Population Pharmacokinetic Comparison and Pharmacodynamic Breakpoints of Ceftazidime in Cystic Fibrosis Patients and Healthy Volunteers

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, Mar. 2010, p. 1275–1282 Vol. 54, No.

3
0066-4804/10/$12.00 doi:10.1128/AAC.00936-09
Copyright © 2010, American Society for Microbiology. All Rights Reserved.

Population Pharmacokinetic Comparison and Pharmacodynamic


Breakpoints of Ceftazidime in Cystic Fibrosis Patients and
Healthy Volunteers䌤
J. B. Bulitta,1† C. B. Landersdorfer,1† S. J. Hüttner,1 G. L. Drusano,2
M. Kinzig,1 U. Holzgrabe,3 U. Stephan,1,4‡ and F. Sörgel1,5*
IBMP Institute for Biomedical and Pharmaceutical Research, Nürnberg-Heroldsberg, Germany1; Ordway Research Institute, Albany,
New York2; Institute of Pharmacy and Food Chemistry, University of Würzburg, Würzburg, Germany3; Department of Pediatrics,
University of Duisburg-Essen, Essen, Germany4; and Department of Pharmacology, University of Duisburg-Essen, Essen, Germany5
Received 7 July 2009/Returned for modification 29 September 2009/Accepted 29 December 2009

Despite the promising activity of ceftazidime against Pseudomonas aeruginosa and Burkholderia cepacia, there
has not yet been a study that directly compared the pharmacokinetics (PK) of ceftazidime in cystic fibrosis
(CF) patients and healthy volunteers by population PK methodology. We assessed the population PK and
PK/pharmacodynamic (PD) breakpoints of ceftazidime in CF patients and healthy volunteers. Eight CF
patients (total body weight [WT] [average ⴞ standard deviation] ⴝ 42.9 ⴞ 18.4 kg) and seven healthy
volunteers (WT ⴝ 66.2 ⴞ 4.9 kg) received 2 g ceftazidime as a 5-min intravenous infusion. High-performance
liquid chromatography (HPLC) was used for drug analysis, and NONMEM (results reported), S-ADAPT, and
NPAG were used for parametric and nonparametric population PK modeling. We considered linear and
allometric body size models to scale clearance and volume of distribution. Monte Carlo simulations were based
on a target time of non-protein-bound plasma concentration of ceftazidime above MIC of >65%, which
represents near-maximal killing. Unscaled total clearance was 19% lower in CF patients, and volume of
distribution was 36% lower. Total clearance was 7.82 liters/h for CF patients and 6.68 liters/h for healthy
volunteers with 53 kg fat-free mass. Allometric scaling by fat-free mass reduced the between-subject variability
by 32% for clearance and by 18 to 26% for volume of both peripheral compartments compared to linear scaling
by WT. A 30-min ceftazidime infusion of 2 g/70 kg WT every 8 h (q8h) achieved robust (>90%) probabilities
of target attainment (PTAs) for MICs of <1 mg/liter in CF patients and <3 mg/liter in healthy volunteers.
Alternative modes of administration achieved robust PTAs up to markedly higher MICs of <8 to 12 mg/liter
in CF patients for 5-h infusions of 2 g/70 kg WT q8h and <12 mg/liter for continuous infusion of 6 g/70 kg WT
daily.

Respiratory tract infections are the primary reason for fre- pared the pharmacodynamic (PD) profiles of CF patients and

Downloaded from https://journals.asm.org/journal/aac on 10 August 2021 by 196.191.48.216.


quent hospitalization of patients with cystic fibrosis (CF) and healthy volunteers based on data from two different studies
the main cause of mortality (4, 60). About 4 of 5 adults with CF and found that CF patients required higher doses than healthy
(age: 26 to 30 years) are infected by Pseudomonas aeruginosa volunteers to achieve the same PD target.
(24). Ceftazidime is still one of the most active antibiotics For beta-lactams, the duration of non-protein-bound plasma
against P. aeruginosa isolates from CF patients and has activity concentration above the MIC (fT⬎MIC) is the PK-PD index
against species belonging to the Burkholderia cepacia complex that best predicts the drug-related responses (2, 22, 25). An
(45, 46, 64). Infections by B. cepacia are of particular interest fT⬎MIC of 65% has been identified as the target for near-
to CF patients, since B. cepacia can cause a rapid deterioration maximal bacterial killing, and an fT⬎MIC target of 40% best
of lung function (29, 71). Efficacious anti-infective therapy predicts bacteriostasis at 24 h for cephalosporins in mouse
against both B. cepacia and P. aeruginosa is critical, in partic- infection models (22, 25). Based on these target values the
ular as eradication of P. aeruginosa in the lungs of CF patients probability of target attainment (PTA) can be predicted via
is possible only in the early stage of infection (19, 34). Monte Carlo simulation (MCS). The effect of an altered body
Two groups compared the pharmacokinetics (PK) in CF composition on PK requires dose adjustment in certain patient
patients and healthy volunteers within the same study (33, 44). populations (21, 31, 49, 55, 71). Therefore, it may be important
CF patients had higher clearances (liters/h/kg) and shorter to use a descriptor that accounts for body size and body com-
terminal half-lives for ceftazidime. Mouton et al. (53) com- position for dose selection and optimization of the PTA. A lack
of adipose tissue in CF patients may require dose adjustments
(16, 48, 62, 71).
* Corresponding author. Mailing address: IBMP Institute for Bio- Due to the hydrophilic properties of ceftazidime, we believed
medical and Pharmaceutical Research, Paul-Ehrlich-Str. 19, D-90562 that fat-free mass (FFM) may better describe body size than total
Nürnberg-Heroldsberg, Germany. Phone: 49-911-518290. Fax: 49-911- body weight (WT), as FFM but not WT accounts for body com-
5182920. E-mail: ibmp@osn.de.
† Present address: Ordway Research Institute, Albany, NY 12208.
position. In addition to explaining the differences in average PK
‡ Deceased. parameters between lean, “normal,” and obese patients, a useful

Published ahead of print on 11 January 2010. body size descriptor should reduce the unexplained between-

1275
1276 BULITTA ET AL. ANTIMICROB. AGENTS CHEMOTHER.

TABLE 1. Demographic data sample handling, storage, and drug analysis procedures for the applied condi-
tions. A LiChrosphere RP 18-5 ␮ (50- by 4-mm) column with a water-acetonitrile
Median 关range兴 for: mixture at pH 5.0 was used. Ceftazidime was detected at a wavelength of 275 nm.
Parameter CF patients Healthy volunteers Calibration was performed by linear regression. The ceftazidime assay was linear
(n ⫽ 8c) (n ⫽ 7d) between 0.6 and 200 mg/liter. The assay was validated, and its performance
(including bias and precision) was deemed acceptable.
Height (cm) 164 关105–176兴 175 关164–191兴 Population PK analysis. (i) Structural model. We tested one-, two-, and
Age (yr) 20 关10–45兴 22 关19–33兴 three-compartment models with linear disposition and evaluated competing
Total body wt (kg) 37.9 关14.2–73.5兴 67 关56–71兴 models using their predictive performance assessed via visual predictive checks
Fat-free massa (kg) 35.9 关13.9–57.7兴 54.0 关46.4–60.5兴 (VPCs), the objective function, and standard diagnostic plots as described pre-
Lean body massb (kg) 34.7 关13.3–58.6兴 54.5 关46.7–60.5兴 viously (16, 18).
Body mass index (kg m⫺2) 15.0 关12.5–23.7兴 21.7 关19.5–23.7兴 (ii) Body size model. The following body size descriptors and body size models
a
were considered: (i) no body size model, (ii) linear scaling by WT, (iii) allometric
Calculated by the formula of Janmahasatian et al. (39). scaling by WT (3, 35, 80), (iv) linear scaling by a new equation estimating FFM
b
Calculated by the formula of Cheymol and James (21, 38).
c (39), and (v) allometric scaling by FFM. We assessed the ability of each body size
Four males and four females.
d
Four males and three females. model to describe the differences in average PK parameters between both subject
groups and to reduce the unexplained (random) BSV. The exponents for the
effect of body size on clearance were fixed to 1.0 for linear scaling and to 0.75 for
allometric scaling as described previously (16). Exponents were fixed to 1.0 for
subject variability (BSV) in PK parameters. Such a body size volumes of distribution for linear and allometric body size models.
(iii) Differences between subject groups. We used scale factors for clearance
descriptor can be used to select dosage regimens that achieve (FCYFCL) and volume of distribution at steady state (FCYFVSS) to describe the
target concentrations and target effects more precisely. difference in average PK parameters after accounting for the effect of body size
Our first objective was to compare the population PK of as described previously (16).
ceftazidime between CF patients and healthy volunteers within (iv) Between-subject variability and observation model. An exponential model
the same study. Second, we assessed whether the difference in was used to describe the BSV of PK parameters, and a combined additive and
proportional error model was used to describe the residual unidentified variabil-
average PK parameters and the BSV in clearance and volume ity as reported previously (16, 18). We used the first-order conditional estimation
of distribution are better described by FFM than by WT. Our with the interaction option (FOCE⫹I) in NONMEM V release 1.1 (10), the
third objective was to predict the PK-PD MIC breakpoints for importance sampling Monte-Carlo parametric expectation maximization method
various ceftazidime dosage regimens in CF patients. We ap- (pmethod ⫽ 8) in S-ADAPT (version 1.56) (9), and the nonparametric adaptive
grid (NPAG) algorithm of the USC*PACK (version 12.00) (43) for population
plied the latest parametric and nonparametric population PK
PK modeling as described previously (16, 18). WinNonlin Professional (version
methodology and evaluated linear and allometric body size 4.0.1; Pharsight Corp., Mountain View, CA) was used for noncompartmental
models comparing the PK in CF patients and healthy volun- analysis and statistics.
teers in order to greatly extend a descriptive noncompartmen- Monte Carlo simulation. PK-PD targets of 65% fT⬎MIC for near-maximal
tal analysis of our study reported previously (65–67). bactericidal activity and of 40% fT⬎MIC for bacteriostasis at 24 h were applied
based on data from animal infection models (22, 25). We studied a range of
(The population PK modeling work has in part been pre- MICs from 0.125 to 128 mg/liter. As the reported protein binding of ceftazidime
sented as part of a meta-analysis [14, 15] and in part as a poster ranges between 2 and 19% (32, 44, 52, 54), we assumed an average protein
[37].) binding of 11%.
We compared the following five dosage regimens, all at a daily dose of 6 g
ceftazidime per 70 kg WT: (i) short-term (30-min) infusion every 8 h (q8h), (ii)
MATERIALS AND METHODS 30-min infusion q6h, (iii) prolonged (5-h) infusion q8h, (iv) prolonged (4-h)

Downloaded from https://journals.asm.org/journal/aac on 10 August 2021 by 196.191.48.216.


Subjects. A total of 15 Caucasian volunteers (8 CF patients and 7 healthy infusion q6h, and (v) continuous infusion. Concentrations of each dosage regi-
volunteers) participated in the study after they had given their written informed men were simulated at steady state in the absence of residual error. We simu-
consent. For the two CF patients 10 and 15 years old, written informed consent lated 11,200 CF patients and 9,800 healthy volunteers with the same demo-
was obtained from their legal representative. General clinical procedures were as graphic data as the subjects in our study. All estimated covariance terms were
described previously (16). The study was performed in 1983; the study protocol included in the VPC and MCS. The PTA was calculated as the fraction of
had been approved by the local ethics committee, and the study was conducted subjects who attained the PK-PD target at each MIC. The highest MIC with a
by following the revised version of the Declaration of Helsinki. PTA of at least 90% was defined as PK-PD MIC breakpoint.
Study design and drug administration. The study was a single-dose, single-
center, open, parallel group trial. Subjects received 2 g ceftazidime as a 5-min
intravenous infusion. As some CF patients were notably smaller or required a RESULTS
higher dose for clinical reasons as judged by the physician, one CF patient
received 1.5 g, one CF patient received 1 g, and one CF patient received 3 g Demographics and noncompartmental analysis. Our CF pa-
instead of 2 g ceftazidime. All infusions were administered with Perfusors tients had a normal renal function and were smaller and leaner
(Braun, Melsungen, Germany). These instruments were checked on a daily basis than our healthy volunteers (Table 1). Peak concentrations
by weighing defined volumes delivered by the Perfusors.
Blood sampling. All blood samples were drawn from a forearm vein via an
in CF patients were approximately twice as high as those in
intravenous catheter contralateral to the one used for drug administration. Blood healthy volunteers (Table 2). We did not scale the noncom-
samples were drawn immediately before the start of the infusion (0 min), at the partmental PK parameters (Table 2) by any body size descrip-
end of the infusion (5 min), and at 5, 10, 15, 20, 30, 45, 60, and 90 min and 2, 2.5, tor. Total clearance was 19% lower, volume of distribution at
3, 3.5, 4, 5, 6, 8, 10, and 12 h after the end of infusion. Conditions of sample
steady state was 36% smaller, and terminal half-life was 24%
handling and storage were the same as described previously (16).
Drug analysis. Ceftazidime concentrations in plasma were determined by a shorter in CF patients than in healthy volunteers.
reversed-phase high-performance liquid chromatography (HPLC) assay with an Population PK analysis. The visual predictive check (Fig. 1)
internal standard. An amount of 100 ␮l of NaH2PO4 buffer was added to 100 ␮l indicated highly sufficient predictive performance for the linear
of each sample. Acetonitrile (400 ␮l) was used to deproteinize each sample. three-compartment model, which was slightly better than that
After centrifugation, 1,000 ␮l of dichloromethane was added for extraction of the
organic phase. From the remaining aqueous phase, 20 ␮l was injected into the
for the linear two-compartment model. A one-compartment
HPLC system. After sample preparation, the prepared samples were stored in an model had insufficient predictive performance and yielded
ice-water bath until injection. We assured the stability of ceftazidime during all poor curve fits. As the objective function in NONMEM was
VOL. 54, 2010 CEFTAZIDIME POPULATION PK AND CF PATIENT BREAKPOINTS 1277

TABLE 2. Unscaled PK parameters from noncompartmental analysis volunteers after adjusting for body size using the respective
Median 关minimum–maximum兴 for:
body size model. The scale factors from NONMEM (Table 4)
Parameter were well comparable to results from S-ADAPT (data not
CF patients Healthy volunteers
shown).
Total clearance (liters/h) 5.37 关3.35–12.8兴 6.59 关4.87–9.50兴 Without accounting for body size, clearance and volume of
Volume of distribution 9.14 关2.77–19.9兴 14.3 关10.8–17.1兴 distribution were 14 to 15% lower in CF patients (Table 4).
at steady state (liters)
The linear body size models yielded 45% or 30% higher clear-
Peak concn (mg/liter) 445 关151–1200兴a 210 关148–397兴
Terminal half-life (h) 1.48 关0.49–1.78兴 1.94 关1.28–2.78兴 ances in CF patients. The allometric body size models ex-
Mean residence time (h) 1.54 关0.62–2.35兴 2.10 关1.71–2.87兴 plained the differences between CF patients and healthy vol-
a
Normalized to a dose of 2 g ceftazidime, since three CF patients received a
unteers better than linear models. The allometric model based
slightly higher or slightly lower dose. on FFM yielded a 17% higher clearance in CF patients (Table
4). Allometric body size models also reduced the unexplained
BSV in clearance and volume of distribution more than the
worse by 46 for the two-compartment model than for the linear body size models. Allometric scaling by FFM reduced
three-compartment model, the latter model was selected as the the unexplained BSV by 32% for total clearance and by ap-
final model. This choice was consistent with results from S- proximately 18 to 26% for volume of distribution for the
ADAPT and NPAG. peripheral compartments (Table 5). As the allometric body size
The estimates for the final population PK model with model based on FFM explained most of the differences in
allometric scaling by FFM were well comparable between clearance and volume of distribution between both subject
NONMEM, S-ADAPT, and NPAG (Table 3) despite the differ- groups, we did not seek to include other potentially confound-
ent estimation algorithms applied. Estimates for volume of the ing variables in the covariate model.
deep peripheral compartment were larger in NPAG, since Monte Carlo simulation. CF patients had lower PTAs than
there were 3 subjects in each group who had large volumes for healthy volunteers, which resulted in 1.5- to 3-times-lower
this compartment. Allometric scaling by FFM explained the PK-PD MIC breakpoints for the studied dosage regimens (Fig.
differences in average PK parameters better than the other 2). Continuous infusion of 6 g/70 kg WT per day had a PK-PD
body size models, since it yielded disease-specific scale factors breakpoint of 12 mg/liter in CF patients and 16 mg/liter in
(FCYFCL and FCYFVSS in Table 4) closest to 1.0. A scale healthy volunteers for both targets. For the near-maximal kill
factor of 1.0 means that the average clearance or volume of target (fT⬎MIC ⱖ 65%), PK-PD MIC breakpoints were 8 to 12
distribution is the same in both patient groups, if the patient mg/liter in CF patients and 16 mg/liter in healthy volunteers for
groups are matched in body size. These scale factors represent both prolonged-infusion regimens. For the near-maximal kill
the ratios of group estimates between CF patients and healthy target, CF patients had breakpoints of 2 mg/liter for short-term

Downloaded from https://journals.asm.org/journal/aac on 10 August 2021 by 196.191.48.216.

FIG. 1. Visual predictive check based on 8,000 CF patients and 7,000 healthy volunteers simulated from the three-compartment model based
on FFM (see Table 3; based on results from NONMEM). The plots show the observed data, the 80% prediction intervals (10 to 90% percentile),
and the interquartile ranges (25 to 75% percentile). Ideally, 50% of the observed data points should fall inside the interquartile range and 80%
of the observed data should fall inside the 80% prediction interval.
1278 BULITTA ET AL. ANTIMICROB. AGENTS CHEMOTHER.

TABLE 3. Estimates from the population PK model based on allometric scaling by fat-free mass
Estimate (CV 关%兴 for between-subject variabilityc) by:

NONMEM S-ADAPT NPAGf


Parametera
Healthy CF Healthy CF Healthy
CF patients
volunteers patients volunteers patients volunteers

CL (liters h⫺1) 7.82b (28) 6.68 (28) 7.70 (30) 6.61 (30) 7.51 6.30
Vssd (liters) 12.8 12.7 12.5 13.6 24.0g 17.9g
V1 (liters) 5.73 (45)e 5.67 (45) 5.43 (40) 6.56 (40) 4.36 6.34
V2 (liters) 3.92 (25) 3.88 (25) 4.34 (43) 4.01 (43) 4.94 3.24
V3 (liters) 3.16 (29) 3.13 (29) 2.75 (33) 3.02 (33) 14.7g 8.35g
CLicshallow (liters h⫺1) 27.9 27.9 19.2 (68) 19.2 (68) 24.3 24.3
CLicdeep (liters h⫺1) 2.57 2.57 1.05 (60) 1.05 (60) 2.24 2.24
CVC 0.122 0.122 0.119 0.119 0.149 0.149
SDC (mg/liter) 0.059 0.059 0.057 0.057 0.078 0.078
a
CL, total clearance; Vss, volume of distribution at steady state; V1, volume of distribution for the central compartment; V2, volume of distribution for the shallow
peripheral compartment; V3, volume of distribution for the deep peripheral compartment; CLicshallow, intercompartmental clearance between the central and the
shallow peripheral compartment; CLicdeep, intercompartmental clearance between the central and the deep peripheral compartment; CVC, proportional residual error
component for the plasma concentrations; SDC, additive residual error component for the plasma concentrations. The duration of zero order input was fixed to 5 min
and not estimated.
b
All clearance and volume estimates are group estimates of the respective PK parameter for subjects of standard size (fat-free mass: 53 kg).
c
Apparent coefficients of variation describing the between-subject variability. As sample size was small, one joint variance was estimated for CF patients and healthy
volunteers for each parameter.
d
Derived from the group means of V1, V2, and V3, not an estimated parameter.
e
Coefficients of correlation for the random variability between pairs of random effects are as follows: r(V1,V2) ⫽ ⫺0.67, r(V1,V3) ⫽ 0.56, r(V2,V3) ⫽ ⫺0.59.
f
Estimates reported for NPAG are geometric means of the support points for each subject group.
g
The nonparametric estimation algorithm in NPAG yielded large volumes of the deep peripheral compartment (between 20 and 50 liters) for three CF patients and
three healthy volunteers. Therefore, the geometric means for V3 in NPAG are larger than those in NONMEM and S-ADAPT.

infusion q6h and of 1 mg/liter for short-term infusion q8h at a this pathogen in CF patients. Ceftazidime is still the cephalo-
daily dose of 6 g/70 kg WT. Healthy volunteers had breakpoints of sporin with the best activity against P. aeruginosa (45) and is a
4 mg/liter for short-term infusion q6h and of 3 mg/liter for short- valuable treatment option in antipseudomonal therapy in CF
term infusion q8h at a daily dose of 6 g/70 kg WT. patients (6, 12, 61, 69).
For the bacteriostasis target (fT⬎MIC ⱖ 40%), the PK-PD MCS can identify dosage regimens that achieve a high PTA
MIC breakpoints were 16 mg/liter in CF patients and 24 mg/ for treatment of a specific patient population (26). To better
liter in healthy volunteers for both prolonged-infusion regi- understand the potentially altered PK in CF patients compared
mens. For this target, CF patients had breakpoints of 6 mg/liter to healthy volunteers, differences in body size and body com-
for short-term infusion q6h and of 4 mg/liter for short-term position should be considered for an MCS. We assessed the
infusion q8h at a daily dose of 6 g/70 kg WT. Healthy volun- effect of these factors on the PK of ceftazidime within the same

Downloaded from https://journals.asm.org/journal/aac on 10 August 2021 by 196.191.48.216.


teers had breakpoints of 12 mg/liter for short-term infusion study. While this assures that the clinical and bioanalytical
q6h and of 8 mg/liter for short-term infusion q8h at a daily dose procedures are standardized, one limitation of our study is that
of 6 g/70 kg WT. the demographic characteristics of CF patients and healthy
volunteers were not matched. The resulting larger range in
DISCUSSION body composition in our study is likely to support a distinction
between different body size models. Another potential limita-
The life expectancy and quality of life of CF patients have
improved impressively during the last 70 years. Optimal treat-
ment of infections by P. aeruginosa (28) is crucially important, TABLE 5. Between-subject variability (variances) for
as it is almost impossible to eradicate chronic lung infections by different body size modelsa
Relative between-subject variance (%) for:
Body size model
CL V1 V2 V3
TABLE 4. Ratiosa of group estimates for clearance and volume of
b b b
distribution for different body size models (based on WT (linear scaling) 100 100 100 100b
results from NONMEM) WT (allometric scaling) 75c 97 96 89
FFM (linear scaling) 85c 105 78 95
Body size model FCYFCL FCYFVSS
FFM (allometric scaling) 68c 104 74 82
No body size model 0.861 0.853 a
Shown are relative between-subject variances for the respective PK param-
WT (linear scaling) 1.45 1.13 eter and body size models (see Table 3 for parameter explanations) based on
WT (allometric scaling) 1.27 1.13 results from NONMEM.
b
FFM (linear scaling) 1.30 1.01 The between-subject variance for linear scaling by WT was used as the
FFM (allometric scaling) 1.17 1.01 reference.
c
The lower this number, the more variability was explained by the respective
a
FCYFCL, ratio of group estimates for total clearance in CF patients divided body size model. These values indicate that the between-subject variability (vari-
by total clearance in healthy volunteers; FCYFVSS, ratio of group estimates for ance) for total clearance was reduced by 25% for allometric scaling by WT, by
volume of distribution at steady state in CF patients divided by volume of 15% for linear scaling by FFM, and by 32% for allometric scaling by FFM, all
distribution at steady state in healthy volunteers. compared to linear scaling by WT.
VOL. 54, 2010 CEFTAZIDIME POPULATION PK AND CF PATIENT BREAKPOINTS 1279

Downloaded from https://journals.asm.org/journal/aac on 10 August 2021 by 196.191.48.216.


FIG. 2. Probability of target attainment for different dosage regimens of 6 g ceftazidime per 70 kg WT daily at steady state (based on results
from NONMEM).

tion of our study from 1983 is that our results for relatively lean it was based only on 15 subjects for whom we had frequent
CF patients may not be directly applicable to therapy of CF plasma samples. However, our results for the PK parameters of
patients with normal body composition nowadays. However, ceftazidime in healthy volunteers were in good agreement with
our results may be very valuable for CF patients in economi- those from other authors (5, 27, 40, 52, 57, 70, 78). We are
cally challenged countries and potentially for other patient aware of only two studies on the PK of ceftazidime in CF
groups with lean body composition. A population PK analysis patients which included a healthy volunteer control group.
with lean CF patients seems important to put PK studies of Leeder et al. (44) found a total clearance of 8.5 ⫾ 1.0 liters/
(relatively) lean CF patients from the 1970s and 1980s in per- h/1.73 m2 and Hedman et al. (33) found 7.6 ⫾ 0.7 liters/h/1.73
spective with more recent studies of CF patients of normal m2 in CF patients. Although some authors report (47, 56, 72)
body size and body composition that are matched to healthy higher clearances of 0.23 to 0.36 liters/h/kg (equivalent to 16 to
volunteers, as in the aztreonam study by Vinks et al. (77). 25 liters/h/70 kg WT) for ceftazidime in CF patients, most
Our final population PK model (Table 3) had highly suffi- studies (41, 50, 53, 58, 75, 76) of juvenile to adult CF patients
cient predictive performance (Fig. 1) for CF patients, whereas find an average total clearance of 8 to 11 liters/h for CF pa-
predictions for healthy volunteers were slightly too variable. tients of standard body size (i.e., 70 kg WT or 1.73 m2 body
This results in slightly more-conservative predictions for the surface area) and a coefficient of variation (CV) of 9 to 26%
PTA in healthy volunteers. One limitation of our MCS is that for the BSV of total clearance. Our geometric mean of 7.82
1280 BULITTA ET AL. ANTIMICROB. AGENTS CHEMOTHER.

liters/h (CV, 28.3%; from NONMEM) for total clearance in healthy volunteers, had shorter drug half-lives (Table 2) and
CF patients with 53 kg FFM (Table 3) was in good agreement required higher beta-lactam doses to achieve similar PK-PD
with the clearances for CF patients in the literature. MIC breakpoints, in particular for short-term infusions. This
The reported average volume of distribution ranges between prediction matches with the higher clinically recommended
0.237 and 0.46 liters/kg WT (equivalent to 16.6 and 32 liters/70 doses (in mg/kg of total body weight) of beta-lactams for pe-
kg WT), with coefficients of variation between 14 and 53% in diatric CF patients than for adult CF patients (30). Mouton et
CF patients (41, 44, 47, 50, 53, 56, 58, 76). Our geometric mean al. (53) found a PTA of ⱖ90% for MICs of ⱕ4 mg/liter in CF
of 12.8 liters (from NONMEM) for CF patients with 53 kg patients and for MICs of ⱕ8 mg/liter in healthy volunteers for
FFM and coefficients of variation between 25 and 45% for the a fixed dose of 2 g q8h and the target fT⬎MIC of ⱖ60%. When
individual volumes (Table 3) was at the lower end of the we used this target and a fixed dose of 2 g q8h as a 30-min
arithmetic means reported in the literature. The nonparamet- infusion, we obtained a comparable breakpoint of 3 mg/liter in
ric estimation algorithm in NPAG yielded larger volumes of CF patients. Daily doses of up to 12 g ceftazidime split into 3
distribution at steady state (Table 3), which were well compa- or 4 intermittent doses have been recommended for CF pa-
rable with literature estimates. Our results on terminal half-life tients (24), whereas other authors recommend lower daily
(Table 2) were in good agreement with the shorter terminal doses of 6 g in adult CF patients (30). To increase the PK-PD
half-life of 1.5 ⫾ 0.2 h in CF patients compared to 1.76 ⫾ 0.2 h MIC breakpoints and clinical outcome, continuous infusion of
in healthy volunteers reported by Leeder et al. (44). The re- ceftazidime has been proposed and studied in CF patients (8,
ported average half-lives in juvenile to adult CF patients range 13, 23, 36, 42, 59, 74–76).
between 1 and 2 h (41, 47, 50, 56, 58, 68, 72, 76). Our MCS assessed the benefit of continuous and prolonged
As our CF patients were smaller and leaner than our healthy infusion in comparison to short-term infusions, all at a daily
volunteers (Table 1), we evaluated various body size models to dose of 6 g/70 kg WT. Both prolonged infusion regimens
describe differences in average PK parameters using popula- achieved PK-PD MIC breakpoints of 8 to 12 mg/liter in CF
tion PK modeling. The differences in average PK parameters patients, which were approximately 10 times higher than the
were better explained by FFM than by WT (Table 4), most breakpoint for short-term infusions q8h at the same daily dose.
likely since FFM accounts for body composition whereas WT Giving the same daily dose as a short-term infusion q6h instead
does not. Lean body mass calculated by the Cheymol and of q8h achieved only a PK-PD MIC breakpoint of 2 mg/liter in
James formula (21, 38) yielded results very similar to those of CF patients. This prediction is in excellent agreement with the
FFM (results not shown). In agreement with theoretical con- better outcome for CF patients with resistant or intermediate
siderations (79) and other studies (3), allometric scaling ex- isolates when they received continuous ceftazidime infusion
plained the difference in average clearance between both pa- compared to when they received short-term infusion (both
tient groups better than linear scaling of clearance (Table 4). coadministered with tobramycin) (36, 63) and with results from
Furthermore, allometric scaling by FFM reduced the unex- in vitro PD models (1, 20, 51).
plained BSV of clearance and of both peripheral volumes of In conclusion, we found a 19% lower unscaled total clear-
distribution most (Table 5). This allows one to design dosage ance and a 36% lower volume of distribution at steady state in
regimens based on FFM that achieve target concentrations and CF patients than in healthy volunteers, because our CF pa-
target effects more precisely. tients were smaller and leaner. Allometric scaling by FFM

Downloaded from https://journals.asm.org/journal/aac on 10 August 2021 by 196.191.48.216.


These results are in agreement with studies on the PK of explained the differences in average PK parameters better than
beta-lactams in CF patients and healthy volunteers (16, 17, 62, linear scaling by WT, probably because WT does not account
71, 77) and are comparable to results from Leeder et al. (44) for body composition. Additionally, allometric scaling by FFM
on ceftazidime. Leeder et al. found a 42% increased total reduced the unexplained BSV by 32% for clearance and by 18
clearance (142 ⫾ 17 ml/min/1.73 m2 in CF patients versus to 26% for volume of the peripheral compartments relative to
101 ⫾ 10 ml/min/1.73 m2 in healthy volunteers) and Hedman linear scaling by WT. Dosage regimens based on FFM can
et al. (33) found a 25% increased renal clearance (125 ⫾ 20 therefore achieve target concentrations more precisely. The
ml/min/1.73 m2 in CF patients and 100 ⫾ 9 ml/min/1.73 m2 in PK-PD MIC breakpoint for near-maximal bactericidal activity
healthy volunteers) for ceftazidime. Hedman et al. explained was 1 mg/liter in CF patients for short-term infusions of 2 g/70
this increased renal clearance primarily by an increased glo- kg WT q8h. As alternative modes of administration, prolonged
merular filtration rate in CF patients determined via inulin (5-h) infusion of 2 g/70 kg WT q8h or continuous infusion of 6
clearance. This might have been in part caused by an altered g/70 kg WT/day achieved a PK-PD MIC breakpoint of 8 to 12
glomerulotubular balance due to a primary tubular transport mg/liter in CF patients. More large clinical trials are warranted
defect (7, 11) or by a 16% higher resting energy expenditure in to compare the time course of clinical responses to prolonged
CF patients (73) due to a higher energy need secondary to or continuous infusion with short-term infusion of beta-lac-
chronic lung infection. A potential limitation of our model is tams in CF patients and to compare clinical responses for dose
that it did not account for renal function. selection via an allometric body size model based on FFM with
Our MCS used an allometric body size model based on FFM standard dosing of CF patients as mg/kg WT.
and predicted lower PTAs for CF patients than for healthy
volunteers (Fig. 2). The PK-PD MIC breakpoint was 1 mg/liter ACKNOWLEDGMENTS
in CF patients and 3 mg/liter in healthy volunteers for short-
This work is dedicated to Klaus-Jürgen Hess at the Gymnasium
term infusion of 2 g/70 kg WT q8h for the near-maximal kill Scheinfeld, Germany, who inspired this work by his continuous support
target. The proposed allometric body size model provides an of one of us (J.B.B.).
explanation why our CF patients, who were smaller than our There are no conflicts of interest for any of the authors.
VOL. 54, 2010 CEFTAZIDIME POPULATION PK AND CF PATIENT BREAKPOINTS 1281

REFERENCES 23. David, T. J., and J. Devlin. 1989. Continuous infusion of ceftazidime in cystic
fibrosis. Lancet i:1454–1455.
1. Alou, L., L. Aguilar, D. Sevillano, M. J. Gimenez, O. Echeverria, M. L.
24. Doring, G., S. P. Conway, H. G. Heijerman, M. E. Hodson, N. Hoiby, A.
Gomez-Lus, and J. Prieto. 2005. Is there a pharmacodynamic need for the Smyth, and D. J. Touw. 2000. Antibiotic therapy against Pseudomonas
use of continuous versus intermittent infusion with ceftazidime against aeruginosa in cystic fibrosis: a European consensus. Eur. Respir. J. 16:749–
Pseudomonas aeruginosa? An in vitro pharmacodynamic model. J. Antimi- 767.
crob. Chemother. 55:209–213. 25. Drusano, G. L. 2004. Antimicrobial pharmacodynamics: critical interactions
2. Ambrose, P. G., S. M. Bhavnani, C. M. Rubino, A. Louie, T. Gumbo, A. of ‘bug and drug’. Nat. Rev. Microbiol. 2:289–300.
Forrest, and G. L. Drusano. 2007. Pharmacokinetics-pharmacodynamics of 26. Drusano, G. L., S. L. Preston, C. Hardalo, R. Hare, C. Banfield, D. Andes,
antimicrobial therapy: it’s not just for mice anymore. Clin. Infect. Dis. 44: O. Vesga, and W. A. Craig. 2001. Use of preclinical data for selection of a
79–86. phase II/III dose for evernimicin and identification of a preclinical MIC
3. Anderson, B. J., and N. H. Holford. 2008. Mechanism-based concepts of size breakpoint. Antimicrob. Agents Chemother. 45:13–22.
and maturity in pharmacokinetics. Annu. Rev. Pharmacol. Toxicol. 48:303– 27. Drusano, G. L., H. C. Standiford, B. Fitzpatrick, J. Leslie, P. Tangtatsa-
332. wasdi, P. Ryan, B. Tatem, M. R. Moody, and S. C. Schimpff. 1984. Compar-
4. Anonymous. 1998. Cystic fibrosis foundation patient registry 1997 annual ison of the pharmacokinetics of ceftazidime and moxalactam and their mi-
data report. Cystic Fibrosis Foundation, Bethesda, MD. crobiological correlates in volunteers. Antimicrob. Agents Chemother. 26:
5. Armstrong, G. C., R. Wise, R. M. Brown, and J. Hancox. 1981. Comparison 388–393.
of ceftazidime and cefamandole pharmacokinetics and blister fluid concen- 28. Frederiksen, B., S. Lanng, C. Koch, and N. Hoiby. 1996. Improved survival
trations. Antimicrob. Agents Chemother. 20:356–358. in the Danish center-treated cystic fibrosis patients: results of aggressive
6. Aronoff, S. C., and J. D. Klinger. 1985. Comparison of cefpiramide (HR-810) treatment. Pediatr. Pulmonol. 21:153–158.
and four anti-pseudomonal beta-lactam agents against pseudomonas isolates 29. George, R. B., Y. Cartier, A. G. Casson, and P. Hernandez. 2006. Suppurative
from children with cystic fibrosis. J. Antimicrob. Chemother. 15:545–549. mediastinitis secondary to Burkholderia cepacia in a patient with cystic
7. Arvidsson, A., G. Alvan, and B. Strandvik. 1983. Difference in renal handling fibrosis. Can. Respir. J. 13:215–218.
of cefsulodin between patients with cystic fibrosis and normal subjects. Acta 30. Gibson, R. L., J. L. Burns, and B. W. Ramsey. 2003. Pathophysiology and
Paediatr. Scand. 72:293–294. management of pulmonary infections in cystic fibrosis. Am. J. Respir. Crit.
8. Bakker, W., A. A. Vinks, J. W. Mouton, P. de Jonge, J. G. Verzijl, and H. G. Care Med. 168:918–951.
Heijerman. 1993. Continuous intravenous home treatment of airway infec- 31. Green, B., and S. B. Duffull. 2004. What is the best size descriptor to use for
tions using ceftazidime administration via portable pump in patients with pharmacokinetic studies in the obese? Br. J. Clin. Pharmacol. 58:119–133.
cystic fibrosis; a multicenter study. Ned. Tijdschr. Geneeskd. 137:2486–2491. 32. Harding, S. M., A. J. Monro, J. E. Thornton, A. J. Munro, and M. I. J. Hogg.
9. Bauer, R. J. 2008. S-ADAPT/MCPEM user’s guide (version 1.56). Software 1981. The comparative pharmacokinetics of ceftazidime and cefotaxime in
for Pharmacokinetic, Pharmacodynamic and Population Data Analysis, healthy volunteers. J. Antimicrob. Chemother. 8(Suppl. B):262–272.
Berkeley, CA. 33. Hedman, A., Y. Adan-Abdi, G. Alvan, B. Strandvik, and A. Arvidsson. 1988.
10. Beal, S. L., A. J. Boeckmann, L. B. Sheiner, and the NONMEM Project Influence of the glomerular filtration rate on renal clearance of ceftazidime
Group. 1999. NONMEM users guides, version 5. University of California at in cystic fibrosis. Clin. Pharmacokinet. 15:57–65.
San Francisco, San Francisco, CA. 34. Hoiby, N. 1993. Antibiotic therapy for chronic infection of pseudomonas in
11. Berg, U., E. Kusoffsky, and B. Strandvik. 1982. Renal function in cystic the lung. Annu. Rev. Med. 44:1–10.
fibrosis with special reference to the renal sodium handling. Acta Paediatr. 35. Holford, N. H. 1996. A size standard for pharmacokinetics. Clin. Pharma-
Scand. 71:833–838. cokinet. 30:329–332.
12. Blumer, J. L., R. C. Stern, J. D. Klinger, T. S. Yamashita, C. M. Meyers, A. 36. Hubert, D., E. Le Roux, T. Lavrut, B. Wallaert, P. Scheid, D. Manach, D.
Blum, and M. D. Reed. 1985. Ceftazidime therapy in patients with cystic Grenet, I. Sermet-Gaudelus, S. Ramel, C. Cracowski, A. Sardet, N. Wizla, E.
fibrosis and multiply-drug-resistant pseudomonas. Am. J. Med. 79:37–46. Deneuville, and R. Garraffo. 2009. Continuous versus intermittent infusions
13. Bosso, J. A., C. R. Bonapace, P. A. Flume, and R. L. White. 1999. A pilot of ceftazidime for treating exacerbation of cystic fibrosis. Antimicrob. Agents
study of the efficacy of constant-infusion ceftazidime in the treatment of Chemother. 53:3650–3656.
endobronchial infections in adults with cystic fibrosis. Pharmacotherapy 19: 37. Hüttner, S., J. B. Bulitta, M. Kinzig, U. Holzgrabe, U. Stephan, and F.
620–626. Sörgel. 2009. Population pharmacokinetics and optimized dosage regimens
of ceftazidime in cystic fibrosis patients and healthy volunteers, abstr. 2798.
14. Bulitta, J., S. B. Duffull, M. Kinzig-Schippers, U. Holzgrabe, U. Stephan,
Abstr. Eur. Congr. Clin. Microbiol. Infect. Dis., Helsinki, Finland.
and F. Sörgel. 2005. Cystic fibrosis patients are pharmacokinetically compa-
38. James, W. 1976. Research on obesity. Her Majesty’s Stationery Office, Lon-
rable to healthy volunteers, abstr. 1923. Abstr. 45th Intersci. Conf. Antimi-
don, United Kingdom.
crob. Agents Chemother.

Downloaded from https://journals.asm.org/journal/aac on 10 August 2021 by 196.191.48.216.


39. Janmahasatian, S., S. B. Duffull, S. Ash, L. C. Ward, N. M. Byrne, and B.
15. Bulitta, J. B., S. B. Duffull, C. B. Landersdorfer, G. L. Drusano, M. Kinzig-
Green. 2005. Quantification of lean bodyweight. Clin. Pharmacokinet. 44:
Schippers, U. Holzgrabe, U. Stephan, and F. Sörgel. 2006. Pharmacody-
1051–1065.
namic comparison of cystic fibrosis patients and healthy volunteers by pop-
40. Kemmerich, B., H. Warns, H. Lode, K. Borner, P. Koeppe, and H. Knothe.
ulation pharmacokinetics (PK) and Monte Carlo simulation, poster W4056.
1983. Multiple-dose pharmacokinetics of ceftazidime and its influence on
Abstr. Am. Assoc. Pharm. Sci. Annu. Meet., San Antonio, TX, 29 October to
fecal flora. Antimicrob. Agents Chemother. 24:333–338.
2 November 2006.
41. Kercsmar, C. M., R. C. Stern, M. D. Reed, C. M. Myers, D. Murdell, and
16. Bulitta, J. B., S. B. Duffull, M. Kinzig-Schippers, U. Holzgrabe, U. Stephan, J. L. Blumer. 1983. Ceftazidime in cystic fibrosis: pharmacokinetics and
G. L. Drusano, and F. Sorgel. 2007. Systematic comparison of the population therapeutic response. J. Antimicrob. Chemother. 12(Suppl. A):289–295.
pharmacokinetics and pharmacodynamics of piperacillin in cystic fibrosis 42. Kuzemko, J., and C. Crawford. 1989. Continuous infusion of ceftazidime in
patients and healthy volunteers. Antimicrob. Agents Chemother. 51:2497– cystic fibrosis. Lancet ii:385.
2507. 43. Leary, R., R. W. Jelliffe, A. Schumitzky, and M. Van Guilder. 2001. An
17. Bulitta, J. B., S. B. Duffull, C. B. Landersdorfer, M. Kinzig, U. Holzgrabe, U. adaptive grid non-parametric approach to pharmacokinetic and dynamic
Stephan, G. L. Drusano, and F. Sorgel. 2009. Comparison of the pharma- (PK/PD) models. IEEE Computer Society, Bethesda, MD.
cokinetics and pharmacodynamic profile of carumonam in cystic fibrosis 44. Leeder, J. S., M. Spino, A. F. Isles, A. M. Tesoro, R. Gold, and S. M.
patients and healthy volunteers. Diagn. Microbiol. Infect. Dis. 65:130–141. MacLeod. 1984. Ceftazidime disposition in acute and stable cystic fibrosis.
18. Bulitta, J. B., C. B. Landersdorfer, M. Kinzig, U. Holzgrabe, and F. Sorgel. Clin. Pharmacol. Ther. 36:355–362.
2009. New semiphysiological absorption model to assess the pharmacody- 45. Livermore, D. M., S. Mushtaq, Y. Ge, and M. Warner. 2009. Activity of
namic profile of cefuroxime axetil using nonparametric and parametric pop- cephalosporin CXA-101 (FR264205) against Pseudomonas aeruginosa and
ulation pharmacokinetics. Antimicrob. Agents Chemother. 53:3462–3471. Burkholderia cepacia group strains and isolates. Int. J. Antimicrob. Agents
19. Canton, R., N. Cobos, J. de Gracia, F. Baquero, J. Honorato, S. Gartner, A. 34:402–406.
Alvarez, A. Salcedo, A. Oliver, and E. Garcia-Quetglas. 2005. Antimicrobial 46. Manno, G., M. Cruciani, L. Romano, S. Scapolan, M. Mentasti, R. Lorini,
therapy for pulmonary pathogenic colonisation and infection by Pseudomo- and L. Minicucci. 2005. Antimicrobial use and Pseudomonas aeruginosa
nas aeruginosa in cystic fibrosis patients. Clin. Microbiol. Infect. 11:690–703. susceptibility profile in a cystic fibrosis centre. Int. J. Antimicrob. Agents
20. Cappelletty, D. M., S. L. Kang, S. M. Palmer, and M. J. Rybak. 1995. 25:193–197.
Pharmacodynamics of ceftazidime administered as continuous infusion or 47. Martini, N., M. Agostini, G. Barlocco, L. Bozzini, L. Castellani, A. Messori,
intermittent bolus alone and in combination with single daily-dose amikacin G. Scroccaro, and G. Mastella. 1984. Serum and sputum concentrations of
against Pseudomonas aeruginosa in an in vitro infection model. Antimicrob. azlocillin, cefoperazone and ceftazidime in patients with cystic fibrosis.
Agents Chemother. 39:1797–1801. J. Clin. Hosp. Pharm. 9:303–309.
21. Cheymol, G. 2000. Effects of obesity on pharmacokinetics: implications for 48. Miller, M. E., and D. M. Kornhauser. 1994. Bromide pharmacokinetics in
drug therapy. Clin. Pharmacokinet. 39:215–231. cystic fibrosis. Arch. Pediatr. Adolesc. Med. 148:266–271.
22. Craig, W. A. 1998. Pharmacokinetic/pharmacodynamic parameters: rationale 49. Morgan, D. J., and K. M. Bray. 1994. Lean body mass as a predictor of drug
for antibacterial dosing of mice and men. Clin. Infect. Dis. 26:1–12. dosage. Implications for drug therapy. Clin. Pharmacokinet. 26:292–307.
1282 BULITTA ET AL. ANTIMICROB. AGENTS CHEMOTHER.

50. Mouton, J., A. Horrevorts, S. Overbeek, et al. 1993. Pharmacokinetics of 66. Sörgel, F., U. Stephan, C. Stehr, R. Seelmann, M. Rey, U. Koch, H. G.
ceftazidime during continuous and intermittent infusion in adult cystic fi- Wiesemann, H. C. Dominick, and B. Gottschalk. 1986. High dose treatment
brosis patients, p. 88–99. In J. W. Mouton (ed.), Pharmacokinetic and phar- with antibiotics in patients with cystic fibrosis (CF)?, abstr. 614. Abstr. 26th
macodynamic studies of betalactam antibiotics in volunteers and patients Intersci. Conf. Antimicrob. Agents Chemother., New Orleans, LA.
with cystic fibrosis. University of Rotterdam, Rotterdam, the Netherlands. 67. Sorgel, F., U. Stephan, H. G. Wiesemann, B. Gottschalk, C. Stehr, M. Rey,
51. Mouton, J. W., and J. G. den Hollander. 1994. Killing of Pseudomonas H. B. Bowing, H. C. Dominick, and M. Geldmacher von Mallinckrodt. 1987.
aeruginosa during continuous and intermittent infusion of ceftazidime in an High dose treatment with antibiotics in cystic fibrosis—a reappraisal with
in vitro pharmacokinetic model. Antimicrob. Agents Chemother. 38:931– special reference to the pharmacokinetics of beta-lactams and new fluoro-
936. quinolones in adult CF-patients. Infection 15:385–396.
52. Mouton, J. W., A. M. Horrevorts, P. G. Mulder, E. P. Prens, and M. F. 68. Strandvik, B., A. S. Malmborg, H. Alfredson, and A. Ericsson. 1983. Clinical
Michel. 1990. Pharmacokinetics of ceftazidime in serum and suction blister results and pharmacokinetics of ceftazidime treatment in patients with cystic
fluid during continuous and intermittent infusions in healthy volunteers. fibrosis. J. Antimicrob. Chemother. 12(Suppl. A):283–287.
Antimicrob. Agents Chemother. 34:2307–2311. 69. Taccetti, G., S. Campana, and L. Marianelli. 1999. Multiresistant non-
53. Mouton, J. W., N. Punt, and A. A. Vinks. 2005. A retrospective analysis using fermentative gram-negative bacteria in cystic fibrosis patients: the results of
Monte Carlo simulation to evaluate recommended ceftazidime dosing regi- an Italian multicenter study. Italian Group for Cystic Fibrosis Microbiology.
mens in healthy volunteers, patients with cystic fibrosis, and patients in the Eur. J. Epidemiol. 15:85–88.
intensive care unit. Clin. Ther. 27:762–772. 70. Tjandramaga, T. B., A. Van Hecken, A. Mullie, R. Verbesselt, P. J. De
54. Norrby, S. R. 1983. Ceftazidime in clinical practice—a summary. J. Antimi- Schepper, and L. Verbist. 1982. Comparative pharmacokinetics of ceftazi-
crob. Chemother. 12(Suppl. A):405–408. dime and moxalactam. Antimicrob. Agents Chemother. 22:237–241.
55. Paap, C. M., and M. C. Nahata. 1990. Clinical pharmacokinetics of antibac- 71. Touw, D. J., A. A. Vinks, J. W. Mouton, and A. M. Horrevorts. 1998. Phar-
terial drugs in neonates. Clin. Pharmacokinet. 19:280–318. macokinetic optimisation of antibacterial treatment in patients with cystic
56. Padoan, R., A. Brienza, R. M. Crossignani, G. Lodi, A. Giunta, B. M. Assael, fibrosis. Current practice and suggestions for future directions. Clin. Phar-
F. Granata, E. Passarella, P. A. Vallaperta, and L. Xerri. 1983. Ceftazidime macokinet. 35:437–459.
in treatment of acute pulmonary exacerbations in patients with cystic fibrosis. 72. Turner, A., S. J. Pedler, F. Carswell, G. R. Spencer, and D. C. Speller. 1984.
J. Pediatr. 103:320–324. Serum and sputum concentrations of ceftazidime in patients with cystic
57. Paulfeuerborn, W., H. J. Muller, K. Borner, P. Koeppe, and H. Lode. 1993. fibrosis. J. Antimicrob. Chemother. 14:521–527.
Comparative pharmacokinetics and serum bactericidal activities of SCE- 73. Vaisman, N., P. B. Pencharz, M. Corey, G. J. Canny, and E. Hahn. 1987.
2787 and ceftazidime. Antimicrob. Agents Chemother. 37:1835–1841. Energy expenditure of patients with cystic fibrosis. J. Pediatr. 111:496–500.
58. Permin, H., C. Koch, N. Hoiby, H. O. Christensen, A. F. Moller, and S. 74. Viaene, E., H. Chanteux, H. Servais, M. P. Mingeot-Leclercq, and P. M.
Moller. 1983. Ceftazidime treatment of chronic Pseudomonas aeruginosa Tulkens. 2002. Comparative stability studies of antipseudomonal beta-lac-
respiratory tract infection in cystic fibrosis. J. Antimicrob. Chemother. tams for potential administration through portable elastomeric pumps
12(Suppl. A):313–323. (home therapy for cystic fibrosis patients) and motor-operated syringes (in-
59. Rappaz, I., L. A. Decosterd, J. Bille, M. Pilet, N. Belaz, and M. Roulet. 2000. tensive care units). Antimicrob. Agents Chemother. 46:2327–2332.
Continuous infusion of ceftazidime with a portable pump is as effective as 75. Vinks, A. A., R. W. Brimicombe, H. G. Heijerman, and W. Bakker. 1997.
thrice-a-day bolus in cystic fibrosis children. Eur. J. Pediatr. 159:919–925. Continuous infusion of ceftazidime in cystic fibrosis patients during home
60. Reed, M. 1997. The pathophysiology and treatment of cystic fibrosis. J. Pe- treatment: clinical outcome, microbiology and pharmacokinetics. J. Antimi-
diatr. Pharm. Pract. 2:285–308. crob. Chemother. 40:125–133.
61. Reed, M. D., R. C. Stern, C. A. O’Brien, D. A. Crenshaw, and J. L. Blumer. 76. Vinks, A. A., D. J. Touw, H. G. Heijerman, M. Danhof, G. P. de Leede, and
1987. Randomized double-blind evaluation of ceftazidime dose ranging in W. Bakker. 1994. Pharmacokinetics of ceftazidime in adult cystic fibrosis
hospitalized patients with cystic fibrosis. Antimicrob. Agents Chemother. patients during continuous infusion and ambulatory treatment at home.
31:698–702. Ther. Drug Monit. 16:341–348.
62. Rey, E., J. M. Treluyer, and G. Pons. 1998. Drug disposition in cystic fibrosis. 77. Vinks, A. A., R. N. van Rossem, R. A. Mathot, H. G. Heijerman, and J. W.
Clin. Pharmacokinet. 35:313–329. Mouton. 2007. Pharmacokinetics of aztreonam in healthy subjects and pa-
63. Riethmueller, J., S. Junge, T. W. Schroeter, K. Kuemmerer, P. Franke, M. tients with cystic fibrosis and evaluation of dose-exposure relationships using
Ballmann, A. Claass, S. Broemme, R. Jeschke, A. Hebestreit, D. Staab, K. Monte Carlo simulation. Antimicrob. Agents Chemother. 51:3049–3055.
Koetz, G. Doering, and M. Stern. 15 September 2009. Continuous vs thrice- 78. Walstad, R. A., K. B. Hellum, S. Blika, L. G. Dale, T. Fredriksen, K. I.
daily ceftazidime for elective intravenous antipseudomonal therapy in cystic Myhre, and G. R. Spencer. 1983. Pharmacokinetics and tissue penetration of
fibrosis. Infection. doi:10.1007/s15010-009-8116-5. ceftazidime: studies on lymph, aqueous humour, skin blister, cerebrospinal
64. Robinson, C. A., R. J. Kuhn, J. Craigmyle, M. I. Anstead, and J. E. Kanga. and pleural fluid. J. Antimicrob. Chemother. 12(Suppl. A):275–282.
2001. Susceptibility of pseudomonas aeruginosa to cefepime versus ceftazi- 79. West, G. B., J. H. Brown, and B. J. Enquist. 1999. The fourth dimension of

Downloaded from https://journals.asm.org/journal/aac on 10 August 2021 by 196.191.48.216.


dime in patients with cystic fibrosis. Pharmacotherapy 21:1320–1324. life: fractal geometry and allometric scaling of organisms. Science 284:1677–
65. Sörgel, F., U. Stephan, H. C. Dominick, H. G. Wiesemann, H. B. Böwing, and 1679.
C. Stehr. 1985. Pharmacokinetics of ceftazidime in patients with cystic fibro- 80. West, G. B., J. H. Brown, and B. J. Enquist. 1997. A general model for the
sis, abstr. 24. Abstr. Eur. Workshop Cystic Fibrosis, Jerusalem, Israel. origin of allometric scaling laws in biology. Science 276:122–126.

You might also like