictory hypotheses and inflated sample sizes. J Clin Epidemiol 51: 537-545.
  5. The United States Pharmacopeia : USP 24 : the National Formulary : NF 19 : by authority of the United States Pharmacopoeial Convention, Inc., meeting at Washington, D.C., March 9-12, 1995 ; prepared by the Committee of Revision and published by the Board of Trustees; United States Pharmacopeial Convention: Rockville, 1999.
  6. McDonnell C, Hum S, Frndova H, Parshuram CS (2009) Pharmacotherapy in Pediatric Critical Illness. Pediatric Drugs 11: 323-331.
  7. Parshuram CS, Ng GYT, Ho TKL, Klein J, Moore AM et al. (2003) Discrepancies between ordered and delivered concentrations of opiate infusions in critical care. Crit Care Med 31: 2483-2487.
  8. Parshuram CS, Dupuis LL, To T, Weitzman SS, Koren G, et al. (2006) Occurrence and impact of unanticipated variation in intravenous methotrexate dosing. Ann Pharmacother 40: 805-811.
  9. Parshuram CS, To T, Seto W, Trope A, Koren G, et al. (2008) Systematic evaluation of errors occurring during the preparation of intravenous medication. Cmaj 178: 42-48.
  10. Research, CfDEa (2019) Data Standards Manual (monographs) - Pediatric Exclusivity Study Age Group.
  11. Stanley D (2016) Prediction Interval Functions for Assessing Replication Study Results.
  12. Frymoyer A, Meng L, Bonifacio SL, Verotta D, Guglielmo BJ (2013) Gentamicin pharmacokinetics and dosing in neonates with hypoxic ischemic encephalopathy receiving hypothermia. Pharmacotherapy 33: 718-726.
  13. Abdel Hadi O, Al Omar S, Nazer LH, Mubarak S, Le J (2016) Vancomycin pharmacokinetics and predicted dosage requirements in pediatric cancer patients. J Oncol Pharm Pract 22: 448-453.
  14. Ali AS, Farouq MF, Al-Faify KA (2012) Pharmacokinetic approach for optimizing gentamicin use in neonates during the first week of life. Indian J Pharmacol 44: 36-40.
  15. Allegaert K, Van der Marel CD, Debeer A, Pluim MAL, Van Lingen RA, et al. (2004) Pharmacokinetics of single dose intravenous propacetamol in neonates: effect of gestational age. Arch Dis Child Fetal Neonatal Ed 89: F25-F28.
  16. Amaker RD, DiPiro JT, Bhatia J (1996) Pharmacokinetics of vancomycin in critically ill infants undergoing extracorporeal membrane oxygenation. Antimicrobial agents and chemotherapy 40: 1139-1142.
  17. Anderson BJ, Woollard GA, Holford NHG (2000) A model for size and age changes in the pharmacokinetics of paracetamol in neonates, infants and children. Br J Clin Pharmacol 50: 125-134.
  18. Anderson R, Huang Y (1992) Fatty acids are precursors of alkylamines in Deinococcus radiodurans. J Bacteriol 174: 7168-7173.
  19. Asbury WH, Darsey EH, Rose WB, Murphy JE, Buffington DE, et al. (1993) Vancomycin pharmacokinetics in neonates and infants: a retrospective evaluation. Ann Pharmacother 27: 490-496.
  20. Asseff IL, Gaucin GB, Olguin HJ, Najera JAG, Lopez AT, et al. (2016) Pharmacokinetics of ranitidine in preterm and term neonates with gastroesophageal reflux. BMC pediatrics 16: 90-90.
  21. Autret E, Dutertre JP, Breteau M, Jonville AP, Furet Y, et al. (1993) Pharmacokinetics of paracetamol in the neonate and infant after administration of propacetamol chlorhydrate. Dev Pharmacol Ther 20: 129-134.
  22. Mohammed BS, Engelhardt T, Cameron GA, Cameron L, Hawksworth GM, et al. (2012) Population pharmacokinetics of single-dose intravenous paracetamol in children. Br J Anaesth 108: 823-829.
  23. Barrett DA, Barker DP, Rutter N, Pawula M, Shaw PN (1996) Morphine, morphine-6-glucuronide and morphine-3-glucuronide pharmacokinetics in newborn infants receiving diamorphine infusions. Br J Clin Pharmacol 41: 531-537.
  24. Barrett DA, Elias-Jones AC, Rutter N, Shaw PN, Davis SS (1991) Morphine kinetics after diamorphine infusion in premature neonates. Br J Clin Pharmacol 32: 31-37.
  25. Begg EJ, Vella-Brincat JWA, Robertshawe B, McMurtrie MJ, Kirkpatrick CMJ, et al. (2009) Eight years' experience of an extended-interval dosing protocol for gentamicin in neonates. J Antimicrob Chemother 63: 1043-1049.
  26. Benefield EC, Hagemann TM, Allen HC, Farmer K, Burton ME, et al. (2016) Vancomycin Dosing and Pharmacokinetics in Postoperative Pediatric Cardiothoracic Surgery Patients. J Pediatr Pharmacol Ther 21: 66-74.
  27. Bhat R, Chari G, Gulati A, Aldana O, Velamati R, et al. (1990) Pharmacokinetics of a single dose of morphine in preterm infants during the first week of life. J Pediatr 117: 477-481.
  28. Bhatt-Mehta V, Johnson CE, Schumacher RE (1992) Gentamicin pharmacokinetics in term neonates receiving extracorporeal membrane oxygenation. Pharmacotherapy 12: 28-32.
  29. Bhatt-Mehta V, Nahata MC, McClead RE, Menke JA (1991) Dopamine pharmacokinetics in critically ill newborn infants. European journal of clinical pharmacology 40: 593-597.
  30. Bialkowski S, Staatz CE, Clark J, Lawson R, Hennig S (2016) Gentamicin Pharmacokinetics and Monitoring in Pediatric Patients with Febrile Neutropenia. Ther Drug Monit 38: 693-698.
  31. Blumer JL, Rothstein FC, Kaplan BS, Yamashita TS, Eshelman FN, et al. (1985) Pharmacokinetic determination of ranitidine pharmacodynamics in pediatric ulcer disease. J Pediatr 107: 301-306.
  32. Buck ML (1998) Vancomycin pharmacokinetics in neonates receiving extracorporeal membrane oxygenation. Pharmacotherapy 18: 1082-1086.
  33. Burtin P, Jacqz-Aigrain E, Girard P, Lenclen R, Magny JF, et al. (1994) Population pharmacokinetics of midazolam in neonates. Clinical Pharmacology and Therapeutics 56: 615-625.
  34. Caceres Guido P, Travaglianti M, Castro G, Licciardone N, Ferreyra O, et al. (2015) Population Pharmacokinetic Study of Vancomycin in Preterm Neonates 34: 124-124.
  35. Chang D (1995) Influence of malignancy on the pharmacokinetics of vancomycin in infants and children. Pediatr Infect Dis J 14: 667-673.
  36. Chang D, Liem L, Malogolowkin MA (1994) prospective study of vancomycin pharmacokinetics and dosage requirements in pediatric cancer patients. Pediatr Infect Dis J 13: 969-974.
  37. Chay PC, Duffy BJ, Walker JS (1992) Pharmacokinetic-pharmacodynamic relationships of morphine in neonates. Clin Pharmacol Ther 51: 334-342.
  38. Choi JJ, Moffett BS, McDade EJ, Palazzi DL (2011) Altered gentamicin serum concentrations in obese pediatric patients. Pediatr Infect Dis J 30: 347-349.
  39. Choonara I, Lawrence A, Michalkiewicz A, Bowhay A, Ratcliffe J (1992) Morphine metabolism in neonates and infants. Br J Clin Pharmacol 34: 434-437.
  40. Choonara IA, McKay P, Hain R, Rane A (1989) Morphine metabolism in children. Br J Clin Pharmacol 28: 599-604.
  41. Cies JJ, Habib T, Bains V, Young M, Menkiti OR (2018) Population Pharmacokinetics of Gentamicin in Neonates with Hypoxemic-Ischemic Encephalopathy Receiving Controlled Hypothermia. Pharmacotherapy 38: 1120-1129.
  42. Cies JJ, Moore WS, Nichols K, Knoderer CA, Carella DM, et al. (2017) Population Pharmacokinetics and Pharmacodynamic Target Attainment of Vancomycin in Neonates on Extracorporeal Life Support. Pediatr Crit Care Med 18: 977-985.
  43. Crom WR, Relling MV, Christensen ML, Rivera GK, Evans WE (1991) Age-related differences in hepatic drug clearance in children: studies with lorazepam and antipyrine. Clin Pharmacol Ther 50: 132-140.
  44. de Hoog M, Schoemaker RC, Mouton JW, van den Anker JN (2000) Vancomycin population pharmacokinetics in neonates. Clin Pharmacol Ther 67: 360-367.
  45. de Wildt SN, de Hoog M, Vinks AA, van der Giesen E, van den Anker JN (2003) Population pharmacokinetics and metabolism of midazolam in pediatric intensive care patients. Crit Care Med 31: 1952-1958.
  46. Demirjian A, Finkelstein Y, Nava-Ocampo A, Arnold A, Jones S, et al. (2013) A randomized controlled trial of a vancomycin loading dose in children. Pediatr Infect Dis J 32: 1217-1223.
  47. Eldadah MK, Schwartz PH, Harrison R, Newth CJ (1991) Pharmacokinetics of dopamine in infants and children. Crit Care Med 19: 1008-1011.
  48. Fontana M, Massironi E, Rossi A, Vaglia P, Gancia GP, et al. (1993) Ranitidine pharmacokinetics in newborn infants. Arch Dis Child 68: 602-603.
  49. Frattarelli DAC, Ergun H, Lulic-Botica M, Lehr VT, Aranda JV (2005) Vancomycin elimination in human infants with intrauterine growth retardation. Pediatr Infect Dis J 24: 979-983.
  50. Frymoyer A, Bonifacio SL, Drover DR, Su F, Wustoff CJ et al. (2017) Decreased Morphine Clearance in Neonates With Hypoxic Ischemic Encephalopathy Receiving Hypothermia. J Clin Pharmacol 57: 64-76.
  51. Frymoyer A, Hersh AL, El-Komy MH, Gaskari S, Su F, et al. (2014) Association between vancomycin trough concentration and area under the concentration-time curve in neonates. Antimicrob Agents Chemother 58: 6454-6461.
  52. Fuchs A, Guidi M, Giannoni E, Werner D, Buclin T, et al. (2014) Population pharmacokinetic study of gentamicin in a large cohort of premature and term neonates. Br J Clin Pharmacol 78: 1090-1101.
  53. Fullas F, Padomek MT, Thieman CJ, Van Gorp AE (2011) Comparative evaluation of six extended-interval gentamicin dosing regimens in premature and full-term neonates. Am J Health Syst Pharm 68: 52-56.
  54. García B, Barcia E, Pérez F, Molina IT (2006) Population pharmacokinetics of gentamicin in premature newborns. J Antimicrob Chemother 58: 372-379.
  55. Gauntlett IS, Fisher DM, Hertzka RE, Kuhls E, Spellman MJ, et al. (1988) Pharmacokinetics of fentanyl in neonatal humans and lambs: effects of age. Anesthesiology 69: 683-687.
  56. Geiduschek JM, Lynn AM, Bratton SL, Sanders JC, Levy FH, et al. (1997) Morphine pharmacokinetics during continuous infusion of morphine sulfate for infants receiving extracorporeal membrane oxygenation. Crit Care Med 25: 360-364.
  57. Germovsek E, Kent A, Metsvaht T, Lutsar I, Klein N, et al. (2016) Development and Evaluation of a Gentamicin Pharmacokinetic Model That Facilitates Opportunistic Gentamicin Therapeutic Drug Monitoring in Neonates and Infants. Antimicrob Agents Chemother 60: 4869-4877.
  58. Giachetto GA, Telechea HM, Speranza N, Oyarzun M, Nanni L, et al. (2011) Vancomycin pharmacokinetic-pharmacodynamic parameters to optimize dosage administration in critically ill children. Pediatr Crit Care Med 12: e250-254.
  59. Gomez DS, Campos EV, de Azevedo RP, Silva JMd, Ferreira MC, et al. (2013). Individualised vancomycin doses for paediatric burn patients to achieve PK/PD targets. Burns 39: 445-450.
  60. González-Martín G, Bravo I, Ibarra N, Arancibia A (1983) Clinical pharmacokinetics of furosemide in children with nephrotic syndrome. Int J Clin Pharmacol Ther Toxicol 21: 598-601.
  61. Gous AG, Dance MD, Lipman J, Luyt DK, Mathivha R, et al. (1995) Changes in vancomycin pharmacokinetics in critically ill infants. Anaesth Intensive Care 23: 678-682.
  62. Gray TM (1993) Gentamicin pharmacokinetics in term newborn twins. Clin Pharm 12: 615-616.
  63. Grimsley C, Thomson AH (1999) Pharmacokinetics and dose requirements of vancomycin in neonates. Archives of Disease in Childhood. Arch Dis Child Fetal Neonatal Ed 81: F221-F227.
  64. Guilhaumou R, Marsot A, Dupouey J, Galambrun C, Boulamery A, et al. (2016) Pediatric Patients With Solid or Hematological Tumor Disease: Vancomycin Population Pharmacokinetics and Dosage Optimization. Ther Drug Monit 38: 559-566.
  65. Hahn A, Frenck RW Jr, Zou Y, Vinks AA (2015) Validation of a pediatric population pharmacokinetic model for vancomycin. Ther Drug Monit 37: 413-416.
  66. Hartley R, Green M, Quinn M, Levene MI (1993) Pharmacokinetics of morphine infusion in premature neonates. Arch Dis Child 69: 55-58.
  67. Hatzopoulos FK, Stile-Calligaro IL, Rodvold KA, Sullivan-Bolyai J, Del Nido P, et al. (1993) Pharmacokinetics of intravenous vancomycin in pediatric cardiopulmonary bypass surgery. Pediatr Infect Dis J 12: 300-304.
  68. Hayani KC, Hatzopoulos FK, Frank AL, Thummala MR, Hantsch MJ, et al. (1997) Pharmacokinetics of once-daily dosing of gentamicin in neonates. The Journal of Pediatrics 131: 76-80.
  69. Hoff DS, Wilcox RA, Tollefson LM, Lipnik PG, Commers AR, et al. (2009) Pharmacokinetic outcomes of a simplified, weight-based, extended-interval gentamicin dosing protocol in critically ill neonates. Pharmacotherapy 29: 1297-1305.
  70. Inparajah M, Wong C, Sibbald C, Boodhan S, Atenafu EG, et al. (2010) Once-daily gentamicin dosing in children with febrile neutropenia resulting from antineoplastic therapy. Pharmacotherapy 30: 43-51.
  71. Neonatal vancomycin dosing on extracorporeal membrane oxygenation. Critical Care Medicine 38: A215-A215.
  72. Kim J, Walker SA, Iaboni DC, Walker SE, Elligsen M, et al. (2014) Determination of vancomycin pharmacokinetics in neonates to develop practical initial dosing recommendations. Antimicrobial Agents and Chemotherapy 58: 2830-2840.
  73. (1981) Fentanyl pharmacokinetics in children with heart disease. Anesthesiology 55: A194-A194.
  74. Chamberlain JM, Capparelli EV, Brown KM, Vance CW, Lillis K, et al. (2012) Pharmacokinetics of Intravenous Lorazepam in Pediatric Patients with and without Status Epilepticus. J Pediatr 160: 667-672.e662.
  75. Jacobson PA, West NJ, Price J, Hutchinson RJ (1997) Gentamicin and tobramycin pharmacokinetics in pediatric bone marrow transplant patients. Ann Pharmacother 31: 1127-1131.
  76. Jacqz-Aigrain E, Daoud P, Burtin P, Maherzi S, Beaufils F (1992) Pharmacokinetics of midazolam during continuous infusion in critically ill neonates. Eur J Clin Pharmacol 42: 329-332.
  77. Jacqz-Aigrain E, Wood C, Robieux I (1990) Pharmacokinetics of midazolam in critically ill neonates. Eur J Clin Pharmacol 39: 191-192.
  78. James A, Koren G, Milliken J, Soldin S, Prober C (1987) Vancomycin pharmacokinetics and dose recommendations for preterm infants. Antimicrobial Agents and Chemotherapy 31: 52-54.
  79. Jones RD, Chan K, Roulson CJ, Brown AG, Smith ID, et al. (1993) Pharmacokinetics of flumazenil and midazolam. Br J Anaesth 70: 286-292.
  80. Koehntop DE, Rodman JH, Brundage DM, Hegland MG, Buckley JJ (1986) Pharmacokinetics of fentanyl in neonates. Anesth Analg 65: 227-232.
  81. Koren G, Goresky G, Crean P, Klein J, MacLeod SM (1984) Pediatric fentanyl dosing based on pharmacokinetics during cardiac surgery. Anesth Analg 63: 577-582.
  82. Koren G, Goresky G, Crean P, Klein J, MacLeod SM (1986) Unexpected alterations in fentanyl pharmacokinetics in children undergoing cardiac surgery: age related or disease related? Dev Pharmacol Ther 9: 183-191.
  83. Krivoy N, Peleg S, Postovsky S, Ben Arush MW (1998) Pharmacokinetic analysis of vancomycin in steady state in pediatric cancer patients. Pediatr Hematol Oncol 15: 333-338.
  84. Lamarre P, Lebel D, Ducharme MP (2000) A population pharmacokinetic model for vancomycin in pediatric patients and its predictive value in a naive population. Antimicrob Agents Chemother 44: 278-282.
  85. Lares-Asseff I, Pérz-Guillé MG, Camacho Vieyra GA, Pérez AG, Peregrina NB, et al. (2016) Population Pharmacokinetics of Gentamicin in Mexican Children With Severe Malnutrition. Pediatr Infect Dis J 35: 872-878.
  86. Le J, Capparelli EV, Wahid U, Wu YSS, Romanowski GL, et al. (2015) Bayesian Estimation of Vancomycin Pharmacokinetics in Obese Children: Matched Case-Control Study. Clin Ther 37: 1340-1351.
  87. Le J, Ngu B, Bradley JS, Murray W, Nguyen A, et al. (2014) Vancomycin monitoring in children using bayesian estimation. Ther Drug Monit 36: 510-518.
  88. Le J, Ny P, Capparelli E, Lane J, Ngu B, et al. (2015) Pharmacodynamic Characteristics of Nephrotoxicity Associated With Vancomycin Use in Children. J Pediatric Infect Dis Soc 4: e109-116.
  89. Lingvall M, Reith D, Broadbent R (2005) The effect of sepsis upon gentamicin pharmacokinetics in neonates. Br J Clin Pharmacol 59: 54-61.
  90. Liu T, Deng C, Cheng D, Zhou T, Lu H, et al. (2017) Population pharmacokinetics of vancomycin in Chinese pediatric patients. Int J Clin Pharmacol Ther 55: 509-516.
  91. Llanos-Paez CC, Staatz CE, Lawson R, Hennig S (2017) A Population Pharmacokinetic Model of Gentamicin in Pediatric Oncology Patients To Facilitate Personalized Dosing. Antimicrob Agents Chemother 61: e00205-e00217.
  92. Lugo RA, Harrison AM, Cash J, Sweeley J, Vernon DD (2001) Pharmacokinetics and pharmacodynamics of ranitidine in critically ill children. Crit Care Med 29: 759-764.
  93. Lulic-Botica M, Sheer T, Edwards D, Thomas RL, Natarajan G (2014) Impact of small-for-gestational age (SGA) status on gentamicin pharmacokinetics in neonates. J Clin Pharmacol 54: 39-45.
  94. Lynn AM, Slattery JT (1987) Morphine pharmacokinetics in early infancy. Anesthesiology 66: 136-139.
  95. Mahmoud MA, Ebid AHIM, Shouman SA, Ebid EN (2014) Pharmacokinetics of vancomycin in oncology egyptian paediatrics: a dosage adjustment trial. Indian J Pharm Sci 76: 82-86.
  96. Mark LF, Solomon A, Northington FJ, Lee CKK (2013) Gentamicin pharmacokinetics in neonates undergoing therapeutic hypothermia. Ther Drug Monit 35: 217-222.
  97. Martínková J, Pokorná P, Záhora J, Chládek J, Vobruba V, et al. (2010) Tolerability and outcomes of kinetically guided therapy with gentamicin in critically ill neonates during the first week of life: an open-label, prospective study. Clin Ther 32: 2400-2414.
  98. Mathews HM, Carson IW, Lyons SM, Orr IA, Collier PS, et al. (1988) A pharmacokinetic study of midazolam in paediatric patients undergoing cardiac surgery. Br J Anaesth 61: 302-307.
  99. McDade EJ, Wagner JL, Moffett BS, Palazzi DL (2010) Once-daily gentamicin dosing in pediatric patients without cystic fibrosis. Pharmacotherapy 30: 248-253.
  100. McDougal A, Ling EW, Levine M (1995) Vancomycin pharmacokinetics and dosing in premature neonates. Ther Drug Monit 17: 319-326.
  101. McKamy S, Hernandez E, Jahng M, Moriwaki T, Deveikis A, et al. (2011) Incidence and risk factors influencing the development of vancomycin nephrotoxicity in children. J Pediatr 158: 422-426.
  102. Medellín-Garibay SE, Rueda-Naharro A, Peña-Cabia S, García B, Romano-Moreno S, et al. (2015) Population pharmacokinetics of gentamicin and dosing optimization for infants. Antimicrobial Agents and Chemotherapy 59: 482-489.
  103. Mikkelsen S, Feilberg VL, Christensen CB, Lundstrøm KE (1994) Morphine pharmacokinetics in premature and mature newborn infants. Acta Paediatrica 83: 1025-1028.
  104. Moffett BS, Kim S, Edwards MS (2011) Vancomycin dosing in obese pediatric patients. Clin Pediatr 50: 442-446.
  105. Muchohi SN, Kokwaro GO, Ogutu BR, Edwards G, Ward SA, et al. Pharmacokinetics and clinical efficacy of midazolam in children with severe malaria and convulsions. Br J Clin Pharmacol 66: 529-538.
  106. Mulla H, McCormack P, Lawson G, Firmin RK, Upton DR (2003) Pharmacokinetics of midazolam in neonates undergoing extracorporeal membrane oxygenation. Anesthesiology 99: 275-282.
  107. Mulla H, Pooboni S (2005) Population pharmacokinetics of vancomycin in patients receiving extracorporeal membrane oxygenation. Br J Clin Pharmacol 60: 265-275.
  108. Murphy JE, Severnak T (1996) Gentamicin pharmacokinetics in Native Americans of Apache ancestry. American journal of health-system pharmacy: AJHP: official journal of the American Society of Health-System Pharmacists 53: 2189-2191.
  109. Pharmacokinetic profile of extended interval gentamicin in critically ill pediatric patients. Pediatric Critical Care Medicine 12: A108-A108.
  110. Nahara MC, McMorrow J, Jones PR, Anglin D, Rosenberg R (2000) Pharmacokinetics of midazolam in critically ill pediatric patients. Eur J Drug Metab Pharmacokinet 25: 219-221.
  111. Naqvi SH, Keenan WJ, Reichley RM, Fortune KP (1986) Vancomycin pharmacokinetics in small, seriously ill infants. Am J Dis Child 140: 107-110.
  112. Padbury JF, Agata Y, Baylen BG, Ludlow JK, Polk DH, et al. (1987) Dopamine pharmacokinetics in critically ill newborn infants. J Pediatr 110: 293-298.
  113. Padbury JF, Agata Y, Baylen BG, Ludlow JK, Polk DH, et al. (1990) Pharmacokinetics of dopamine in critically ill newborn infants. J Pediatr 117: 472-476.
  114. Payne K, Mattheyse FJ, Liebenberg D, Dawes T (1989) The pharmacokinetics of midazolam in paediatric patients. Eur J Clin Pharmacol 37: 267-272.
  115. Peterson RG, Simmons MA, Rumack BH, Levine RL, Brooks JG (1980) Pharmacology of furosemide in the premature newborn infant. J Pediatr 97: 139-143.
  116. Ploessl C, White C, Manasco K (2015) Correlation of a Vancomycin Pharmacokinetic Model and Trough Serum Concentrations in Pediatric Patients. Pediatr Infect Dis J 34: e244-247.
  117. Postovsky S, Ben Arush MW, Kassis E, Elhasid R, Krivoy N (1997) Pharmacokinetic analysis of gentamicin thrice and single daily dosage in pediatric cancer patients. Pediatric Hematology and Oncology 14: 547-554.
  118. Rasmussen MB, Gramsbergen JB, Eriksen VR, Greisen G (2018) Dopamine plasma clearance is increased in piglets compared to neonates during continuous dopamine infusion. Acta Paediatrica 107: 249-254.
  119. Reed MD, Kliegman RM, Weiner JS, Huang M, Yamashita TS, et al. (1987) The clinical pharmacology of vancomycin in seriously ill preterm infants. Pediatr Res 22: 360-363.
  120. Relling MV, Mulhern RK, Dodge RK, Johnson D, Pieper JA, et al. (1989) Lorazepam pharmacodynamics and pharmacokinetics in children. J Pediatr 114: 641-646.
  121. Rey E, Delaunay L, Pons G, Murat I, Richard MO, et al. (1991) Pharmacokinetics of midazolam in children: comparative study of intranasal and intravenous administration. Eur J Clin Pharmacol 41: 355-357.
  122. Rocha MJ, Almeida AM, Afonso E, Martins V, Santos J, et al. (2000) The kinetic profile of gentamicin in premature neonates. J Pharm Pharmacol 52: 1091-1097.
  123. Saarenmaa E, Neuvonen PJ, Fellman V (2000) Gestational age and birth weight effects on plasma clearance of fentanyl in newborn infants. J Pediatr 136: 767-770.
  124. Salonen M, Kanto J, Iisalo E, Himberg JJ (1987) Midazolam as an induction agent in children: a pharmacokinetic and clinical study. Anesth Analg 66: 625-628.
  125. Santeiro ML, Christie J, Stromquist C, Torres BA, Markowsky SJ (1997) Pharmacokinetics of continuous infusion fentanyl in newborns. J Perinatol 17: 135-139.
  126. Scott CS, Riggs KW, Ling EW, Fitzgerald CE, Hill ML, et al. (1999) Morphine pharmacokinetics and pain assessment in premature newborns. J Pediatr 135: 423-429.
  127. Seay RE, Brundage RC, Jensen PD, Schilling CG, Edgren BE (1994) Population pharmacokinetics of vancomycin in neonates. Clin Pharmacol Ther 56: 169-175.
  128. Seixas GTF, Araujo OR, Silva DCB, Arduini RG, Petrilli AS (2016) Vancomycin Therapeutic Targets and Nephrotoxicity in Critically Ill Children With Cancer. J Pediatr Hematol Oncol 38: e56-62.
  129. Shankar SM, Jew RK, Bickert BM, Cavalieri GE, Bell LM, et al. (1999) Pharmacokinetics of single daily dose gentamicin in children with cancer. J Pediatr Hematol Oncol 21: 284-288.
  130. Silva DC, Seixas GT, Araujo OR, Arduini RG, Carlesse FA, et al. (2012) Vancomycin serum concentrations in pediatric oncologic/hematologic intensive care patients. Braz J Infect Dis 16: 361-365.
  131. Song L, He CY, Yin NG, Liu F, Jia YT, et al. (2017) A population pharmacokinetic model for individualised dosage regimens of vancomycin in Chinese neonates and young infants. Oncotarget 8: 105211-105221.
  132. Southgate WM, DiPiro JT, Robertson AF (1989) Pharmacokinetics of gentamicin in neonates on extracorporeal membrane oxygenation. Antimicrob Agents Chemother 33: 817-819.
  133. Spivey JM, Gal P (1986) Vancomycin pharmacokinetics in neonates. American Journal of Diseases of Children 140: 859-859.
  134. Ting JY, Kwan E, McDougal A, Osiovich H (2015) Pharmacokinetics of gentamicin in newborns with moderate-to-severe hypoxic-ischemic encephalopathy undergoing therapeutic hypothermia. Indian J Pediatr 82: 119-125.
  135. Tolia V, Brennan S, Aravind MK, Kauffman RE (1991) Pharmacokinetic and pharmacodynamic study of midazolam in children during esophagogastroduodenoscopy. J Pediatr 119: 467-471.
  136. Tseng SH, Lim CP, Chen Q, Tang CC, Kong ST, et al. (2018) Evaluating the Relationship between Vancomycin Trough Concentration and 24-Hour Area under the Concentration-Time Curve in Neonates. Antimicrobial Agents and Chemotherapy 62: e01647-17.
  137. Tuck S, Morselli P, Broquaire M, Vert P (1983) Plasma and urinary kinetics of furosemide in newborn infants. J Pediatr 103: 481-485.
  138. Vandenberghe H, Mac Leod S, Chinyanga H, Endrenyi L, Soldin S (1983) Pharmacokinetics of intravenous morphine in balanced anesthesia: studies in children. Drug Metab Rev 14: 887-903.
  139. Vert P, Broquaire M, Legagneur M, Morselli PL (1982) Pharmacokinetics of furosemide in neonates. Eur J Clin Pharmacol 22: 39-45.
  140. Zhao W, Zhang,D, Fakhoury M, Fahd M, Duquesne F, et al. (2014) Population pharmacokinetics and dosing optimization of vancomycin in children with malignant hematological disease. Antimicrob Agents Chemother 58: 3191-3199.
  141. Watterberg KL, Kelly HW, Angelus P, Backstrom C (1989) The need for a loading dose of gentamicin in neonates. Ther Drug Monit 11: 16-20.
  142. Wells TG, Heulitt MJ, Taylor BJ, Fasules JW, Kearns GL (1998) Pharmacokinetics and pharmacodynamics of ranitidine in neonates treated with extracorporeal membrane oxygenation. J Clin Pharmacol 38: 402-407.
  143. Welzing L, Junghaenel S, Weiss V, Roth B, Mueller C, et al. (2013) Disposition of midazolam in asphyxiated neonates receiving therapeutic hypothermia - A pilot study. Klin Padiatr 225: 398-404.
  144. Williams BS, Ransom JL, Gal P, Carlos RQ, Smith M, et al. Gentamicin pharmacokinetics in neonates with patent ductus arteriosus. Crit Care Med 25: 273-275.
  145. Wrishko RE, Levine M, Khoo D, Abbott P, Hamilton D (2000) Vancomycin pharmacokinetics and Bayesian estimation in pediatric patients. Ther Drug Monit 22: 522-531.
  146. Zakova M, Pong S, Trope A, Atenafu EG, Papaioannou V, et al. (2014) Dose derivation of once-daily dosing guidelines for gentamicin in critically ill pediatric patients. Ther Drug Monit 36: 288-294.
  147. Zaritsky A, Lotze A, Stull R, Goldstein DS (1988) Steady-state dopamine clearance in critically ill infants and children. Crit Care Med 16: 217-220.
  148. Zhang H, Wang Y, Gao P, Hu J, Chen Y, et al. (2016) Pharmacokinetic Characteristics and Clinical Outcomes of Vancomycin in Young Children With Various Degrees of Renal Function. J Clin Pharmacol 56: 740-748.
  149. Zuppa AF, Hammer GB, Barrett JS, Kenney BF, Kassir N, et al. (2011) Safety and population pharmacokinetic analysis of intravenous acetaminophen in neonates, infants, children, and adolescents with pain or Fever. J Pediatr Pharmacol Ther 16: 246-261.
  150. Zhao W, Lopez E, Biran V, Durrmeyer X, Fakhoury M, et al. (2013) Vancomycin continuous infusion in neonates: dosing optimisation and therapeutic drug monitoring. Arch Dis Child 98: 449-453.
  151. Barrett JS, Narayan M, Patel D, Zuppa AF, Adamson PC (2011) Prescribing habits and caregiver satisfaction with resources for dosing children: rationale for more informative dosing guidance. BMC Pediatr 11: 25.
  152. Bartelink IH, Rademaker CMA, Schobben AF, van den Anker JN (2006) Guidelines on Paediatric Dosing on the Basis of Developmental Physiology and Pharmacokinetic Considerations. Clin Pharmacokinet 45: 1077-1097.
  153. Ceelie I, van der Starre C, Tibboel D, Stol K, Koren G, et al. (2011) Evaluation of drug formularies for pediatric intensive care. Pediatr Crit Care Med 12: e14-e19.
  154. Kurz H, Mauser-Ganshorn A, Stickel H (1977) Differences in the binding of drugs to plasma proteins from newborn and adult man. I. Eur J Clin Pharmacol 11: 463-467.
  155. Crom WR (1994) Pharmacokinetics in the child. Environ Health Perspect 102: 111-117.
  156. Anderson B, Holford N (2008) Mechanism-based concepts of size and maturity in pharmacokinetics. Annu Rev Pharmacol Toxicol 48: 303-332.
  157. Madadi P, Koren G (2008) Pharmacogenetic insights into codeine analgesia: implications to pediatric codeine use. Pharmacogenomics 9: 1267-1284.
  158. Stojiljkovic M, Patrinos G, Pavlovic S (2011) Clinical applicability of sequence variations in genes related to drug metabolism. Curr Drug Metab 12: 445-454.
  159. Mahmood I, Ahmad T, Mansoor N, Sharib SM (2017) Prediction of Clearance in Neonates and Infants (≤ 3 Months of Age) for Drugs That Are Glucuronidated: A Comparative Study Between Allometric Scaling and Physiologically Based Pharmacokinetic Modeling. J Clin Pharmacol 57: 476-483.
  160. Brussee JM, Vet NJ, Krekels EH, Valkenburg AJ, Jacqz‐Aigrain E, et al. (2018) Predicting CYP3A‐mediated midazolam metabolism in critically ill neonates, infants, children and adults with inflammation and organ failure. Br J Clin Pharmacol 84: 358-368.
  161. Koch G, Datta AN, Jost K, Schulzke SM, van den Anker J, et al. (2017) Caffeine citrate dosing adjustments to assure stable caffeine concentrations in preterm neonates. J Pediatr 191: 50-56.
  162. Shekar K, Fraser JF, Taccone FS, Welch S, Wallis SC, et al. (2014) The combined effects of extracorporeal membrane oxygenation and renal replacement therapy on meropenem pharmacokinetics: a matched cohort study. Critical Care 18: 565.
  163. Shakhnovich V, Hornik CP, Kearns GL, Weigel J, Abdel-Rahman SM (2019) How to Conduct Clinical Trials in Children: A Tutorial. Clin Transl Sci 12: 218-230.
  164. Goldstein SL, Nolin TD (2014) Lack of Drug Dosing Guidelines for Critically Ill Patients Receiving Continuous Renal Replacement Therapy. Clin Pharmacol Ther 96: 159-161.
  165. Wildschut ED, Ahsman MJ, Houmes RJ, Pokorna P, de Wildt SN, et al. (2012) Pharmacotherapy in neonatal and pediatric extracorporeal membrane oxygenation (ECMO). Curr Drug Metab 13: 767-777.
  166. Wang Z, Kim S, Quinney SK, Guo Y, Hall SD, et al. (2009) Literature mining on pharmacokinetics numerical data: a feasibility study. J Biomed Inform 42: 726-735.
  167. Benjamin DK, Smith PB, Murphy MD, Roberts R, Mathis L, et al. (2006) Peer-reviewed publication of clinical trials completed for pediatric exclusivity. Jama 296: 1266-1273.

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. With this license, readers can share, distribute, download, even commercially, as long as the original source is properly cited. Read More.

Journal of Hospital and Healthcare Administration

cara menggunakan pola slot mahjongrtp tertinggi hari inislot mahjong ways 1pola gacor olympus hari inipola gacor starlight princessslot mahjong ways 2strategi olympustrik mahjong ways 2trik olympus hari inirtp koi gatertp pragmatic tertinggicheat jackpot mahjongpg soft link gamertp jackpotelemen sakti mahjongpola maxwin mahjongslot olympus mudah mainrtp live starlightrumus slot mahjongmahjong scatter hitamslot pragmaticjam gacor mahjongpola gacor mahjongstrategi maxwin olympusslot jamin menangrtp slot gacorscatter wild banditopola slot mahjongstrategi maxwin sweet bonanzartp slot terakuratkejutan scatter hitamslot88 resmimaxwin olympuspola mahjong pgsoftretas mahjong waystrik mahjongtrik slot olympusewallet modal recehpanduan pemula slotpg soft primadona slottercheat mahjong androidtips dewa slot mahjongslot demo mahjonghujan scatter olympusrtp caishen winsrtp sweet bonanzamahjong vs qilinmaxwin x5000 starlight princessmahjong wins x1000rtp baru wild scatterpg soft trik maxwinamantotorm1131