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

European Journal of Clinical Pharmacology

https://doi.org/10.1007/s00228-020-03001-z

PHARMACOKINETICS AND DISPOSITION

Ceftriaxone dosing in patients admitted from the emergency


department with sepsis
Aaron J. Heffernan 1,2 & Rebecca A. Curran 3 & Kerina J. Denny 4 & Fekade B. Sime 2,5 & Claire L. Stanford 6 &
Brett McWhinney 7 & Jacobus Ungerer 7,8 & Jason A. Roberts 2,5,9,10,11 & Jeffrey Lipman 5,10,11

Received: 5 April 2020 / Accepted: 16 September 2020


# Springer-Verlag GmbH Germany, part of Springer Nature 2020

Abstract
Purpose Unbound ceftriaxone pharmacokinetics in adult patients have been poorly characterised. The objective of this study is to
determine the ceftriaxone dose that achieves an unbound trough concentration ≥ 0.5 mg/L in > 90% of adult patients receiving
once-daily dosing presenting to the emergency department (ED) with sepsis.
Methods We performed a prospective single-centre pharmacokinetic study. A single unbound plasma ceftriaxone concentration
was obtained from each patient using blood collected as part of routine clinical practice within the first dosing interval. Samples
were analysed using a validated ultra-high pressure liquid chromatography method. Population pharmacokinetic analysis and
Monte Carlo simulations (n = 1000) were performed using Pmetrics for R.
Results A ceftriaxone concentration obtained throughout the first dosing interval was available for fifty adult patients meeting
sepsis criteria. Using this concentration time-curve data, a pharmacokinetic model was developed with acceptable predictive
performance per the visual predictive check. Simulations show that a 1-g once-daily dose is unlikely to achieve the minimum
therapeutic ceftriaxone exposure in > 90% patients with a creatinine clearance ≥ 60 mL/min. However, a 2-g once-daily dose will
provide a therapeutic exposure for target pathogens infecting patients with a creatinine clearance ≤ 140 mL/min.
Conclusions Ceftriaxone administered as a 1-g once-daily dose is unlikely to achieve a therapeutic exposure in > 90% of patients
presenting to the ED with sepsis. Increasing the ceftriaxone dose to 2 g once daily will likely achieve the desired exposure against
target pathogens. Future clinical trials are required to determine any potential clinical benefit of optimised ceftriaxone dosing.

Keywords Ceftriaxone . Sepsis . Pharmacokinetics . Dose

Aaron J. Heffernan and Rebecca A. Curran are joint first authors

* Aaron J. Heffernan 6
Department of Emergency Medicine, Gold Coast University
aaron.heffernan@griffithuni.edu.au Hospital, Southport, Australia
7
Department of Chemical Pathology, Pathology Queensland,
1
School of Medicine, Griffith University, Southport, Australia Herston, Australia
8
2 Faculty of Biomedical Science, University of Queensland, St
Centre for Translational Anti-Infective Pharmacodynamics, School
Lucia, Australia
of Pharmacy, University of Queensland, Woolloongabba, Australia
9
3
Department of Pharmacy, Royal Brisbane and Women’s Hospital,
Department of Pharmacy, Gold Coast University Hospital, Brisbane, Australia
Southport, Australia
10
Department of Intensive Care Medicine, Royal Brisbane and
4
Department of Intensive Care, Gold Coast University Hospital, Women’s Hospital, Brisbane, Australia
Southport, Australia 11
Division of Anaesthesiology Critical Care Emergency and Pain
5
University of Queensland Centre for Clinical Research, Faculty of Medicine, Nîmes University Hospital, University of Montpellier,
Medicine, University of Queensland, Herston, Australia Nîmes, France
Eur J Clin Pharmacol

Introduction matched patients receiving 2 g once-daily showed no signifi-


cant difference in clinical cure rates [19]. In contrast, pharma-
Ceftriaxone is a clinically important intravenous broad- cokinetic studies in patients with sepsis admitted to the inten-
spectrum cephalosporin β-lactam antibiotic commonly pre- sive care unit show that a 1 g twice-daily dose likely achieves
scribed in the emergency department (ED) for the manage- the therapeutic target ceftriaxone exposure [20].
ment of severe pneumonia, urinary tract infections and intra- In the absence of robust clinical evidence directly compar-
abdominal infections [1]. It is clear that prescribing an empiric ing ceftriaxone dosing regimens with patient-centered out-
antibiotic to which the causative pathogen is sensitive to re- comes, an approach that aims to determine the likely optimal
duces the risk of mortality in patients with sepsis; therefore, dose based on pharmacokinetic principles may be beneficial.
ceftriaxone may be used empirically given its favourable ac- This research therefore aims to determine if a 1 g once-
tivity against pathogens such as Escherichia coli, daily ceftriaxone dose achieves recommended therapeutic tar-
Streptococcus pneumoniae, Klebsiella pneumoniae and get of an unbound trough concentration exceeding 0.5 mg/L
Haemophilus influenzae [2]. However, the clinician should for > 90% of adult patients with sepsis not admitted to the
also optimise the antibiotic dose. Administering a dose that intensive care unit.
achieves a cephalosporin concentration exceeding the patho-
gen minimum inhibitory concentration (MIC) throughout the
dosing interval (% fT>MIC) is associated with an approximate Methods
threefold increase in clinical cure rates and an approximate
twofold increase in bacterial eradication rates compared with Setting
patients not meeting this target exposure [3, 4]. These are
important considerations as antibiotics prescribed in the ED This single-centre prospective observational pharmacokinetic
are likely to be continued on admission to hospital; therefore, study was conducted within a tertiary hospital in South-East
the selection of an appropriate antibiotic given at the optimal Queensland. Waiver of informed consent was granted by the
dose in the ED is highly likely to contribute to positive patient Gold Coast University Hospital Ethics Committee HREC/18/
outcomes [5, 6]. QGC/100.
Prescribing the ceftriaxone dosing regimen that achieves
optimal exposure in patients with sepsis is challenging due Study population
to the underlying pathophysiological changes that occur.
These can include the following: fluid shifts from third spac- Adult patients (age ≥ 18 years) admitted to hospital via the ED
ing, augmented renal clearance (creatinine clearance ≥ 130 for ≥ 24 h receiving ceftriaxone 1 g intravenously once-daily
mL/min/1.73 m2), altered hepatic/biliary elimination and for the treatment of sepsis were eligible for inclusion in the
hypoalbuminaemia increasing the unbound ceftriaxone frac- study. Sepsis was defined as having a presumed or microbio-
tion of this highly protein-bound (> 90%) antibiotic [7, 8]. logical confirmed infection with a Systemic Inflammatory
Some of these physiological changes may be exacerbated by Response Score (SIRS) ≥ 2 of a temperature ≥ 38 or ≤ 36
fluid resuscitation, which may further dilute albumin and con- °C, a heart rate ≥ 90 beats per minute, respiratory rate ≥ 20
tribute to an increased volume that the antibiotic must distrib- per minute or a white cell count ≥ 12 × 109 cells/L; or a Quick
ute into [7]. Moreover, these physiological changes are likely Sequential Organ Failure Assessment (qSOFA) Score ≥ 2 of a
to be pronounced on presentation to ED, early in the course of systolic blood pressure ≤ 90 mmHg, Glasgow Coma Scale ≤
sepsis. Previous studies in critically ill patients admitted to the 14 or a respiratory rate ≥ 22 per minute [21]. The following
intensive care unit (ICU) highlight that 1-g or 2-g once-daily patients were excluded: pregnant, receiving either intermittent
dosing regimens are unlikely to achieve the optimal exposure or continuous dialysis, received a ceftriaxone dosing regimen
associated with improved clinical outcomes, a 100% fT>MIC other than 1 g once-daily, or required intensive care unit ad-
[9–12]. However, as patients with sepsis may not be confined mission. Patients admitted to ICU were excluded as most
to the intensive care unit, ceftriaxone dose optimisation for would receive a 1 g twice-daily dose, consistent with current
other patients who present to the ED should also be reviewed evidence in the ICU population, and the primary aim of this
to potentially improve patient outcomes [13, 14]. However, study was to enable a robust evaluation of the 1-g once-daily
many guidelines provide a wide dosing range, from 1 g once- dosing regimen that may be used on general wards. Clinical
daily to 2 g twice-daily [1, 15–17]. This may prompt clinicians and anthropometric data including the patient’s age (years),
to use a lower dose, which remains in accordance with man- weight (kg), height (cm), systolic blood pressure (mmHg),
ufacturer’s recommendations [18]. Importantly, there is con- heart rate (beats per minute), temperature (°C), respiratory rate
flicting evidence for a higher dosing regimen. One small study (per minute), Glasgow Coma Scale, suspected infection
of patients in Japan with community-acquired pneumonia re- source, microbiological results, liver transaminases (aspartate
ceiving a 1-g once-daily dose compared with propensity- aminotransferase, alanine aminotransferase, gamma-glutamyl
Eur J Clin Pharmacol

transferase, alkaline phosphatase [U/L]), albumin (g/L), lac- on physiological plausibility. Variables assessed against the
tate (mmol/L), unconjugated and conjugated bilirubin volume of distribution and clearance included age (years), sex,
(μmol/L) and creatinine (μmol/L) were extracted from the weight (kg), height (cm), creatinine clearance (determined
patient’s medical record where available. Suspected infection using the Cockcroft-Gault equation using total body weight)
source was per the treating clinician’s working diagnosis. A expressed as mL/min [26], lactate (mmol/L), albumin (g/L),
sample size of 50 patients has previously been shown to ade- conjugated and unconjugated bilirubin (μmol/L), and liver
quately describe the population mean area under the transaminases (aspartate aminotransferase, alanine amino-
concentration-time curve [22]. transferase, gamma-glutamyl transferase, alkaline phospha-
tase [U/L]). The incorporation of different patient variables
Sample handing, storage and measurement into the model that reduced the LLR, AIC, or BIC and/or
improving the goodness-of-fit plots was included.
Blood specimens taken in an ethylenediaminetetraacetic acid
(EDTA)–containing vial as part of routine clinical practice Probability of target attainment and fractional target
were used to determine the ceftriaxone concentration. Only attainment
one sample was taken per patient at variable times throughout
the first ceftriaxone dosing interval. Samples were centrifuged Monte Carlo simulations (n = 1000) were performed using
at 3000 rpm for 10 min and stored at − 80 °C until analysis. ceftriaxone doses of 1 or 2 g administered once- or twice-
Analysis was conducted using a previously validated and pub- daily in patients with a creatinine clearance ranging from 20
lished methodology [23] with the following modifications. to 140 mL/min. The probability of these regimens achieving a
Briefly, unbound ceftriaxone was separated from plasma target unbound ceftriaxone concentration (probability of tar-
using an Amicon Ultra Centrifugal 30-kDa filter. Unbound get attainment; PTA) exceeding the bacterial MIC throughout
ceftriaxone plasma concentrations were quantified using an the dosing interval (fT>MIC 100%) for MICs ranging from
Ultra Performance Liquid Chromatography (UPLC) coupled 0.032 to 1 mg/L was determined. Given that many microbio-
with QDa Mass Detection (Waters Corporation, Milford, MA, logical laboratories do not report a specific MIC, target expo-
USA). The lower limit of quantification was 0.1 mg/L and sures for this study were a free minimum concentration (fCmin)
imprecision was < 10% at all concentrations tested (0.1–50 ≥ 0.5 mg/L based on the current European Committee of
mg/L). No bias has been identified compared with target total Antimicrobial Susceptibility Testing (EUCAST) breakpoints
ceftriaxone concentrations. Our assay is approved as an [27].
in vitro diagnostic product with the appropriate certified ref- The fractional target attainment (FTA) represents the prob-
erence material used to calibrate our method. ability that patients would achieve the antibiotic therapeutic
target against a specific pathogen assuming a wild-type distri-
Population pharmacokinetic modelling bution of susceptible MICs. The FTA, assuming an unbound
ceftriaxone target of fT>MIC 100%, was calculated for the
Unbound ceftriaxone concentrations were analysed using above ceftriaxone dosing regimens for susceptible
Pmetrics version 1.5.0 software package for R, which allows S. pneumoniae, H. influenzae, E. coli, and K. pneumoniae
for non-parametric parameter estimation thereby making no isolates using the MIC distribution obtained from the
assumption of the underlying parameter estimate data distri- EUCAST database (available from www.eucast.org).
bution [24]. Parametric approaches may underestimate the Dosing regimens were considered successful if the PTA
population inter-individual parameter variance when com- was ≥ 90% of the simulated patient population.
pared with a non-parametric approach that has been shown
to appropriately describe extreme parameter estimates [25].
A one-compartment pharmacokinetic model was assessed. Results
Both the additive and multiplicative error models incorporat-
ing a linear residual error estimate within Pmetrics were tested Study population
and optimised using an iterative process based on model di-
agnostics. Diagnostic plots, including the observed vs. pre- Ceftriaxone concentrations were available for 50 patients
dicted concentrations, residuals vs. predicted concentrations (Table 1). These patients had a mean age of 69 years (standard
and vs. time; as well as statistical comparison tests including deviation 15.6 years), with mild-moderate renal impairment
the Akaike Information Criterion (AIC), Bayesian (eGFR 63.48 [standard deviation 23.6] mL/min/1.73 m2).
Information Criterion (BIC) and log-likelihood ratio (LLR) Patients are presented to the ED with predominantly respira-
were considered to compare different models. tory tract infections (86%). Two isolates had an MIC avail-
Patient variables were selected a priori for potential inclu- able. One S. pneumoniae and one H. influenzae had MICs of
sion in the model to account for inter-patient variability based 0.016 and 0.032 mg/L respectively.
Eur J Clin Pharmacol

Table 1 Patient demographic details value (63 mL/min) (Eq. 1). Incorporating TVNR with TVR
Median (IQR) or N (%) reduced the LLR, AIC, and BIC by 6, 3.6, and 2 respectively.
as appropriate The linear adjustment of TVR with creatinine normalised to
63 mL/min reduced the LLR, AIC, and BIC each by 25.5. The
Age, years 73 (58.25–80.75) final model was adequately fit as described by the visual pre-
Weight, kg 72.4 (59.26–81.38) dictive check (Fig. 1) and a population- and individual-
Lactate, mmol/L 1.7 (1.2–2.6) predicted model R2 of 0.47 and 0.99 respectively. Other pa-
Albumin, g/L 34 (31–37) tient covariates (age, weight, albumin, lactate, liver transami-
Creatinine, μmol/L 82.5 (63.25–114.75) nases, unconjugated bilirubin, and conjugated bilirubin) did
Creatinine clearance, mL/min 62.39 (39.15–82.64) not improve the model performance. Final model parameter
Conjugated bilirubin, μmol/L 4 (4–6) estimates are described in Table 2. The moderate-high coeffi-
Unconjugated bilirubin, μmol/L 14 (8–20) cient of variation (CV%) may suggest, at least in part, poor
Alkaline phosphatase, U/L 78 (68–104.5) model precision for the parameter estimates, and also impor-
Gamma-glutamyl transferase, U/L 28 (16–70.75) tantly, reflecting the pharmacokinetic variability of unbound
Aspartate aminotransferase, U/L 21 (16.25–38.75) ceftriaxone in this patient cohort. Furthermore, the TVNR
Alanine aminotransferase, U/L 19 (11–33.5) estimates are skewed as indicated by the difference between
qSOFA Score the mean and median values. The cause of this is likely related
0, n (%) 11 (22) to the heterogeneous patient population included in the study.
1, n (%) 28 (56)
≥ 2, n (%) 11 (22) 
CrCl
SIRS Score Ceftriaxone Clearance ¼ TVNR þ TVR  Þ ð1Þ
1, n (%) 4 (8)
63
2, n (%) 15 (30)
3, n (%) 22 (15)
4, n (%) 9 (18) Dosing simulations
Infection source
Gastrointestinal (%) 3 (6) The PTA for common ceftriaxone dosing regimens is present-
Respiratory (%) 43 (86) ed in Fig. 2. The probability of attaining a therapeutic expo-
Urinary tract (%) 3 (6) sure (100% fT>MIC) within the first 24 h of therapy in ≥ 90%
Unknown (%) 1 (2) of patients receiving a 1 g once-daily dose is only likely for
Causative Pathogen patients with an impaired renal function (CrCL ≤ 60 mL/min)
(total n, susceptible n) for an isolate with an MIC ≥ 0.25 mg/L. Patients with a CrCl
S. pneumoniae 2, 2 between 60 and 120 mL/min require a minimum of a 2 g once-
H. influenzae 2, 1 daily dose for optimal activity against susceptible bacteria per
C. perfringens 1, 0 current EUCAST clinical breakpoints [27]. When considering
Influenza A 2, NA
E. coli 2, 2
S. aureus 3, 3
Parainfluenzae 3 1, NA

IQR, interquartile range; N, number

Pharmacokinetic model

The unbound ceftriaxone concentration time profile was ade-


quately described using a one-compartment model with linear
elimination incorporating an additive error model where error
= (standard deviation2 + 0.32)0.5 and the standard deviation is
Fig. 1 Observed unbound ceftriaxone concentration time data and visual
0.1 + 0.012. The model fit was improved by including a typ- predictive check of the final model. Blue lines represent the model
ical population value for non-renal clearance (TVNR) and the predicted 5th and 95th centiles. Red lines represent the model predicted
typical value for renal clearance (TVR) incorporating creati- 25th and 75th quartiles. Filled dots represent observed patient unbound
ceftriaxone concentrations.
nine clearance (CrCl) normalised to the population median
Eur J Clin Pharmacol

Table 2 Ceftriaxone pharmacokinetic model parameter estimates Discussion


Parameter Mean (SD) Median CV%
The present study has demonstrated that a 1 g once-daily dose
TVNR (L/h) 4.27 (4.77) 1.66 111.66 is unlikely to achieve a therapeutic ceftriaxone exposure in
TVR (L/h) 4.83 (3.69) 4.70 76.43 patients presenting to the ED with a creatinine clearance ≥
Vd (L) 116.21 (62.22) 99.87 53.54 60 mL/min, infected with a susceptible pathogen with an
MIC ≥ 0.25 mg/L. Overall, a 2 g once-daily dose is likely to
TVNR, non-renal ceftriaxone clearance; TVR, renal ceftriaxone clearance;
be optimal in most adult patients admitted to the hospital from
Vd, volume of distribution
the ED with sepsis. The use of a once-daily dose is also con-
venient and may limit the potential for a delayed second anti-
biotic dose that can occur between patient transfers from the
the FTA for specific bacterial species, a 1 g once-daily ceftri- ED to another team [28].
axone dose is adequate for H. influenzae for patients with any A recent study by Bos and colleagues conducted in adult
renal function, including augmented renal clearance (Table 3). patients with sepsis from Africa who were not admitted to the
In contrast, a 1 g once-daily dose is insufficient for patients ICU showed that a dose of 2 g twice-daily may be required to
with a CrCl ≥ 120 mL/min against K. pneumoniae, E. coli, and achieve the same therapeutic exposure (100% fT>MIC with an
S. pneumoniae. Increasing the ceftriaxone dose to 2 g once- isolate MIC of 0.5 mg/L) as targeted in our study [29].
daily will reliably achieve a fT>MIC of 100% for ≥ 90% of all Furthermore, the mean unbound volume of distribution and
isolates, even in patients with augmented renal clearance clearance identified by Bos et al. was 48 L and 11 L/h com-
(Table 3). pared with our values of 116.87 L and 9.1 L/h respectively.

Table 3 Fractional target attainment for different ceftriaxone dosing regimens and bacterial species

Dosing regimen and CrCL (mL/min) Fractional target attainment (%) for 100% fT>MIC by bacterial species

E. coli H. influenzae K. pneumoniae S. pneumoniae

1 g once-daily
20 100 99 99 98
40 98 99 97 96
60 97 97 97 94
80 97 96 96 92
100 95 95 93 89
120 93 93 90 85
140 89 90 85 81
1 g twice-daily
20 100 100 100 100
40 100 100 100 100
60 100 100 100 100
80 100 100 99 99
100 100 100 99 99
120 98 99 98 98
140 98 99 98 97
2 g once-daily
20 100 100 100 100
40 100 100 100 100
60 100 100 100 100
80 100 100 99 99
100 100 100 99 99
120 98 99 98 98
140 98 99 98 97
2 g twice-daily
20 100 100 100 100
40 100 100 100 100
60 100 100 100 100
80 100 100 100 100
100 100 100 99 99
120 99 99 99 99
140 98 99 98 98
Eur J Clin Pharmacol

Although the ceftriaxone model included total and unbound Joynt et al. [20] described a two-compartment pharmacokinet-
concentrations from an intensive sampling strategy in the ic model for ceftriaxone in patients admitted to the ICU, with a
study conducted by Bos et al., the key difference between Vd of the central compartment of 6 L and a clearance of 9.2 h;
studies is the patient cohort. Bos et al. primarily included a however, this represents the total, not the unbound ceftriax-
younger patient cohort (median age 35 years, range 18–76) one. In contrast, our unbound ceftriaxone mean Vd was
with a lower albumin (median 29 g/L, range 11-44) who were 116.87 L with a total average clearance of 9.1 L/h, which is
also seemingly of a much lower weight (mean BMI 18.9 kg/ related to the different (total or unbound) concentrations
m2) compared with our study [29]. Moreover, both our study assayed. A separate pharmacokinetic study, also conducted
and the study conducted by Bos et al., like most pharmacoki- in ICU patients [33], identified an unbound total volume of
netic studies, only modelled plasma ceftriaxone concentra- distribution of 87.5 L and unbound clearance of 10.61 L/h,
tions and not the antibiotic concentration at the site of which is similar to that identified in our study. Therefore, the
infection. differences in dosing recommendations between studies (the
In the case of patients with pneumonia, infection often current study 2 g once-daily compared with 1–2 g twice-daily
involves the epithelial lining fluid (ELF) [30, 31]. There is recommended by Ollivier et al.) likely pertain to the lower
no currently available data regarding ceftriaxone ELF concen- albumin concentrations in patients admitted to the ICU and
trations, which may be as low as 20% that of concurrent plas- potential augmented renal clearance observed in such patients
ma concentrations in the case of many other β-lactam antibi- [33]. It has been previously demonstrated that ceftriaxone un-
otics [31]. Although there is limited data regarding target site dergoes biliary (non-renal) elimination [34]. This would likely
antibiotic concentrations and outcomes, current evidence account for our observation that the pharmacokinetic model
would suggest that optimising the antibiotic concentration at was improved by incorporating a non-renal clearance
the site of infection would improve bactericidal activity parameter.
[30–32]. This may be an important consideration given the Despite the potential benefits of dose optimisation, the con-
low FTA in plasma samples for the current 1 g once-daily cern of clinicians may be that increasing the dose from the
ceftriaxone dosing, particularly for patients with manufacturer recommendation of 1 g once daily may both
S. pneumoniae (Table 3). increase the potential for dose-related adverse events. Data
Other ceftriaxone pharmacokinetic studies have been con- regarding ceftriaxone adverse events is lacking, but this risk
ducted in critically ill patients admitted to ICU which are is likely low even with doses exceeding 4 g/day in patients
likely to be a more seriously affected cohort than studied here. without renal dysfunction [33]. In a small study, the absolute

a Ceftriaxone 1 g once-daily b Ceftriaxone 1 g twice-daily


1.0 1.0
CrCl 20 mL/min CrCl 20 mL/min
Probability of Success

Probability of Success

0.9 0.9
0.8 CrCl 40 mL/min 0.8 CrCl 40 mL/min
0.7 0.7
CrCl 60 mL/min CrCl 60 mL/min
0.6 0.6
0.5 CrCl 80 mL/min 0.5 CrCl 80 mL/min
0.4 CrCl 100 mL/min 0.4 CrCl 100 mL/min
0.3 0.3
CrCl 120 mL/min CrCl 120 mL/min
0.2 0.2
0.1 CrCl 140 mL/min 0.1 CrCl 140 mL/min
0.0 0.0
0.032 0.064 0.125 0.25 0.5 1 0.032 0.064 0.125 0.25 0.5 1
Pathogen MIC (mg/L) Pathogen MIC (mg/L)

c Ceftriaxone 2 g once-daily
d Ceftriaxone 2 g twice-daily
1.0 1.0
CrCl 20 mL/min CrCl 20 mL/min
Probability of Success

Probability of Success

0.9 0.9
0.8 CrCl 40 mL/min 0.8 CrCl 40 mL/min
0.7 0.7
CrCl 60 mL/min CrCl 60 mL/min
0.6 0.6
0.5 CrCl 80 mL/min 0.5 CrCl 80 mL/min
0.4 CrCl 100 mL/min 0.4 CrCl 100 mL/min
0.3 0.3
CrCl 120 mL/min CrCl 120 mL/min
0.2 0.2
0.1 CrCl 140 mL/min 0.1 CrCl 140 mL/min
0.0 0.0
0.032 0.064 0.125 0.25 0.5 1 0.032 0.064 0.125 0.25 0.5 1
Pathogen MIC (mg/L) Pathogen MIC (mg/L)

Fig. 2 Probability of target attainment (unbound ceftriaxone concentration 100% fT>MIC) for ceftriaxone administered as a 1 g once-daily dose (a), 1 g
twice-daily dose (b), 2 g once-daily dose (c), and 2 g twice-daily dose (d)
Eur J Clin Pharmacol

risk of a suspected ceftriaxone adverse reaction was 8.7%, Acknowledgements Aaron J. Heffernan would like to acknowledge
funding from a Griffith School of Medicine Research Higher degree
which was associated with total trough concentrations ≥ 100
scholarship; Fekade B. Sime acknowledges funding from the University
mg/L, greatly exceeding the target exposure used for dosing of Queensland Post-Doctoral Fellowship (W. T. Allen Bequest);and
simulations in our study [35]. Together, the low risk of dose- Jason Roberts would like to acknowledge funding for a National Health
related adverse events and the potential benefits may promote and Medical Research Council (NHMRC) Centre of Research Excellence
(APP1099452), an NHMRC Project Grant (APP1062040) and a
the adoption of a convenient 2 g once-daily or 1 g twice-daily
Practitioner Fellowship (APP1117065).
dosing regimen in most patients with sepsis.
Authors’ contributions AJH—study design, data collection, data
curation, data analysis, and writing—original draft
RAC—study design, data collection, writing—original draft, review,
Limitations and editing
KJD—study design, data analysis, writing, review, and editing
Our study is not without limitations. First, a sparse sampling FBS—data analysis, writing, review, and editing
CLS—study design, writing, review, and editing
strategy was used that may result in imprecise parameter esti- BMcW—study design, sample analysis, writing, review, and editing
mates within a specific patient’s dosing interval. Nonetheless, the JU—study design, sample analysis, writing, review, and editing
data presented herein are similar to previous studies conducted in JAR—study design, data analysis, writing, review, and editing
intensive care unit patients receiving a 2 g once-daily dose [20]. JL—study design, project administration, funding acquisition, writing,
review, and editing
Second, samples were not taken on different days of therapy to
assess temporal changes in ceftriaxone pharmacokinetics. This is Data availability Data will not be shared per specific ethics requirements.
particularly relevant given that patient’s physiology may fluctu-
ate with time and biological markers, such as creatinine, and may Compliance with ethical standards
not reflect the current physiological state. Third, only the un-
bound ceftriaxone concentration was assayed and as such we Conflict of interest The authors declare that they have no conflict o
were unable to account for changes in ceftriaxone binding that interest.
is highly likely to influence the extent of tissue distribution and
vary with albumin concentration. Fourth, the small sample size Ethics statement Waiver of informed consent was granted by the Gold
Coast University Hospital Ethics Committee HREC/18/QGC/100.
including predominantly older patients may not generalise to a
younger patient cohort. Fifth, the MIC distribution for the FTA Code availability R code and model files will be available on specific
was obtained from the EUCAST database, which may reflect a reasonable request.
different pathogen MIC distribution depending on the geograph-
ical location. Sixth, as only patients receiving a 1 g once-daily
ceftriaxone dose were included, future studies are required to
determine if the recommended 2 g once-daily dose is able to References
achieve the desired targets as demonstrated in our simulations.
1. Antibiotic Expert Group (2014) Therapeutic guidelines: antibiotic,
Seventh, sepsis was defined using the SIRS criteria, which does version 15. Therapeutic Guidelines Limited, Melbourne
not reflect the latest Sepsis-3 guidelines [21]; nonetheless, we 2. Kumar A, Ellis P, Arabi Y, Roberts D, Light B, Parrillo JE, Dodek
have shown in this ill patient cohort that a dose of 2 g once- P, Wood G, Kumar A, Simon D, Peters C, Ahsan M, Chateau D
daily is the minimum dose that is likely to achieve target expo- (2009) Initiation of inappropriate antimicrobial therapy results in a
fivefold reduction of survival in human septic shock. Chest 136:
sures. Last, patient-centered outcomes such as mortality or tox-
1237–1248
icity were not assessed in this study—an important focus for 3. Craig WA (1998) Pharmacokinetic/pharmacodynamic parameters:
future studies assessing different dosing regimens. Rationale for antibacterial dosing of mice and men. Clin Infect Dis
26:1–10
4. McKinnon PS, Paladino JA, Schentag JJ (2008) Evaluation of area
under the inhibitory curve (AUIC) and time above the minimum
Conclusions inhibitory concentration (T > MIC) as predictors of outcome for
cefepime and ceftazidime in serious bacterial infections. Int J
Antimicrob Agents 31:345–351
In conclusion, based on the potential benefits of optimised
5. Shah MN, Schmit J, Croley WC, Meltzer D (2003) Continuity of
ceftriaxone dosing and the limited risk of adverse events, we antibiotic therapy in patients admitted from the emergency depart-
would recommend the routine use of a 2 g once-daily dose of ment. Ann Emerg Med 42:117–123
ceftriaxone for the management of adult patients with sepsis. 6. Denny KJ, Gartside JG, Alcorn K, Cross JW, Maloney S, Keijzers
Future studies should be conducted determining if there is a G (2018) Appropriateness of antibiotic prescribing in the emergen-
cy department. J Antimicrob Chemother 74:515–520
clinical benefit for the use of a higher daily dose of 1 g twice- 7. Roberts JA, Abdul-Aziz MH, Lipman J, Mouton JW, Vinks AA,
daily or 2 g once-daily as compared with a 1 g once-daily Felton TW, Hope WW, Farkas A, Neely MN, Schentag JJ, Drusano
regimen. G, Frey OR, Theuretzbacher U, Kuti JL (2014) Individualised
Eur J Clin Pharmacol

antibiotic dosing for patients who are critically ill: challenges and 22. Tam VH, Kabbara S, Yeh RF, Leary RH (2006) Impact of sample
potential solutions. Lancet Infect Dis 14:498–509 size on the performance of multiple-model pharmacokinetic simu-
8. Schleibinger M, Steinbach CL, Töpper C, Kratzer A, Liebchen U, lations. Antimicrob Agents Chemother 50:3950–3952
Kees F, Salzberger B, Kees MG (2015) Protein binding character- 23. Briscoe SE, McWhinney BC, Lipman J, Roberts JA, Ungerer JP
istics and pharmacokinetics of ceftriaxone in intensive care unit (2012) A method for determining the free (unbound) concentration
patients. Br J Clin Pharmacol 80:525–533 of ten beta-lactam antibiotics in human plasma using high perfor-
9. Tsai D, Stewart P, Goud R, Gourley S, Hewagama S, mance liquid chromatography with ultraviolet detection. J
Krishnaswamy S, Wallis SC, Lipman J, Roberts JA (2016) Total Chromatogr B 907:178–184
and unbound ceftriaxone pharmacokinetics in critically ill 24. Neely MN, van Guilder MG, Yamada WM, Schumitzky A, Jelliffe
Australian Indigenous patients with severe sepsis. Int J RW (2012) Accurate detection of outliers and subpopulations with
Antimicrob Agents 48:748–752 Pmetrics, a nonparametric and parametric pharmacometric model-
10. Roberts JA, Paul SK, Akova M, Bassetti M, Waele JJD, ing and simulation package for R. Ther Drug Monit 34:467–476
Dimopoulos G, Kaukonen K, Koulenti D, Martin C, Montravers 25. Bustad A, Terziivanov D, Leary R, Port R, Schumitzky A, Jelliffe R
P, Rello J, Rhodes A, Starr T, Wallis SC, Lipman J (2014) DALI: (2006) Parametric and nonparametric population methods. Clin
defining antibiotic levels in intensive care unit patients: are current Pharmacokinet 45:365–383
beta-lactam antibiotic doses sufficient for critically ill patients? Clin 26. Sunder S, Jayaraman R, Mahapatra HS, Sathi S, Ramanan V,
Infect Dis 58:1072–1083 Kanchi P, Gupta A, Daksh S, Ram P (2014) Estimation of renal
11. Patel IH, Chen S, Parsonnet M, Hackman MR, Brooks MA, function in the intensive care unit: the covert concepts brought to
Konikoff J, Kaplan SA (1981) Pharmacokinetics of ceftriaxone in light. J Intensive Care 2:31. https://doi.org/10.1186/2052-0492-2-
humans. Antimicrob Agents Chemother 20:634–641 31
12. Pollock AA, Tee PE, Patel IH, Spicehandler J, Simberkoff MS, 27. The European Committee on Antimicrobial Susceptibility Testing
Rahal JJ Jr (1982) Pharmacokinetic characteristics of intravenous (2019) Breakpoint tables for interpretation of MICs and zone diam-
ceftriaxone in normal adults. Antimicrob Agents Chemother 22: eters, version 9.0. Available from: http://eucast.org/clinical_
816–823 breakpoints. Accessed 15 Sept 2019
13. Esteban A, Frutos-Vivar F, Ferguson ND, Penuelas O, Lorente JA, 28. Leisman D, Huang V, Zhou Q, Gribben J, Bianculli A, Bernshteyn
Gordo F et al (2007) Sepsis incidence and outcome: contrasting the M, Ward MF, Schneider SM (2017) Delayed second dose antibi-
intensive care unit with the hospital ward. Crit Care Med 35:1284– otics for patients admitted from the emergency department with
1289 sepsis: prevalence, risk factors, and outcomes. Crit Care Med 45:
14. Glickman SW, Cairns CB, Otero RM, Woods CW, Tsalik EL, 956–965
Langley RJ, van Velkinburgh JC, Park LP, Glickman LT, Fowler 29. Bos JC, Prins JM, Misticio MC, Nunguiane G, Lang CN, Beirao JC
VG Jr, Kingsmore SF, Rivers EP (2010) Disease progression in et al (2018) Pharmacokinetics and pharmacodynamic target attain-
hemodynamically stable patients presenting to the emergency de- ment of ceftriaxone in adult severely ill sub-Saharan African pa-
partment with sepsis. Acad Emerg Med 17:383–390 tients: a population pharmacokinetic modelling study. J Antimicrob
15. Adult Sepsis Pathway. Queensland Government (2019) Available Chemother 73:1620–1629
from: https://clinicalexcellence.qld.gov.au/priority-areas/safety-
30. Jager NGL, van Hest RM, Lipman J, Roberts JA, Cotta MO (2019)
and-quality/resist-sepsis-program/adult-sepsis. Accessed 17
Antibiotic exposure at the site of infection: principles and assess-
Aug 2019
ment of tissue penetration. Expert Rev Clin Pharmacol 12:623–634
16. Metlay JP, Waterer GW, Long AC, Anzueto A, Brozek J, Crothers
31. Heffernan AJ, Sime FB, Lipman J, Dhanani J, Andrews K, Ellwood
K, Cooley LA, Dean NC, Fine MJ, Flanders SA, Griffin MR,
D, Grimwood K, Roberts JA (2019) Intrapulmonary pharmacoki-
Metersky ML, Musher DM, Restrepo MI, Whitney CG (2019)
netics of antibiotics used to treat nosocomial pneumonia caused by
Diagnosis and treatment of adults with community-acquired pneu-
Gram-negative bacilli: a systematic review. Int J Antimicrob
monia. An official clinical practice guideline of the American
Agents 53:234–245
Thoracic Society and Infectious Diseases Society of America. Am
32. Steed ME, Vidaillac C, Winterfield P, Biek D, Rybak MJ (2012)
J Respir Crit Care Med 200:e45–e67
Evaluation of ceftaroline activity versus ceftriaxone against clinical
17. Solomkin JS, Mazuski JE, Bradley JS, Rodvold KA, Goldstein EJ,
isolates of Streptococcus pneumoniae with various susceptibilities
Baron EJ et al (2010) Diagnosis and management of complicated
to cephalosporins in an in vitro pharmacokinetic/pharmacodynamic
intra-abdominal infection in adults and children: guidelines by the
model. Antimicrob Agents Chemother 56:2691–2695
Surgical Infection Society and the Infectious Diseases Society of
America. Clin Infect Dis 50:133–164 33. Ollivier J, Carrié C, d’Houdain N, Djabarouti S, Petit L, Xuereb F,
18. Ceftriaxone Product Information. Therapeutic goods administration Legeron R, Biais M, Breilh D (2019) Are standard dosing regimens
Available from: https://www.ebs.tga.gov.au/ebs/picmi/ of ceftriaxone adapted for critically ill patients with augmented
picmirepository.nsf/pdf?OpenAgent&id=CP-2010-PI-02576- creatinine clearance? Antimicrob Agents Chemother 63:e02134–
3&d=2017123016114622483. Accessed 26 Jan 2020 e02118
19. Hasegawa S, Sada R, Yaegashi M, Mori T (2018) 1975. 1 g vs. 2 g 34. Hayton WL, Schandlik R, Stoeckel K (1986) Biliary excretion and
daily intravenous ceftriaxone in the therapy of community-onset pharmacokinetics of ceftriaxone after cholecystectomy. Eur J Clin
pneumonia: a propensity score analysis from a data of Japanese Pharmacol 30:445–451
Multicenter Registry. Open Forum Infect Dis 5:S573 35. Le Turnier P, Navas D, Dalichampt M, Dailly E, Garot D, Guimard
20. Joynt GM, Lipman J, Gomersall CD, Young RJ, Wong ELY, Gin T T, Bernard L, Tattevin P, Vandamme Y, Hoff J, Chiffoleau A,
(2001) The pharmacokinetics of once-daily dosing of ceftriaxone in Leclair L, Sebille V, Asseray Madani N (2017) Is there a concen-
critically ill patients. J Antimicrob Chemother 47:421–429 tration threshold for ceftriaxone toxicity? Preliminary results of
21. Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, high-dose ceftriaxone in central nervous system infections, a pro-
Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, spective multicenter cohort study. European Congress Clinical
Coopersmith CM, Hotchkiss RS, Levy MM, Marshall JC, Martin Microbiology
GS, Opal SM, Rubenfeld GD, van der Poll T, Vincent JL, Angus
DC (2016) The Third International Consensus Definitions for Publisher’s note Springer Nature remains neutral with regard to jurisdic-
Sepsis and Septic Shock (Sepsis-3). J Am Med Assoc 315:801–810 tional claims in published maps and institutional affiliations.

You might also like