You are on page 1of 10

ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, Aug. 2009, p. 34623471 Vol. 53, No.

8
0066-4804/09/$08.000 doi:10.1128/AAC.00054-09
Copyright 2009, American Society for Microbiology. All Rights Reserved.

New Semiphysiological Absorption Model To Assess the Pharmacodynamic


Profile of Cefuroxime Axetil Using Nonparametric and Parametric
Population Pharmacokinetics
J. B. Bulitta,1 C. B. Landersdorfer,1 M. Kinzig,1 U. Holzgrabe,2 and F. Sorgel1,3*
Institute for Biomedical and Pharmaceutical Research (IBMP), Nurnberg-Heroldsberg, Germany1; Institute of Pharmacy and
Food Chemistry, University of Wurzburg, Wurzburg, Germany2; and Department of Pharmacology, University of
DuisburgEssen, Essen, Germany3

Downloaded from http://aac.asm.org/ on February 5, 2017 by GLAXOSMITHKLINE


Received 14 January 2009/Returned for modification 26 March 2009/Accepted 3 June 2009

Cefuroxime axetil is widely used to treat respiratory tract infections. We are not aware of a population
pharmacokinetic (PK) model for cefuroxime axetil. Our objectives were to develop a semiphysiological popu-
lation PK model and evaluate the pharmacodynamic profile for cefuroxime axetil. Twenty-four healthy volun-
teers received 250 mg oral cefuroxime as a suspension after a standardized breakfast. Liquid chromatography-
tandem mass spectrometry was used for drug analysis, NONMEM and S-ADAPT (results reported) were used
for parametric population PK modeling, and NPAG was used for nonparametric population PK modeling.
Monte Carlo simulations were used to predict the duration for which the non-protein-bound-plasma concen-
tration was above the MIC (fT>MIC). A model with one disposition compartment, a saturable and time-
dependent drug release from the stomach, and fast drug absorption from the intestine yielded precise (r >
0.992) and unbiased curve fits and an excellent predictive performance. The apparent clearance was 21.7
liters/h (19.8% coefficient of variation [CV]) and the volume of distribution 38.7 liters (18.3% CV). Robust
(>90%) probabilities of target attainment (PTAs) were achieved by 250 mg cefuroxime given every 12 h (q12h)
or q8h for MICs of <0.375 mg/liter or <0.5 mg/liter, respectively, for the bacteriostasis target fT>MIC of >40%
and for MICs of <0.094 mg/liter or <0.375 mg/liter, respectively, for the near-maximal-killing target fT>MIC
of >65%. For the >40% fT>MIC target, the PTAs for 250 mg cefuroxime q12h were >97.8% for Streptococcus
pyogenes and penicillin-susceptible Streptococcus pneumoniae. Cefuroxime at 250 mg q12h or q8h achieved PTAs
below 73% or 92%, respectively, for Haemophilus influenzae, Moraxella catarrhalis, and penicillin-intermediate
S. pneumoniae for susceptibility data from various countries. Depending on the MIC distribution, 250 mg oral
cefuroxime q8h instead of q12h should be considered, especially for more-severe infections that require
near-maximal killing by cefuroxime.

Cefuroxime axetil is the acetoxyethyl-ester prodrug of cefu- are 1 mg/liter for S. pneumoniae and 4 mg/liter for Hae-
roxime. Cefuroxime axetil is reliably absorbed and can be taken mophilus spp., Enterobacteriaceae, and Staphylococcus spp.
with or without a meal, although its extent of bioavailability is Several authors (31, 37, 40) determined the pharmacoki-
enhanced under the influence of food (20, 54). Cefuroxime has netic-pharmacodynamic (PK-PD) MIC breakpoint for cefu-
been successfully used in the treatment of upper and lower roxime axetil on the basis of the average plasma concentration
respiratory tract infections as well as genitourinary tract profiles but did not incorporate between-subject variability
infections (45) and is active against Haemophilus influenzae, (BSV) in their analysis. Ambrose et al. (2) determined the
Moraxella catarrhalis, Streptococcus pyogenes, Klebsiella pneu- PK-PD MIC breakpoint for intravenous cefuroxime via Monte
moniae, Neisseria gonorrhoeae, penicillin-susceptible Strepto- Carlo simulation (MCS) based on literature data, and Viberg
coccus pneumoniae, and some isolates of penicillin-intermedi- et al. (51, 52) developed a population PK model for intrave-
ate S. pneumoniae (6, 7, 2527, 34, 35, 37, 38, 41, 55). nous cefuroxime. We are not aware of a population PK model
A susceptibility breakpoint of 1 mg/liter has been determined or MCS for cefuroxime axetil.
for cefuroxime by national organizations in Britain (BSAC) (8)
Population PK and the MCS methodology account for the
and Germany (DIN) (17). The susceptibility breakpoints from
BSV in PK parameters and for the variability in the bacterial
the Clinical and Laboratory Standards Institute (CLSI) (11)
susceptibility. A PK-PD target is used as a surrogate measure
to predict successful microbiological or clinical outcome (13,
18, 22, 28, 29, 48). For beta-lactams, the duration for which the
* Corresponding author. Mailing address: Institute for Biomedical non-protein-bound-plasma concentration exceeds the MIC
and Pharmaceutical Research (IBMP), Paul-Ehrlich-Str. 19, D-90562 (fTMIC) best predicts these outcomes (3, 14, 18). For cepha-
Nurnberg-Heroldsberg, Germany. Phone: 49-911-518290. Fax: 49-911-
5182920. E-mail: ibmp@osn.de. losporins, data from animal infection models showed that a
This article is dedicated to Ulrich Stephan, the cofounder of target fTMIC of 40% correlates with bacteriostasis at 24 h and
IBMP, who passed away on 6 February 2009. Without his inspiration that an fTMIC of 60 to 70% is required for near-maximal
and support, IBMP would not exist, and neither would the present
bactericidal activity at 24 h (3, 12, 14, 18). On the basis of these
research have been performed. We keep him in our hearts.
Present address: Ordway Research Institute, Albany, NY 12208. PK-PD targets, MCS can predict the probability of target at-

Published ahead of print on 15 June 2009. tainment (PTA) at various MICs. If the PTA-versus-MIC pro-

3462
VOL. 53, 2009 POPULATION PK AND PD OF CEFUROXIME AXETIL 3463

file is combined with the expected MIC distribution of the


pathogen(s) of interest in a local hospital, the probability of
successful microbiological or clinical outcome can be pre-
dicted.
In addition to increasing the extent of bioavailability (20,
54), administration after a meal may cause a lower rate of
cefuroxime absorption due to a prolonged gastric transit time.
The rate of gastric emptying after a high-fat meal is likely to be
variable and may change over time. Parameters describing the
absorption phase may also not be normally or log-normally
distributed. As MCS based on parametric population PK mod-

Downloaded from http://aac.asm.org/ on February 5, 2017 by GLAXOSMITHKLINE


els uses parametric distributions to describe the variability in FIG. 1. Structure of the final PK model. For the simulation of
PK parameters, we additionally applied nonparametric popu- Vmax/Vmax 0 profiles, Emax values of 3 (long dashed line), 1 (continuous
line), or 0.5 (dotted line); a TC50 of 2.5 h; and a Hill coefficient of 10
lation PK modeling. The latter offers the advantage that it does were used. CL, clearance. (See Table 2 and Materials and Methods for
not assume any shape for the multivariate distribution of PK further explanations of the parameters.)
parameters. However, sample sizes larger than 24 subjects may
be required to adequately describe the shape of the multivar-
iate distribution by a nonparametric variability model. metry C8), eluted with an isocratic solvent system consisting of ammonium
Our first objective was to develop a semiphysiological acetate buffer and acetonitrile (70%-30%, vol/vol), and monitored by liquid
chromatography-tandem mass spectrometry with a multiple-reaction-monitoring
population PK model for cefuroxime axetil by using para-
method as follows: precursor 3 product ion for cefuroxime (m/z 423 3 m/z 207)
metric and nonparametric population PK methods. Second, and an internal standard (m/z 426 3 m/z 156). Both analyses were in negative
we sought to determine the PTA-versus-MIC profiles and mode. Under these conditions, cefuroxime and the internal standard were eluted
the PTAs for specific MIC distributions of various patho- after approximately 1.4 and 1.5 min. The Mac Quan software program (version
gens for dosage regimens with oral cefuroxime given every 1.5; PE Sciex, Thornhill, Ontario, Canada [1991 to 1997]) was used for evaluation
of chromatograms.
12 h (q12h) or q8h. The linearity of the cefuroxime calibration curve was shown from 0.00900
(This work was presented in part at the 46th Interscience mg/liter to 10.2 mg/liter. The coefficient of correlation for all measured se-
Conference on Antimicrobial Agents and Chemotherapy, 2006 quences of cefuroxime was at least 0.999. The lowest calibration standard of
[8a].) 0.00900 mg/liter was set as the lower limit of quantification of the assay for
cefuroxime in human plasma. There was no observation below this quantification
limit. For the spiked quality control standards of cefuroxime, the interday pre-
MATERIALS AND METHODS cision ranged from 3.2 to 5.0%, with interday accuracy (relative error) between
Subjects. Twenty-four healthy male Caucasian volunteers participated in the 4.3 and 2.1%. The intraday precision and relative error of the cefuroxime assay
study after they had given their written informed consent. Their demographic ranged from 0.7 to 4.0% and from 0.1 to 3.4%, respectively.
data were as follows (means standard deviations [SD]): age, 24.5 3.3 years Population PK analysis. (i) Computation. We applied the first-order condi-
(range, 18 to 31 years); weight, 73.8 9.2 kg (range, 58.2 to 93.6 kg); and height, tional estimation method with the interaction estimation option in NONMEM,
179 8.0 cm (range, 166 to 193 cm). The subjects health statuses were assessed version VI, release 1.2 (NONMEM Project Group, University of California, San
by physical examination, electrocardiography, and laboratory tests, including Francisco) (5). Initial models were developed in NONMEM V. Model develop-
urinalysis and screening for drugs of abuse. Intake of food and fluid was strictly ment was primarily performed in NONMEM. The final population PK model
standardized during the study days. Consumption of tobacco, methylxanthines, was additionally estimated in S-ADAPT (version 1.55; parallelized on a com-
and alcohol in any form was prohibited from 12 h before administration of the puter cluster), using the importance-sampling parametric Monte Carlo expecta-
study drug until acquisition of the last sample. The volunteers were closely tion maximization method (with a pmethod value of 8 in S-ADAPT) (4) and by
observed by physicians for the occurrence of adverse events on the study days. the nonparametric adaptive grid (NPAG) algorithm implemented in the
The study protocol had been approved by the local ethics committee, and the USC*PACK program (version 12.00) (33). WinNonlin Professional (version
study was conducted by following the revised version of the Declaration of 4.0.1; Pharsight Corp., Mountain View, CA) was used for noncompartmental
Helsinki. analysis and statistics.
Study design and drug administration. The study was a single-dose, single- Parameter uncertainty was assessed by standard asymptotic formulas in S-
center study. All subjects received an oral suspension of 300.72 mg cefuroxime ADAPT (4). As NONMEM could not compute asymptotic standard errors in the
axetil (equivalent to 250 mg cefuroxime) with 240 ml low-carbonated, calcium- $COV step for the final model, nonparametric bootstrap methods with 100
poor mineral water at room temperature. The study drug was administered replicates were used to calculate standard errors in NONMEM as described
directly after intake of a standardized breakfast with a significant amount of fat. previously (9).
This breakfast contained four slices of crisp bread (50 g), 20 g margarine, 2 slices (ii) Structural model. We considered one- and two-compartment disposition
of cheese (40 g; 30% fat content), 25 g jam, 100 ml fruit tea, and 100 ml milk models with first-order, zero-order, or mixed-order (Michaelis-Menten) absorp-
(3.5% fat content). tion with or without a lag time. Additionally, a semiphysiological model with a
Blood sampling. All blood samples were drawn in heparinated tubes from a time-dependent release from the stomach to the intestine and subsequent ab-
forearm vein via an intravenous catheter. Blood samples were drawn immedi- sorption into the central compartment was developed (Fig. 1). The differential
ately before administration and at 30, 60, and 90 min and at 2, 2.33, 2.67, 3, 3.33, equations for this model are as follows:
3.67, 4, 4.5, 5, 6, 8, 10, and 12 h after administration of the study drug. Samples dA1 Vmax A1
were immediately centrifuged and immediately frozen and stored at 70C until (1)
dt Km A1
analysis.
Drug analysis. Samples were analyzed by means of a liquid chromatography- dA2 Vmax A1
tandem mass spectrometry method validated for 0.1-ml samples of human kabs A2 (2)
dt Km A1
plasma. Plasma samples (0.1 ml) were diluted with buffer containing the internal
standard and deproteinized by addition of 400 l of acetonitrile. After thorough dA3 CL
mixing, the samples were centrifuged for 5 min at 3,600 rpm at approximately kabs A2 A3 (3)
dt V
4C, and acetonitrile was removed by extraction with 1 ml dichloromethane.
The mixture was centrifuged again, and 30 l of the aqueous phase of each where A1 is the amount of drug in the stomach, A2 is the amount of drug in the
sample was then chromatographed on a reversed-phase column (Waters Sym- intestine, and A3 is the amount in the central compartment (see Table 2 for
3464 BULITTA ET AL. ANTIMICROB. AGENTS CHEMOTHER.

parameter explanations). All initial conditions for all three compartments were TABLE 1. PK parameters for noncompartmental analysis for
zero. The stomach compartment (A1) received a bolus dose of 250 mg cefu- 250 mg oral cefuroxime given as cefuroxime axetil
roxime at zero hour. The model is simplified, as it did not contain a specific
compartment for the prodrug cefuroxime axetil. It was assumed that cefuroxime Parameter Avg SDa Median (range)
axetil is converted to cefuroxime before the ester prodrug reaches the peripheral
Area under the curve from 11.9 2.49 11.6 (8.4918.1)
sampling site. This assumption is considered justifiable for an ester prodrug. The
time zero to infinity
maximum rate of release (Vmax) of drug from the stomach compartment was
(mg/h/liter)
described by a time-dependent function:
Peak concn (mg/liter) 2.64 0.64 2.54 (1.653.90)

VmaxTPM Vmax 0 1
Emax TPM
TC50 TPM (4)
Time of peak concn (h)
Terminal half-life (h)
Apparent total clearance
2.98 0.73
1.34 0.13
21.8 4.29
2.83 (2.005.00)
1.35 (1.081.54)
21.5 (13.829.4)
Time is denoted as time past meal (TPM). Starting from a Vmax at time zero (liters/h)
(Vmax 0), the Hill coefficient () modifies the Vmax over time, with TC50 denoting Apparent vol of distribution at 54.1 15.7 51.2 (34.7102)
steady statea (liters)

Downloaded from http://aac.asm.org/ on February 5, 2017 by GLAXOSMITHKLINE


the time past the meal at which Vmax changed by 50% and Emax the extent of the
change in Vmax. For TPM values of TC50, the Vmax approaches Vmax 0 (1 Apparent vol of distribution 41.7 7.64 44.4 (27.454.9)
Emax). Therefore, an Emax of 1 represents complete inhibition of gastric re- during the terminal phase
lease, an Emax of 0 an unchanged maximum rate of gastric release, and an Emax (liters)
of 1 a maximum rate of gastric release twice as fast. Time of total concn above 4.97 0.79 4.90 (3.437.08)
Competing models were discriminated by their predictive performance as- 1 mg/liter (h)
sessed via visual predictive checks (VPCs), their objective function (or log like- Time of total concn above 6.90 0.77 6.75 (5.139.02)
lihood), and standard diagnostic plots. 0.5 mg/liter (h)
(iii) Parameter variability and observation model. For parametric population a
To calculate the mean residence time after intravenous bolus administration,
PK modeling in NONMEM and S-ADAPT, we estimated the BSV of PK pa-
mean input time was estimated as 0.5 times the time of peak concentration,
rameters by assuming a log-normal distribution. The maximum extent of inhibi- assuming approximately zero-order kinetics of drug input.
tion or stimulation of gastric release for the ith subject (Emax i) was constrained
to a lower bound of 1 by the following logit transformation:

Emax i 1 10
expLg_Emax BSV Emax i
1 expLg_Emax BSV Emax i (5)
2002 and 2003 for H. influenzae (n 581), M. catarrhalis (n 269), and S.
pneumoniae (n 519); susceptibility data from Canada (55) collected in 2001
and 2002 for H. influenzae (n 1,350); and susceptibility data from Germany (7)
Lg_Emax is the estimated population mean (arithmetic mean) on the transformed collected in 2002 for H. influenzae (n 300), M. catarrhalis (n 308), S.
scale and BSV Emax i the random deviate for the ith subject on transformed scale. pneumoniae (n 331), and S. pyogenes (n 340). Additionally, we used sus-
This transformation ensures that all Emax i values range from 1 to 9. A sensi- ceptibility data from a global surveillance study (6) collected between 1997 and
tivity analysis showed that the upper limit of 9 did not affect the curve fits or 2000 for penicillin-susceptible S. pneumoniae (n 2,102) and penicillin-inter-
predictive performance of this model. mediate S. pneumoniae (n 1,024); susceptibility data from a European surveil-
Plasma concentration time profiles were simulated for at least 4,800 subjects lance study (26) collected between 1997 and 1999 for S. pneumoniae (n 2,018)
for each competing model to calculate the median and nonparametric 80% and S. pyogenes (n 662); and susceptibility data from North America (25)
prediction interval (10th to 90th percentile) for the predicted concentrations. collected between 1997 and 1999 for S. pyogenes (n 119), S. pneumoniae (n
The same percentiles were calculated for the observations to visually assess 417), H. influenzae (n 300), and M. catarrhalis (n 231). The PTA expectation
whether the simulated percentiles closely matched the percentiles of the obser- values were also calculated for the MIC distributions for H. influenzae (n
vations. For nonparametric population PK models in NPAG, this VPC was 66,947), K. pneumoniae (n 34,629), M. catarrhalis (n 14,308), Staphylococcus
performed either based on the nonparametric distribution of PK parameters aureus (n 10,620), N. gonorrhoeae (n 655), and Neisseria meningitidis (n
characterized by the support point matrix or based on a parametric, multivariate 257) on the basis of the multinational database of the European Committee on
log-normal distribution of PK parameters. In all three programs full variance- Antimicrobial Susceptibility Testing (EUCAST) (http://www.eucast.org/mic
covariance matrices were estimated and used for MCS. _distributions_of_wild_type_microorganisms/).
The residual unidentified variability was described by a combined additive and
proportional error model. We used the adaptive gamma option in NPAG to
estimate the residual error described by the assay error polynomial. RESULTS
(iv) MCS. A target fTMIC of 60 to 70% has been identified for near-maximal
bactericidal activity of cephalosporins, and a target of 40% is required for The PK parameters from noncompartmental analysis (Table
bacteriostasis (14, 18). Therefore, we used PK-PD target fTMICs of 65% for 1) were in good agreement with the literature (41, 45). We
near-maximal bactericidal activity and 40% for bacteriostasis. A range of MICs found (average SD) a terminal half-life of 1.34 0.13 h and
from 0.031 to 64 mg/liter was considered. As the protein binding levels for
a peak concentration of 2.64 0.64 mg/liter between 2 and 5 h
cefuroxime have been reported to range between 33 and 50% (21, 23, 45), we
assumed an average protein binding of 42% for cefuroxime. postdose. The variability in terminal half-life (9.4% coefficient
We compared dosage regimens of 250 mg and 500 mg oral cefuroxime given of variation) was lower than the variabilities in apparent clear-
q12h and q8h at steady state. For the final population PK models in NONMEM, ance (20%), peak concentration (24%), and time of peak
S-ADAPT, and NPAG, we simulated 10,000 subjects for each dosage regimen in (24%).
the absence of residual error. NONMEM was used to simulate the full concen-
tration time profiles at steady state with very frequent sampling based on the final
A biphasic absorption pattern was found for 5 of 24 subjects
population PK model and the estimated full variance-covariance matrix. The and a plateau-like peak for 8 of 24 subjects (Fig. 2). These
fTMIC values were calculated by linear interpolation between simulated time shapes could not be described by standard first-order or zero-
points as previously described (9). order absorption models that included a lag time. Compared to
The PTA was estimated by calculating the fraction of subjects who attained the
the objective function value for the final semiphysiological
PK-PD target at each MIC. The highest MIC with a PTA of at least 90% was
used as the PK-PD MIC breakpoint. model, the objective function differences in NONMEM were
To put these PTAs into clinical perspective, we calculated the PTA expecta- 721 points for the first-order absorption model with a lag time,
tion value (39) for successful treatment against pathogens from specific MIC 359 points for the zero-order absorption model with a lag time,
distributions as described previously (9). The PTA expectation value is the PTA and 189 points for the Michaelis-Menten absorption model
for treatment of infections caused by bacteria from a specific MIC distribution
(ideally the MIC distribution of each local hospital).
with a lag time (likelihood ratio test: P 0.0001 for all com-
The PTA expectation value was calculated based on published MIC distribu- parisons). The semiphysiological absorption model with a two-
tions. We used susceptibility data from the United Kingdom (38) collected in compartment disposition model had a 25-point-lower objective
VOL. 53, 2009 POPULATION PK AND PD OF CEFUROXIME AXETIL 3465

Downloaded from http://aac.asm.org/ on February 5, 2017 by GLAXOSMITHKLINE


FIG. 2. Individual curve fits from NONMEM (dashed line), S-ADAPT (dotted line), and NPAG (continuous line) overlaid on observations
(markers).

function than the same absorption model with one disposition more notably stimulated, since the median (90th percentile) of
compartment. As the latter model showed precise curve fits individual Emax values was 1.82 (5.55). NPAG estimated the
and an excellent predictive performance, we chose the simpler release from the stomach to the intestine primarily as a first-
model as the final model. order process (Km/dose, 343%), and stimulation of gastric
The individual curve fits for the semiphysiological model emptying was more pronounced than for S-ADAPT. The mean
(Fig. 1) were excellent in all three programs (Fig. 2). The TC50 values for the rate of gastric emptying after a standard-
model was flexible enough to fit profiles with sharp peaks, ized breakfast were 1.61 h in NONMEM and S-ADAPT and
plateau-like peaks, and dual peaks. No estimation algorithm 2.08 h in NPAG. Individual TC50 estimates were variable (Ta-
(program) provided the best fit for every subject. The linear ble 2).
regression plot of individual fitted versus observed concentra- The VPCs (Fig. 3) indicated that the nonparametric simu-
tions had a slope of 1.007 and an intercept of 0.014 mg/liter lation based on the support points from NPAG yielded the
in NONMEM (r 0.9941), a slope of 1.011 and an intercept of closest match between predicted and observed concentrations.
0.011 mg/liter in S-ADAPT (r 0.9928), and a slope of 1.003 This was expected, since this simulation is based on the non-
and an intercept of 0.011 mg/liter in NPAG (r 0.9935). parametric distribution of PK parameters that yielded precise
The final estimates (Table 2 and Table 3) were precise. The and unbiased fits for all 24 concentration-time profiles. The
relative standard errors were 31% or less for all population parametric simulations based on the estimates from S-ADAPT
means (except for Km, 70% in NONMEM and 39% in S- and NPAG matched the median and 10th to 90th percentiles
ADAPT) and 41% or less for all BSV estimates. The estimates for the observations more closely between approximately 1 and
for apparent clearance and volume of distribution were similar 4 h than those based on NONMEM (Fig. 3). For the three
for all three programs. For the absorption parameters, the parametric simulations, S-ADAPT yielded the best represen-
differences were more apparent. The low Km/dose value tation of the observations during the terminal phase, followed
(mean, 0.433%) from NONMEM indicated that the release by NONMEM. The predicted variability was slightly too wide
from the stomach was estimated to be essentially a zero-order during the terminal phase for the parametric simulation (Fig.
process and that Vmax was inhibited in some subjects and stim- 3) using the variance-covariance matrix derived from the sup-
ulated in others, as indicated by the negative or positive indi- port point matrix in NPAG.
vidual Emax value. As the VPCs showed that the nonparametric simulation
In S-ADAPT, the release from the stomach to the intestine based on NPAG and the parametric simulation based on S-
had partial first-order and zero-order properties, as indicated ADAPT had the best predictive performance, breakpoints for
by the estimate of 42.6% for Km/dose. This rate of release was MCS are reported for these two models. The breakpoints for
3466
TABLE 2. Population PK parameters for 250 mg oral cefuroxime given as cefuroxime axetil after a breakfast with a significant amount of fat
Value for:

NONMEM S-ADAPT NPAG


Parameter Symbol
Population Median (1090 percentile) Population Median (1090 percentile) Population Median (1090 percentile)
mean (% SEi) of individual estimates mean (% SEi) of individual estimates mean (% SEi) of individual estimates

Fixed effects
Apparent clearance (liters/h) CL/F 23.0 (8.1) 21.7 (16.827.9) 21.7 (4.1) 21.5 (17.127.8) 21.8c 21.8 (17.028.3)
BULITTA ET AL.

Apparent vol of distribution V/F 40.7 (6.3) 39.7 (28.847.6) 38.7 (4.1) 40.2 (30.445.8) 34.8 35.6 (25.548.0)
(liters)
Maximum rate of release from Vmax 0 /dose 0.381j (12)a 0.308 (0.2000.471) 0.505 j (22)a 0.418 (0.1641.48) 1.53a, j 1.35 (0.5002.66)
stomach to intestine at time
zero divided by dose (1/h)
Fraction of dose associated with Km/dose 0.433 (70)a 0.488 (0.1803.09) 42.6 (39)a 38.1 (4.70478) 343a 363 (193498)
50% of Vmax 0 (%)
Fastest half-life of gastric Ln(2)/Vmax 0 Km 0.534 (0.2706.36) 30.4 (13.0120) 104 (49.3155)
release, if fraction of dose in
stomach is Km/dose (min)
Absorption half-life from Tabsf 9.00 (29) 14.2 (3.4830.9) 9.34 (15) 9.20 (4.7016.2) 16.9 17.5 (4.0032.2)
intestine to central
compartment (min)
Time past meal at which Vmax TC50 1.61 (14) 2.06 (1.093.26) 1.61 (20) 1.53 (0.6195.05) 2.08 2.33 (1.145.31)
changed by 50% (h)
c
Maximum fractional change on Lg_Emax 3.59 (8.9) 3.14 (4.15 to 1.79) 0.762 (31) 0.937 (1.96 to 0.641) 0.515 0.50 (3.44 to 4.95)
transformed scale
Maximum fractional change Emax 0.582 (0.845 to 0.426) 1.82 (0.2425.55) 2.78 (0.582 to 8.93)
Hill coefficient 10g 10g 10g

Random effectsb
BSV(CL/F) 0.202b (18)h 0.198b (31)h 0.193e
BSV(V/F) 0.201 (16) 0.183 (32) 0.258
BSV(Vmax 0 /dose) 0.401 (22) 0.900 (30) 0.518
BSV(Km /dose) 1.23 (37) 1.80 (27) 0.403
BSV(Tabs) 0.867 (23) 0.600 (41) 0.634
BSV(TC50) 0.536 (25) 0.946 (31) 0.580
BSV(Lg_Emax) 1.09d (30) 0.990d (29) 3.03d
Proportional residual error 0.0754 (15) 0.0851 (5.5) 0.0620
Additive residual error (mg/liter) 0.0065 (24) 0.0029 (48) 0.0062
a
Vmax 0 and Km were estimated and are reported as fractions of the cefuroxime dose. This assumes that the rate of drug release from the stomach is primarily determined by the meal and not by the amount of cefuroxime
axetil. This assumption yields a linear increase in peak concentrations and no change in time to peak with dose, which is consistent with literature data.
b
These estimates for random effects represent the apparent coefficient of variation for the BSV. All variability estimates are reported as square roots of the estimated variance, since this square root is an approximation
of the coefficient of variation of a normal distribution on natural logarithmic scale.
c
For NPAG, the medians of the support point matrix are provided. For Lg_Emax, the arithmetic mean is reported, since the distribution of Lg_Emax was more symmetric and since this choice for Lg_Emax yielded a
better predictive performance in the VPC.
d
The variability estimate for Lg_Emax is reported as the SD on the transformed scale.
e
Estimates are coefficients of variation calculated based on the means SD of results from the support point matrix.
f
The corresponding rate constant kabs (in liters/h) is calculated as ln(2)/(Tabs/60).
g
The Hill coefficient was initially estimated, and estimates ranged between 10 and 15. To improve model stability, the Hill coefficient was subsequently fixed at 10. This had no notable effect on the curve fits and
the objective function.
h
The value in parentheses represents the uncertainty of the respective BSV estimate and is the relative standard error of the estimated variance (reported as %).
i
Standard errors are derived from a nonparametric bootstrap for NONMEM and are asymptotic standard errors for S-ADAPT.
j
This estimate represents the maximum fraction of stomach content that can be released into the intestine per hour. This maximum fraction can change over time as defined in the model.
ANTIMICROB. AGENTS CHEMOTHER.

Downloaded from http://aac.asm.org/ on February 5, 2017 by GLAXOSMITHKLINE


VOL. 53, 2009 POPULATION PK AND PD OF CEFUROXIME AXETIL 3467

TABLE 3. Variance-covariance matrix for the final population 0.375 mg/liter for 250 mg q8h, 0.188 mg/liter for 500 mg q12h,
PK model in S-ADAPTa and 0.75 mg/liter for 500 mg q8h). Additional simulations with
Parameter CL/F V/F Vmax 0/dose Km/dose Tabs TC50 Lg_Emax a hypothetical higher rate of absorption showed that the
PK-PD MIC breakpoints were lower if the rate of absorption
CL/F 0.0393
V/F 0.0326 0.0333 was faster. The decrease in breakpoints was most pronounced
Vmax 0/dose 0.1143 0.0877 0.8100 for the 65% fTMIC target and q12h dosing.
Km/dose 0.2693 0.2066 1.5925 3.2560
Tabs 0.0083 0.0142 0.1036 0.1640 0.3604 High PTA expectation values (97.8% for the 40% fTMIC
TC50 0.0931 0.0389 0.5324 1.0200 0.0459 0.8945 target) were achieved by all three dosage regimens shown in
Lg_Emax 0.1530 0.1131 0.5702 1.1824 0.0414 0.8197 0.9793
Table 4 for S. pyogenes, penicillin-susceptible S. pneumoniae,
a
V/F, apparent volume of distribution in the central compartment. See Table N. gonorrhoeae, and N. meningitidis (results not shown for the
2 for explanations of other parameters.
latter two pathogens). High (90%) PTA expectation values

Downloaded from http://aac.asm.org/ on February 5, 2017 by GLAXOSMITHKLINE


were achieved for some but not all MIC distributions for S.
pneumoniae. The PTA expectation values were notably lower
the other two simulation models were within one 1.5-fold di-
for penicillin-intermediate S. pneumoniae, H. influenzae, M.
lution. The PTA-versus-MIC profiles were similar for these
catarrhalis, S. aureus, and K. pneumoniae (results not shown for
two models (Fig. 4). For the bacteriostasis target fTMIC of
the latter two pathogens).
40%, the PK-PD MIC breakpoints in NPAG and S-ADAPT
were 0.5 and 0.375 mg/liter, respectively, for 250 mg q12h and
0.5 and 0.5 mg/liter, respectively, for 250 mg q8h. As these PK DISCUSSION
models are linear with dose, 500 mg q12h achieved breakpoints
of 1 and 0.75 mg/liter and 500 mg q8h achieved breakpoints of Cefuroxime axetil has been used widely for treatment of
1 and 1 mg/liter in NPAG and S-ADAPT, respectively, for the community-acquired upper and lower respiratory tract infec-
40% fTMIC target. For the near-maximal-killing target tions (including community-acquired pneumonia) (45) that are
fTMIC of 65%, the PK-PD MIC breakpoints for S-ADAPT often caused by S. pneumoniae, H. influenzae, M. catarrhalis,
and NPAG were identical (0.094 mg/liter for 250 mg q12h, and S. pyogenes. The reported MIC90s of cefuroxime against H.

FIG. 3. VPC for a single oral dose of 250 mg cefuroxime as cefuroxime axetil after a breakfast with a significant amount of fat for the final model
in each program. The continuous lines represent the 10th, 50th, and 90th percentiles for the simulated concentrations, the broken lines represent
the same percentiles for the observations, and the markers show the observations. Ideally, the continuous and corresponding broken lines should
fall onto each other. The insets show the terminal phase on a semilogarithmic scale.
3468 BULITTA ET AL. ANTIMICROB. AGENTS CHEMOTHER.

one dose level (results not shown). However, such a mixed-


order absorption model would predict a notable increase in
Tmax with cefuroxime doses. This is in disagreement with the
data reported by Finn et al. (20). A mixed-order absorption
model cannot describe profiles with a dual peak.
Food increases the extent of bioavailability of cefuroxime
axetil from 36% in fasting subjects to 52% after a meal (20).
Similar results were found by Williams and Harding (54). In
both studies (20, 54), Tmax is prolonged by approximately 0.6 to
0.7 h for administration with food. Potential reasons for the
increased bioavailability and slightly longer Tmax observed un-

Downloaded from http://aac.asm.org/ on February 5, 2017 by GLAXOSMITHKLINE


der fed conditions include a more complete dissolution of
cefuroxime axetil due to a longer residence time in the stomach
and due to bile acid secretion stimulated by the presence of
lipids in the intestine (36, 50).
The proposed absorption model (Fig. 1) is in agreement with
the observations of literature studies for various dose levels of
cefuroxime axetil (20, 54). One limitation of our study is that
we had data only for 250 mg oral cefuroxime. Therefore, our
simulation results for 500 mg oral cefuroxime q12h should be
interpreted conservatively. The saturable rate of absorption is
described by a mixed-order release of drug from the stomach
to the intestine that is primarily saturated due to the presence
of food and not due to the cefuroxime axetil dose. In our
FIG. 4. PTA-versus-MIC profiles for the PK-PD target fTMICs of
40% (top) and 65% (bottom) for the nonparametric MCS based model, Vmax and Km are expressed as fractions of dose, and this
on NPAG (continuous lines) and the parametric MCS based on S- causes Tmax to be independent of dose. The second peak in
ADAPT (broken lines). some profiles was described by an increase in the rate of gastric
release over time.
This semiphysiological model proved to be robust (Table 2)
influenzae and M. catarrhalis are often 2 or 4 mg/liter (41, 45). and to yield excellent individual curve fits (Fig. 2) for all three
Whereas most isolates would be deemed susceptible by the population PK algorithms and programs. This absorption
CLSI breakpoint of 4 mg/liter for those pathogens, a signif- model was able to capture relevant features of complex oral
icant number of isolates would be considered resistant accord- absorption profiles (24, 32, 53) which showed that the rate of
ing to the BSAC and DIN susceptibility breakpoint of 1 gastric emptying is important for the absorption of amoxicillin
mg/liter. To evaluate which breakpoint is in better agreement (amoxicilline) and clavulanic acid (53).
with the predicted PK-PD MIC breakpoint, we applied para- Estimation of a full variance-covariance matrix and its use
metric and nonparametric population PK modeling and MCS during simulations yielded the best predictive performance,
for cefuroxime axetil. caused no instability during estimation, and did not prolong
The fTMIC best predicts the clinical and microbiological suc- run times in S-ADAPT and NPAG. This saves modeling time,
cess for cephalosporins. As the prolonged absorption phase of since there are fewer decisions about the choice of the param-
cefuroxime axetil has a notable influence on the fTMIC values, it eter variability model in S-ADAPT and NPAG than in
was critical to develop a population PK model that adequately NONMEM. NPAG does not directly estimate the variance-
captures the rates of absorption and BSVs that were observed in covariance matrix but always derives this full matrix from the
our studies and in the literature. We intensively qualified the estimated support points. Estimating a full variance-covariance
predictive performance of our population PK model to ensure matrix in NONMEM tended to cause model instability (i.e.,
that the model-predicted PK-PD MIC breakpoints are sound. An unsuccessful termination messages and the inability of
8-fold increase (from 125 to 1,000 mg) in the oral cefuroxime NONMEM to obtain asymptotic standard errors) and notably
axetil dose after a meal causes a 7.5-fold increase in the area increased estimation times in NONMEM.
under the curve and a 6.5-fold increase in peak concentrations The most important difference between the parametric and
(20) and causes no systematically altered time of peak concentra- nonparametric approaches is that the former describes BSV by
tion (Tmax) (20). Data from rats suggest a saturable component a parametric, multivariate distribution (often a multivariate
for the rate of absorption (4244). log-normal distribution). In contrast, nonparametric methods
We found a range of complex absorption patterns in healthy use a discrete set of support points to exactly store the BSV
volunteers (Fig. 2). Models with first-order or zero-order ab- and correlation structure of all estimated PK parameters in the
sorption with or without a lag time can describe the dose studied patient population. In the simplest case, each support
proportionality in area under the curve and peak concentration point essentially represents a complete set of PK parameters
(20) but cannot describe a mixed-order rate of absorption and for one patient and has a probability of 1 divided by the
the complex absorption profiles observed in our study. A number of subjects.
mixed-order absorption model with a lag time could describe As the variability of individual PK parameter estimates in
the plasma concentration time profiles of cefuroxime axetil at the studied subject population is exactly represented by the
VOL. 53, 2009 POPULATION PK AND PD OF CEFUROXIME AXETIL 3469

TABLE 4. PTA expectation values for various cefuroxime axetil dosage regimens and PK-PD targets for the parametric
MCS based on S-ADAPT
PTA expectation value (%) for indicated PK-PD target dosage regimen

fTMIC 40% fTMIC 65%


Pathogen, source, and yr (no. of isolates)a Source or reference
250 mg 250 mg 500 mg 250 mg 250 mg 500 mg
q12h q8h q8h q12h q8h q8h

S. pyogenes
United States and Canada, 1997-1999 (119) 25 99.2 99.2 99.2 98.9 99.2 99.2
Germany, 2002 (340) 7 99.6 99.8 100 99.3 99.6 99.9
Europe, 1997-1999 (662) 26 98.9 99.3 99.6 96.7 98.9 99.4

Downloaded from http://aac.asm.org/ on February 5, 2017 by GLAXOSMITHKLINE


S. pneumoniae
United States and Canada, 1997-1999 (417) 25 65.9 67.6 70.2 56.4 65.6 68.1
United Kingdom, 2002-2003 (519) 38 94.4 95.0 96.2 90.6 94.2 95.2
Germany, 2002 (331) 7 97.3 98.0 98.2 93.4 97.0 98.0
Europe, 1997-1999 (2,018) 26 71.5 74.1 77.8 63.2 70.8 74.7

PSSP
United States and Canada, 1997-1999 (249) 25 97.8 98.5 99.3 91.4 97.6 98.6
Global, 1997-2000 (2,102) 6 98.2 99.0 99.6 92.7 98.0 99.2
Europe, 1997-1999 (1,274) 26 98.4 99.1 99.6 93.5 98.2 99.2

PISP
United States and Canada, 1997-1999 (70) 25 44.5 52.5 65.1 10.9 43.4 55.0
Global, 1997-2000 (1,024) 6 46.0 53.9 64.4 19.9 44.0 55.7
Europe, 1997-1999 (458) 26 39.9 48.8 60.8 18.0 37.8 50.9

H. influenzae
United States and Canada, 1997-1999 (300) 25 18.5 45.3 83.8 1.7 16.0 53.4
Canada, 2001-2002 (1,350) 55 19.3 44.4 81.6 4.8 17.3 52.2
United Kingdom, 2002-2003 (581) 38 41.1 67.6 88.8 3.9 33.7 72.4
Germany, 2002 (300) 7 72.4 91.6 98.1 13.2 64.7 93.6
EUCAST (66,947) EUCAST MIC 41.5 72.7 99.1 4.9 34.9 79.3
distribution websiteb

M. catarrhalis
United States and Canada, 1997-1999 (231) 25 26.2 49.5 82.5 3.6 22.7 56.0
United Kingdom, 2002-2003 (269) 38 35.1 63.0 93.0 3.6 30.0 69.7
Germany, 2002 (308) 7 11.8 35.5 77.4 1.5 10.5 43.6
a
PSSP, penicillin-susceptible S. pneumoniae; PISP, penicillin-intermediate S. pneumoniae.
b
Available at http://www.eucast.org/mic_distributions_of_wild_type_microorganisms/ (last accessed on 20 December 2008).

set of support points, it was expected that the nonparametric We did not manually increase the BSV in clearance and
VPC had the best predictive performance (Fig. 3). The para- volume of distribution to mirror the higher variability in criti-
metric VPC using estimates from S-ADAPT had the best pre- cally ill patients, as the relatively low PK-PD MIC breakpoints
dictive performance among the parametric VPCs. We reported for oral cefuroxime do not support treatment of critically ill
the results from a parametric MCS in S-ADAPT with 10,000 patients. A higher variability in PK parameters and lower av-
virtual patients and from a nonparametric MCS in NPAG. As erage extent of bioavailability after intake of cefuroxime in the
every support point had the same probability for this study with fasting state (20, 54) are expected to result in lower PK-PD
frequent sampling, the latter MCS was identical to simulation MIC breakpoints than those reported here. As the sample size
from the individual PK parameter estimates of the 24 subjects. of 24 subjects probably did not allow us to obtain precise
The PTA-versus-MIC profiles from S-ADAPT and NPAG estimates of the BSV in the whole patient population, the
were similar (Fig. 4). For 250 mg oral cefuroxime q12h or q8h, results of our MCS should be interpreted conservatively. The
the PK-PD MIC breakpoints fell between 0.375 and 0.5 mg/ probability of clinical success of the simulated cefuroxime ax-
liter for the bacteriostasis target fTMIC of 40% but were etil dosage regimens will ultimately depend on the MIC distri-
approximately fourfold lower (0.094 mg/liter) for dosing of 250 bution of the pathogen(s) of interest in the local hospital.
mg q12h and the near-maximal-killing target fTMIC of 65%. Dosing at 250 mg cefuroxime q8h instead of q12h had only a
Dosing of 250 mg or 500 mg q8h increased the latter break- small benefit for S. pyogenes and penicillin-susceptible S. pneu-
points to 0.375 mg/liter or 0.75 mg/liter, respectively. Koeth et moniae, since 250 mg q12h achieved high PTA expectation
al. (31) determined a PK-PD MIC breakpoint of 1 mg/liter values, especially for the bacteriostasis target (Table 4).
for susceptibility for standard cefuroxime dosage regimens Administering cefuroxime axetil q8h yielded notably higher
based on an fTMIC of 40 to 50%. This higher breakpoint is PTA expectation values for some but not all MIC distributions
expected, since Koeth et al. (31) used average PK parameters of H. influenzae and M. catarrhalis. Although 500 mg oral
for simulation and did not include BSV. cefuroxime q8h is above the typically recommended oral cefu-
3470 BULITTA ET AL. ANTIMICROB. AGENTS CHEMOTHER.

roxime dose, parenteral doses of up to 6,000 mg split into four liter provided by the BSAC and DIN, whereas the CLSI break-
daily doses are recommended for severe infections. point of 4 mg/liter for most pathogens is higher than the
To put the results of our MCS into a clinical perspective, we PK-PD MIC breakpoints predicted by MCS in this study. Oral
compared our PTA expectation values to the microbiological cefuroxime (250 mg q12h or q8h) achieved high PTA expec-
and clinical outcomes in clinical studies. Clinical data for chil- tation values for S. pyogenes (96.7%) and penicillin-suscep-
dren with pneumococcal acute otitis media suggest a break- tible S. pneumoniae (91.4%) but notably lower PTA expec-
point of about 0.5 mg/liter for oral cefuroxime (15). Shah et al. tation values for M. catarrhalis, penicillin-intermediate S.
(47) studied hospitalized patients and outpatients in 14 coun- pneumoniae, and H. influenzae for most studied MIC distribu-
tries in Europe, Africa, and South America with acute exacer- tions for various countries. Administering 250 mg oral cefu-
bation of chronic bronchitis (AECB) and found a 60% bacte- roxime q8h instead of q12h was most beneficial for the near-
riological overall cure rate for 250 mg oral cefuroxime given maximal-killing target for MICs between 0.094 and 0.375 mg/

Downloaded from http://aac.asm.org/ on February 5, 2017 by GLAXOSMITHKLINE


twice daily (BID). This cure rate is comparable to the placebo liter. Future clinical studies that assess the MIC of the
response rate for AECB (30, 46), depending on the severity of causative pathogen are warranted to validate these predictions
disease. Interestingly, 56% (22 of 39) of the H. influenzae for the clinical and microbiological success for q12h and q8h
isolates were eradicated. Using per-protocol and intention-to- cefuroxime axetil dosage regimens.
treat analyses, the authors reported clinical cure rates of 66%
and 61%, respectively, at the clinical endpoint (5 to 14 days ACKNOWLEDGMENT
posttreatment) and of 53% and 39%, respectively, at the fol- We thank George Drusano for fruitful discussions about this project.
low-up 3 to 4 weeks posttreatment. Although the authors did
not report the MICs in those patients, the failures for the REFERENCES

treatment of H. influenzae show a suboptimal effectiveness of 1. Alvarez-Sala, J. L., P. Kardos, J. Martinez-Beltran, P. Coronel, and L.
Aguilar. 2006. Clinical and bacteriological efficacy in treatment of acute
oral cefuroxime against this pathogen. exacerbations of chronic bronchitis with cefditoren-pivoxil versus cefu-
Chodosh et al. (10) found a significantly lower microbiolog- roxime-axetil. Antimicrob. Agents Chemother. 50:17621767.
2. Ambrose, P. G., S. M. Bhavnani, R. N. Jones, W. A. Craig, and M. N. Dudley.
ical eradication rate for 500 mg oral cefuroxime BID (82%) 2004. Use of pharmacokinetic-pharmacodynamic and Monte-Carlo simula-
versus 500 mg oral ciprofloxacin BID (96%) in an outpatient tion as decision support for the re-evaluation of NCCLS cephem suscepti-
trial with AECB patients. Cefuroxime eradicated S. pneu- bility breakpoints for Enterobacteriaceae. Abstr. 44th Intersci. Conf. Antimi-
crob. Agents Chemother., abstr. A-138.
moniae in 100% of the cases (13/13) but had eradication rates 3. Ambrose, P. G., S. M. Bhavnani, C. M. Rubino, A. Louie, T. Gumbo, A.
of only 76% (19/25) for M. catarrhalis and 86% (19/22) for H. Forrest, and G. L. Drusano. 2007. Pharmacokinetics-pharmacodynamics of
influenzae. In an outpatient trial with AECB patients, de Abate antimicrobial therapy: its not just for mice anymore. Clin. Infect. Dis. 44:
7986.
et al. (16) found a clinical cure rate of 77% for 250 mg oral 4. Bauer, R. J. 2007. S-ADAPT/MCPEM users guide (version 1.55). Software
cefuroxime BID, which was significantly lower than the clinical for pharmacokinetic, pharmacodynamic and population data analysis. Bio-
medical Simulations Resource, Berkeley, CA.
cure rate of 89% for 400 mg gatifloxacin once daily. The mi- 5. Beal, S. L., L. B. Sheiner, and A. J. Boeckmann. 2006. NONMEM users
crobiological eradication rates were 77% for cefuroxime and guides (1989 to 2006). Icon Development Solutions, Ellicott City, MD.
90% for gatifloxacin. 6. Bouchillon, S. K., D. J. Hoban, J. L. Johnson, B. M. Johnson, D. L. Butler,
K. A. Saunders, L. A. Miller, and J. A. Poupard. 2004. In vitro activity of
A trial using patients with community-acquired pneumonia gemifloxacin and contemporary oral antimicrobial agents against 27,247
(19) showed a significantly lower rate of microbiological erad- Gram-positive and Gram-negative aerobic isolates: a global surveillance
ication of H. influenzae for combinations of intravenous ceftri- study. Int. J. Antimicrob. Agents 23:181196.
7. Brauers, J., S. Bagel, and M. Kresken. 2005. Aktuelle Resistenzsituation bei
axone (1 to 2 g once daily or BID) and/or oral cefuroxime bakteriellen Erregern von ambulant erworbenen Atemwegsinfektionen.
axetil (500 mg BID) than for intravenous and/or oral levofloxa- Chemother. J. 14:4553.
8. British Society for Antimicrobial Chemotherapy. 2008. BSAC methods for
cin (500 mg once daily). The former regimen had an eradica- antimicrobial susceptibility testing, version 7.1. British Society for Antimi-
tion rate of 79% and the latter 100%. Upchurch et al. (49) crobial Chemotherapy, Birmingham, United Kingdom.
found a clinical cure rate of 74.5% for treatment of acute 8a.Bulitta, J., C. Landersdorfer, M. Kinzig-Schippers, V. Jakob, M. Rodamer,
G. L. Drusano, U. Thyroff-Friesinger, U. Holzgrabe, and F. Sorgel. 2006.
bacterial sinusitis with 250 mg oral cefuroxime for 10 days but Abstr. 46th Intersci. Conf. Antimicrob. Agents Chemother., poster A-1119.
did not document the bacterial etiology. Alvarez-Sala et al. (1) 9. Bulitta, J. B., S. B. Duffull, M. Kinzig-Schippers, U. Holzgrabe, U. Stephan,
found a clinical success rate of approximately 82% for patients G. L. Drusano, and F. Sorgel. 2007. Systematic comparison of the population
pharmacokinetics and pharmacodynamics of piperacillin in cystic fibrosis
with H. influenzae and S. pneumoniae for 250 mg oral cefu- patients and healthy volunteers. Antimicrob. Agents Chemother. 51:2497
roxime q12h. 2507.
10. Chodosh, S., J. McCarty, S. Farkas, M. Drehobl, R. Tosiello, M. Shan, L.
In conclusion, we developed a semiphysiological population Aneiro, S. Kowalsky, et al. 1998. Randomized, double-blind study of cipro-
PK model for oral cefuroxime which provided precise and floxacin and cefuroxime axetil for treatment of acute bacterial exacerbations
unbiased individual curve fits for complex absorption profiles of chronic bronchitis. Clin. Infect. Dis. 27:722729.
11. Clinical and Laboratory Standards Institute. 2007. Performance standards
and had an excellent predictive performance. The nonpara- for antimicrobial susceptibility testing; 17th informational supplement, vol.
metric VPC based on NPAG showed a better predictive per- 27. Clinical and Laboratory Standards Institute, Wayne, PA.
formance than the best parametric VPC in S-ADAPT. The 12. Craig, W. A. 2003. Basic pharmacodynamics of antibacterials with clinical
applications to the use of beta-lactams, glycopeptides, and linezolid. Infect.
PK-PD MIC breakpoints were 0.375 to 0.5 mg/liter for 250 mg Dis. Clin. N. Am. 17:479501.
oral cefuroxime q12h and 0.5 mg/liter for 250 mg oral cefu- 13. Craig, W. A. 1998. Choosing an antibiotic on the basis of pharmacodynamics.
Ear Nose Throat J. 77:712.
roxime q8h for the bacteriostasis target fTMIC of 40%. 14. Craig, W. A. 1998. Pharmacokinetic/pharmacodynamic parameters: rationale
Dosing at 250 mg cefuroxime q8h instead of q12h increased the for antibacterial dosing of mice and men. Clin. Infect. Dis. 26:112.
breakpoint for the near-maximal-killing target fTMIC of 65% 15. Dagan, R., O. Abramson, E. Leibovitz, D. Greenberg, R. Lang, S. Goshen, P.
Yagupsky, A. Leiberman, and D. M. Fliss. 1997. Bacteriologic response to
from 0.094 mg/liter to 0.375 mg/liter. These breakpoints were oral cephalosporins: are established susceptibility breakpoints appropriate in
(slightly) lower than the susceptibility breakpoint of 1 mg/ the case of acute otitis media? J. Infect. Dis. 176:12531259.
VOL. 53, 2009 POPULATION PK AND PD OF CEFUROXIME AXETIL 3471

16. de Abate, C. A., R. A. McIvor, P. McElvaine, et al. 1999. Smokers treated Investigations into the mechanism for the improved oral systemic bioavail-
with gatifloxacin had a high clinical cure rate: gatifloxacin vs. cefuroxime ability of cefuroxime from cefuroxime axetil when taken after food (proceed-
axetil in patients with acute exacerbations of chronic bronchitis. J. Respir. ings of the BPS, Sept. 1991). Br. J. Clin. Pharmacol. 33:226P227P.
Dis. 20(Suppl.):S23S29. 37. Mason, E. O., Jr., L. B. Lamberth, N. L. Kershaw, B. L. Prosser, A. Zoe, and
17. Deutsches Institut fur Normung e.V. 2004. Medizinische Mikrobiologie P. G. Ambrose. 2000. Streptococcus pneumoniae in the USA: in vitro sus-
Empfindlichkeitsprufung von mikrobiellen Krankheitserregern gegen Che- ceptibility and pharmacodynamic analysis. J. Antimicrob. Chemother. 45:
motherapeutikaTeil 4: Bewertungsstufen fur die minimale Hemmkonzen- 623631.
trationMHK-Grenzwerte von antibakteriellen Wirkstoffen. DIN 58940-4. 38. Morrissey, I., M. Robbins, L. Viljoen, and D. F. Brown. 2005. Antimicrobial
Beuth Verlag, Berlin, Germany. susceptibility of community-acquired respiratory tract pathogens in the UK
18. Drusano, G. L. 2004. Antimicrobial pharmacodynamics: critical interactions during 2002/3 determined locally and centrally by BSAC methods. J. Anti-
of bug and drug. Nat. Rev. Microbiol. 2:289300. microb. Chemother. 55:200208.
19. File, T. M., Jr., J. Segreti, L. Dunbar, R. Player, R. Kohler, R. R. Williams, 39. Mouton, J. W., M. N. Dudley, O. Cars, H. Derendorf, and G. L. Drusano.
C. Kojak, and A. Rubin. 1997. A multicenter, randomized study comparing 2005. Standardization of pharmacokinetic/pharmacodynamic (PK/PD) ter-
the efficacy and safety of intravenous and/or oral levofloxacin versus ceftri- minology for anti-infective drugs: an update. J. Antimicrob. Chemother.
axone and/or cefuroxime axetil in treatment of adults with community- 55:601607.

Downloaded from http://aac.asm.org/ on February 5, 2017 by GLAXOSMITHKLINE


acquired pneumonia. Antimicrob. Agents Chemother. 41:19651972. 40. Peric, M., F. A. Browne, M. R. Jacobs, and P. C. Appelbaum. 2003. Activity
20. Finn, A., A. Straughn, M. Meyer, and J. Chubb. 1987. Effect of dose and food of nine oral agents against gram-positive and gram-negative bacteria encoun-
on the bioavailability of cefuroxime axetil. Biopharm. Drug Dispos. 8:519 tered in community-acquired infections: use of pharmacokinetic/pharmaco-
526. dynamic breakpoints in the comparative assessment of beta-lactam and mac-
21. Foord, R. D. 1976. Cefuroxime: human pharmacokinetics. Antimicrob. rolide antimicrobial agents. Clin. Ther. 25:169177.
Agents Chemother. 9:741747. 41. Perry, C. M., and R. N. Brogden. 1996. Cefuroxime axetil. A review of its
22. Garraffo, R., H. B. Drugeon, and D. Chiche. 1997. Pharmacokinetics and antibacterial activity, pharmacokinetic properties and therapeutic efficacy.
pharmacodynamics of two oral forms of cefuroxime axetil. Fundam. Clin. Drugs 52:125158.
Pharmacol. 11:9095. 42. Ruiz-Balaguer, N., A. Nacher, V. G. Casabo, and M. Merino. 1997. Nonlin-
23. Hara, T., H. Saeki, A. Yamamoto, K. Ohta, T. Suzuki, M. Ara, M. Sugawara, ear intestinal absorption kinetics of cefuroxime axetil in rats. Antimicrob.
Y. Sano, H. Sakuma, and K. Satoh. 1988. Concentrations of cefuroxime in Agents Chemother. 41:445448.
the skin. Jpn. J. Antibiot. 41:1017. 43. Ruiz-Balaguer, N., A. Nacher, V. G. Casabo, and M. Merino Sanjuan. 2002.
24. Higaki, K., S. Y. Choe, R. Lobenberg, L. S. Welage, and G. L. Amidon. 2008. Intestinal transport of cefuroxime axetil in rats: absorption and hydrolysis
Mechanistic understanding of time-dependent oral absorption based on gas- processes. Int. J. Pharm. 234:101111.
tric motor activity in humans. Eur. J. Pharm. Biopharm. 70:313325. 44. Ruiz-Carretero, P., M. Merino-Sanjuan, A. Nacher, and V. G. Casabo. 2004.
25. Hoban, D. J., S. K. Bouchillon, J. L. Johnson, G. G. Zhanel, D. L. Butler, Pharmacokinetic models for the saturable absorption of cefuroxime axetil
K. A. Saunders, L. A. Miller, and J. A. Poupard. 2003. Comparative in vitro and saturable elimination of cefuroxime. Eur. J. Pharm. Sci. 21:217223.
potency of amoxycillin-clavulanic acid and four oral agents against recent 45. Scott, L. J., D. Ormrod, and K. L. Goa. 2001. Cefuroxime axetil: an updated
North American clinical isolates from a global surveillance study. Int. J. review of its use in the management of bacterial infections. Drugs 61:1455
Antimicrob. Agents 21:425433. 1500.
26. Hoban, D. J., S. K. Bouchillon, J. L. Johnson, G. G. Zhanel, D. L. Butler, 46. Sethi, S., and T. F. Murphy. 2004. Acute exacerbations of chronic bronchitis:
K. A. Saunders, L. A. Miller, and J. A. Poupard. 2003. Comparative in vitro new developments concerning microbiology and pathophysiologyimpact
surveillance of amoxicillin-clavulanic acid and four oral comparators against on approaches to risk stratification and therapy. Infect. Dis. Clin. N. Am.
21232 clinical isolates from europe. Eur. J. Clin. Microbiol. Infect. Dis. 18:861882.
22:261267. 47. Shah, P. M., F. P. Maesen, A. Dolmann, N. Vetter, E. Fiss, and R. Wesch.
27. Ison, C. A., J. W. Mouton, K. Jones, K. A. Fenton, and D. M. Livermore. 1999. Levofloxacin versus cefuroxime axetil in the treatment of acute exac-
2004. Which cephalosporin for gonorrhoea? Sex. Transm. Infect. 80:386 erbation of chronic bronchitis: results of a randomized, double-blind study.
388. J. Antimicrob. Chemother. 43:529539.
28. Jacobs, M. R. 2003. How can we predict bacterial eradication? Int. J. Infect. 48. Stoeckel, K., W. L. Hayton, and D. J. Edwards. 1995. Clinical pharmacoki-
Dis. 7(Suppl. 1):S13S20. netics of oral cephalosporins. Antibiot. Chemother. 47:3471.
29. Jacobs, M. R., S. Bajaksouzian, A. Zilles, G. Lin, G. A. Pankuch, and P. C. 49. Upchurch, J., M. Rosemore, R. Tosiello, S. Kowalsky, and R. Echols. 2006.
Appelbaum. 1999. Susceptibilities of Streptococcus pneumoniae and Hae- Randomized double-blind study comparing 7- and 10-day regimens of faro-
mophilus influenzae to 10 oral antimicrobial agents based on pharmacody- penem medoxomil with a 10-day cefuroxime axetil regimen for treatment of
namic parameters: 1997 U.S. surveillance study. Antimicrob. Agents Che- acute bacterial sinusitis. Otolaryngol. Head Neck Surg. 135:511517.
mother. 43:19011908. 50. Vasu, S., C. Adithan, C. H. Shashindran, M. Asad, K. Koumaravelou, and I.
30. Jorgensen, A. F., J. Coolidge, P. A. Pedersen, K. P. Petersen, S. Waldorff, and Topno. 2000. Effect of two types of Indian breakfast on bioavailability of
E. Widding. 1992. Amoxicillin in treatment of acute uncomplicated exacer- cefuroxime axetil. Indian J. Med. Res. 112:104108.
bations of chronic bronchitis. A double-blind, placebo-controlled multicen- 51. Viberg, A., O. Cars, M. O. Karlsson, and S. Jonsson. 2008. Estimation of
tre study in general practice. Scand. J. Prim. Health Care 10:711. cefuroxime dosage using pharmacodynamic targets, MIC distributions, and
31. Koeth, L. M., D. Felmingham, M. R. Jacobs, and F. Rossi. 2004. Antimicro- minimization of a risk function. J. Clin. Pharmacol. 48:12701281.
bial resistance of Streptococcus pneumoniae and Haemophilus influenzae in 52. Viberg, A., A. Lannergard, A. Larsson, O. Cars, M. O. Karlsson, and M.
Sao Paulo, Brazil from 1996 to 2000. Int. J. Antimicrob. Agents 23:356361. Sandstrom. 2006. A population pharmacokinetic model for cefuroxime using
32. Kong, F., and R. P. Singh. 2008. A model stomach system to investigate cystatin C as a marker of renal function. Br. J. Clin. Pharmacol. 62:297303.
disintegration kinetics of solid foods during gastric digestion. J. Food Sci. 53. Weitschies, W., C. Friedrich, R. S. Wedemeyer, M. Schmidtmann, O. Kosch,
73:E202E210. M. Kinzig, L. Trahms, F. Sorgel, W. Siegmund, S. Horkovics-Kovats, F.
33. Leary, R., R. W. Jelliffe, A. Schumitzky, and M. Van Guilder. 2001. An Schwarz, J. Raneburger, and H. Monnikes. 2008. Bioavailability of amoxi-
adaptive grid non-parametric approach to pharmacokinetic and dynamic cillin and clavulanic acid from extended release tablets depends on intragas-
(PK/PD) models. IEEE Computer Society, Bethesda, MD. tric tablet deposition and gastric emptying. Eur. J. Pharm. Biopharm. 70:
34. Lesmana, M., C. I. Lebron, D. Taslim, P. Tjaniadi, D. Subekti, M. O. Wasfy, 641648.
J. R. Campbell, and B. A. Oyofo. 2001. In vitro antibiotic susceptibility of 54. Williams, P. E., and S. M. Harding. 1984. The absolute bioavailability of oral
Neisseria gonorrhoeae in Jakarta, Indonesia. Antimicrob. Agents Chemother. cefuroxime axetil in male and female volunteers after fasting and after food.
45:359362. J. Antimicrob. Chemother. 13:191196.
35. Llanes, R., J. Sosa, D. Guzman, A. Llop, E. A. Valdes, I. Martinez, S. Palma, 55. Zhanel, G. G., L. Palatnick, K. A. Nichol, D. E. Low, and D. J. Hoban. 2003.
and M. I. Lantero. 2003. Antimicrobial susceptibility of Neisseria gonor- Antimicrobial resistance in Haemophilus influenzae and Moraxella catarrhalis
rhoeae in Cuba (1995-1999): implications for treatment of gonorrhea. Sex. respiratory tract isolates: results of the Canadian Respiratory Organism
Transm. Dis. 30:1014. Susceptibility Study, 1997 to 2002. Antimicrob. Agents Chemother. 47:1875
36. Mackay, J., A. E. Mackie, J. L. Palmer, A. Moult, and N. S. Baber. 1992. 1881.

You might also like