J Antimicrob Chemother 2013; 68: 648 – 658
doi:10.1093/jac/dks442 Advance Access publication 27 November 2012
Activity of ceftaroline against extracellular (broth) and
intracellular (THP-1 monocytes) forms of methicillin-resistant
Staphylococcus aureus: comparison with vancomycin, linezolid
and daptomycin
Aurélie Mélard1, Laetitia G. Garcia1, Debaditya Das1, Raoul Rozenberg2, Paul M. Tulkens1*, Françoise Van Bambeke1
and Sandrine Lemaire1
1
Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium; 2Analyse
structurale moléculaire, Institute of Condensed Matter and Nanosciences, Université catholique de Louvain, Louvain-la-Neuve, Belgium
*Corresponding author. Pharmacologie cellulaire et moléculaire, Université catholique de Louvain, Avenue E. Mounier 73 B1.73.05, B-1200 Brussels,
Belgium. Tel: +32-2-7647371; Fax: +32-2-7647373; E-mail:
[email protected]
Received 22 August 2012; returned 25 September 2012; revised 26 September 2012; accepted 11 October 2012
Background: Ceftaroline fosamil is approved for treatment of acute bacterial skin and skin structure infections
caused by methicillin-resistant Staphylococcus aureus (MRSA). We examined the activity of its active metabolite
(ceftaroline) against intracellular forms of S. aureus in comparison with vancomycin, daptomycin and linezolid.
Methods: Two methicillin-susceptible S. aureus (MSSA) and 11 MRSA strains with ceftaroline MICs from 0.125 to
2 mg/L [two strains vancomycin- and one strain linezolid-resistant (EUCAST interpretative criteria); VISA and
cfr+] were investigated. The activity was measured in broth and after phagocytosis by THP-1 monocytes in
concentration-dependent experiments (24 h of incubation) to determine: (i) relative potencies (EC50) and
static concentrations (Cs) (mg/L and ×MIC); and (ii) relative activities at human Cmax (ECmax) and maximal
relative efficacies (Emax) (change in log10 cfu compared with initial inoculum). Ceftaroline stability and cellular
accumulation (at 24 h) were measured by mass spectrometry.
Results: Ceftaroline showed similar activities in broth and in monocytes compared with vancomycin, daptomycin and linezolid, with no impact of resistance mechanisms to vancomycin or linezolid. For all four antibiotics,
intracellular ECmax and Emax were considerably lower than in broth (≏0.5 log10 versus 4– 5 log10 cfu decrease),
but the EC50 and Cs showed comparatively little change (all values between ≏0.3 and ≏6× MIC). The mean
cellular to extracellular ceftaroline concentration ratios (20 mg/L; 24 h) were 0.66+0.05 and 0.90+0.36 in
uninfected and infected cells, respectively.
Conclusion: In vitro, ceftaroline controls the growth of intracellular MRSA to an extent similar to that of
vancomycin, linezolid and daptomycin for strains with a ceftaroline MIC ≤2 mg/L.
Keywords: Hill equation, maximal relative efficacy, static concentration, recursive partitioning analysis, mass spectrometry, VISA,
linezolid resistant, cfr
Introduction
Ceftaroline,1 originally known as T-91825, is a novel cephalosporin
with in vitro activities against methicillin-resistant Staphylococcus
aureus (MRSA) comparable to those of vancomycin, linezolid and
daptomycin towards susceptible strains,2,3 and with unimpaired
activity against strains non-susceptible (VISA) or resistant (VRSA)
to vancomycin.4 In in vitro time–kill studies, ceftaroline is also
more rapidly cidal against MRSA than vancomycin and linezolid.2
Developed for clinical use as a water-soluble N-phosphono
prodrug [ceftaroline fosamil (TAK-599)],5 it has proven efficacious,
thus receiving approval in the USA and the EU for the treatment
of acute bacterial skin and skin structure infections caused by
susceptible organisms including MRSA.6 – 9a In these studies a numerically higher clinical response was achieved for ceftaroline
over a vancomycin/aztreonam combination at an early stage
of treatment.10 Ceftaroline fosamil has also been approved for
the treatment of community-acquired bacterial pneumonia
caused by susceptible organisms including methicillinsusceptible S. aureus (MSSA) and Streptococcus pneumoniae.
These studies compared ceftaroline with ceftriaxone and again
# The Author 2012. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved.
For Permissions, please e-mail:
[email protected]
648
Ceftaroline and intracellular MRSA
ceftaroline clinical response rates were numerically superior to
those with the comparator agent.
While historically considered as an extracellular organism
only, there is now increasing evidence that S. aureus invades,
sojourns and thrives intracellularly.11,12 This creates a potential
therapeutic challenge for clinicians, since it has been clearly
documented using in vitro and in vivo models that most antistaphylococcal agents are considerably less active against intracellular S. aureus than expected given their respective intrinsic
activity and/or level of cellular accumulation.13,14 Evaluation of
novel antistaphylococcal agents must therefore include an
assessment of their ability to control intracellular infections.
In this context, we observed that, contrary to most assumptions,
b-lactams actually display significant intracellular activity
against S. aureus, despite a reported poor cellular accumulation.15,16 In a previous study we also showed that ceftobiprole,
another anti-MRSA cephalosporin,17 was equally active against
intracellular forms of MSSA and MRSA, regardless of origin (community or hospital acquired) and resistance phenotype towards
vancomycin.18 The present study extends these observations to
ceftaroline and compares it with vancomycin, daptomycin and
linezolid using a panel of strains with increasing MICs towards
these antibiotics [including VISA and linezolid resistant (LZDR)].
Our study used a previously established pharmacodynamic
model of infected human THP-1 monocytes that allows a quantitative assessment of key properties such as intracellular
maximal efficacy and potency of antibiotics.15 We show that
all four antibiotics display similar intracellular efficacy, with the
activity of ceftaroline remaining essentially unimpaired across
all strains investigated up to the highest MIC of ceftaroline
observed (2 mg/L).
Materials and methods
Materials
Ceftaroline (potency 85.3%; lot no. FMD-CEF-035) was provided by Forest
Laboratories, Inc. (New York, NY, USA). Oxacillin was purchased from
Sigma– Aldrich (St Louis, MO, USA). Other antibiotics were obtained as
the corresponding branded products for human parenteral use distributed for clinical use in Belgium (gentamicin as Geomycinew, GlaxoSmithKline, Wavre, Belgium; vancomycin as Vancomycin Sandozw, Sandoz n.v.,
Vilvoorde, Belgium; and linezolid as Zyvoxidw, Pfizer s.a., Brussels,
Belgium) or in France (daptomycin as Cubicinw, Novartis Europharm
Ltd, Horsham, UK). Culture media and sera were from Invitrogen Corporation (Carlsbad, CA, USA) and Becton Dickinson (Franklin Lakes, NJ, USA),
and other reagents were from Sigma–Aldrich or Merck KGaA (Darmstadt,
Germany).
Bacterial strains and susceptibility testing
The clinical isolates used in the present study are listed in Table 1, with
information on their origin and their resistance phenotypes. MICs were
determined following the general recommendations of the CLSI19 for
vancomycin, linezolid and daptomycin (with addition of Ca2+ for daptomycin). For ceftaroline, we used both plain and cation-adjusted
Mueller– Hinton (MH) broth, but no difference was observed between
these two media. There was also no effect of the addition of 2% NaCl
to MH broth. MICs were also measured in broth adjusted to pH 5.5 to
mimic the phagolysosomal environment where S. aureus sojourns after
phagocytosis.20,21 Strains for which ceftaroline showed an MIC ≥1 mg/L
were retested using arithmetic dilutions (0.25 mg/L intervals) between
JAC
1 and 4 mg/L to determine their MICs in a more accurate fashion in
that range than with the conventional geometric (log2) dilution method.
Cell lines, cell infection and determination of cell viability
Experiments were performed with THP-1 cells (ATCC TIB-202; supplied
through LGC Promochem Ltd, Teddington, UK), a human myelomonocytic
cell line displaying macrophage-like activity,22 maintained in our laboratory as previously described.23 Cell infection was performed as previously
described.15,16,18 Briefly, S. aureus in the stationary phase (overnight
culture) were opsonized in the presence of 10% human serum (Lonza
Ltd, Basel, Switzerland) in RPMI-1640 medium and mixed with THP-1
cells (0.5×106 cells/mL) for 1 h at a ratio of four bacteria per macrophage
after which extracellular and non-internalized bacteria were eliminated
by washing and exposure to gentamicin (100× MIC; 45 min). This
yielded a typical post-phagocytosis bacterial load of 1 –3×106 cfu/mg
of cell protein. Cells were thereafter incubated for 24 h at 378C. Cell viability was checked by measuring the release of lactate dehydrogenase
and trypan blue exclusion assay.
Determination of the extracellular and intracellular
activities of antibiotics (concentration– response curves)
and pharmacodynamic descriptors
Extracellular and intracellular activities were measured at a fixed timepoint (24 h) using a large array of antibiotic concentrations (typically
from 0.01 to 100× MIC) to obtain a complete description of the
concentration-dependent response, as described in detail in our previous
publications.15,16,18 For extracellular activity, experiments were performed in MH broth (supplemented with Ca2+ for daptomycin) with an
initial inoculum of 106 cfu/mL and results expressed as the change in
cfu/mL from the initial inoculum as measured by colony counting. Bactericidal activity was defined as a reduction of 99.9% (≥3 log10 cfu/mL
decrease) of the total counts. For intracellular activity, infected THP-1
cells were collected by centrifugation, washed once in PBS and lysed in
distilled water. The resulting solution was analysed for protein content
(using the Folin-Ciocalteu/biuret method)24 and were plated on
TrypticaseTM soy agar (Becton Dickinson) to enumerate bacteria as
described in detail in a previous publication (including validation and
determination of the lowest limit of detection).15 Results were expressed
as the change in cfu/mg of cell protein. Data were used to fit a sigmoidal
function (Hill equation; slope factor¼1) by non-linear regression (GraphPad Prismw version 4.03; GraphPad Software, La Jolla, CA, USA) to obtain
for each condition numeric values of five key pharmacodynamic descriptors, namely: (i) the increase in the number of cfu for an infinitely low
antibiotic concentration [relative minimal efficacy (Emin; in log10 cfu
units)] compared with the original inoculum; (ii) the decrease in the
number of cfu for an infinitely large concentration of antibiotic [relative
maximal efficacy (Emax; in log10 cfu units); limit of detection
≏5.5 log10 cfu decrease from the original inoculum]; (iii) the decrease
in the number of cfu at a concentration corresponding to the maximal
serum concentration (Cmax) of the drug as observed in humans receiving
standard therapies (ECmax; in log10 cfu); (iv) the concentration of antibiotic
yielding a response halfway between Emin and Emax [relative potency
(EC50; in mg/L or in multiples of MIC)]; and (v) the concentration of antibiotic
resulting in no apparent bacterial growth compared with the original inoculum [static concentration (Cs; in mg/L or in multiples of MIC)].
Stability of ceftaroline and measurement of its cellular
to extracellular concentration ratio
Ceftaroline stability at 378C over 24 h in water, broth (adjusted to pH 7.4
and 5.5) and in the cell culture medium (pH ≏7.4) was checked by
649
Mélard et al.
measuring its MIC for S. aureus ATCC 25923 using an arithmetic dilution
progression (0.1 mg/L intervals) starting from media containing 10 or
100 mg/L. In parallel, broth and culture medium samples incubated
with 20 mg/L ceftaroline and cell samples from uninfected and infected
THP-1 cells incubated with 20 mg/L ceftaroline for 24 h (collected by centrifugation, followed by washing in PBS and final resuspension in distilled
water) were used for measurement of ceftaroline concentrations using
liquid chromatography (LC) and tandem mass spectrometry (MS/MS).
In brief, samples (100 mL) were mixed with 1.125 mL of methanol/acetonitrile (4: 21, v/v), stored at 2208C for 30 min (to facilitate protein
denaturation), thawed and centrifuged. The supernatant was collected,
evaporated to dryness under a gentle stream of nitrogen and resuspended in 100 mL of methanol/water (1: 1) with care to obtain full dissolution of any visible material. Samples were then subjected to LC
separation using a ThermoFischer LC system equipped with a C18
XBridge column (150×2.1 mm, i.d. 3.5 mm) (Waters Corp., Milford, MA,
USA) and using 100 mM ammonium formate/water/methanol/isopropanol (100: 780: 80: 40, v/v/v/v) as eluent and a flow rate of 0.2 mL/min.
Chromatography was performed at 308C, but samples were maintained
at 78C prior to injection (10 mL). High collision dissociation spectra were
recorded with a Q-Exactive in LC-MS/MS [quadrupole precursor selection
with accurate mass (HR/AM) Orbitrap detection; ThermoFisher Scientific,
Waltham, MA, USA] at relative collision energy of 40%. Multiple reaction
monitoring mode was used for the quantification of the analytes by
monitoring the transition m/z 605 208 by high-resolution mass spectrometry. Calculation of the actual concentration was made using a calibration curve [external standard; linearity 1– 5000 ng/mL (R 2 ¼0.9976);
limit of detection and limit of quantification: 0.1 and 0.5 ng/mL,
respectively] and corrected for actual extraction efficiency from cells by
running samples of control cells to which a known amount of ceftaroline
had been added and which were then treated exactly as the samples
from incubated cells. The cell content in ceftaroline was expressed as
ng/mg of cell protein and the ratio of the apparent cellular to extracellular concentrations was calculated using a conversion factor of 5 mL of cell
volume per mg of cell protein, as in our previous publications.15
Statistical analyses
Statistical analyses of the differences between values of the pharmacological descriptors were made with GraphPad Instat version 3.06 (GraphPad Software). Recursive partitioning analysis was made with JMP version
9.0.3 (SAS Institute, Cary, NC, USA) using a single-pass decision tree
method with node splitting based on the LogWorth statistic (see
details and justification in the white paper ‘Monte Carlo Calibration of
Distributions of Partition Statistics’, available at http://www.jmp.com).
Results
Strains and susceptibility to ceftaroline and
comparator antibiotics at neutral and acid pH
Table 1 shows that the MICs of ceftaroline at pH 7.4 for the
strains used in this study ranged from 0.125 to 0.25 mg/L for
MSSA and from 0.25 to 2 mg/L for MRSA irrespective of their
resistance phenotype to vancomycin (range 0.5–4 mg/L),
Table 1. Strains used in this study (origin, resistance phenotype and MICs in broth at neutral and acidic pH)
MIC (mg/L)a
ceftaroline
Strain
ATCC 25923b
34843/33134c
ATCC 33591b
19210/18057c
SA 555d
SA 1984d
36065/34090c
NRS18e
35165/33258c
48046/44800c
CM05g
062-13091 Ac
062-13101 Ac
vancomycin
daptomycin
linezolid
Resistance phenotype
pH 7.4
pH 5.5
pH 7.4
pH 5.5
pH 7.4
pH 5.5
pH 7.4
pH 5.5
MSSA
MSSA
MRSA
MRSA
MRSA/VISA
MRSA
MRSA
MRSA/VISA
MRSA
MRSA
MRSA/LZDR
MRSA
MRSA
0.125
0.25
0.5
0.5
0.5
0.5
0.25
0.5–1
1 (0.75)f
2 (2.25)f
2 (1.75)f
2 (1.75)f
2 (2)e
0.125
0.125–0.25
0.25
0.25
0.125–0.25
0.25
0.125–0.25
0.25
0.25
0.5
0.5–1
1
0.5
0.5
1
0.5
0.5
4
0.5
2
2 –4
0.5
1
1 –2
1
1
0.5– 1
1
1
1
2
0.5
2
2
1
1
0.5– 1
2
2
0.125– 0.25
1
0.25– 0.5
0.5
4
0.25– 0.5
1
1 –2
0.5
1
0.5–1
0.25
0.25
0.5
1
0.5
1
ND
2
2
2
1–2
2
0.5– 1
1
1
1
4
1– 2
2
0.5
1
4
0.5–2
4
2
4– 8
2
1– 2
1 –2
4
2
2
0.5
0.5
2
0.5
2
1
4
2
2
LZDR, linezolid resistant; ND, not determined.
Triplicate determinations using conventional 1 log2 dilution progression unless specified otherwise (lower and higher values shown in case of
divergence).
b
From the ATCC collection (Manassas, VA, USA).
c
From JMI Laboratories (North Liberty, IA, USA); the first number is the strain number and the second is the bank number.
d
From K. Kosowska-Shick and P. C. Appelbaum (Hershey Medical Center, Hershey, PA, USA); VISA phenotype based on vancomycin MIC determination.
e
From the Network on Antimicrobial Resistance in S. aureus (managed by the Eurofins Global Central Laboratory, Chantilly, VA, USA; supported under
NIAID/NIH contract no. HHSN2722007 00055C); SCCmec group II.
f
Values in parentheses refer to MICs measured using arithmetic dilutions (0.25 mg/L intervals over the 1 –4 mg/L range).
g
From J. Quinn (Pfizer, Groton, CT, USA); cfr+ mechanism of resistance.
a
650
JAC
Ceftaroline and intracellular MRSA
Pharmacological descriptors of the activity of ceftaroline
and comparator antibiotics against extracellular and
intracellular forms of MRSA strain ATCC 33591
daptomycin (range 0.125–4 mg/L) or linezolid (range 0.5–8 mg/L).
Strains 35165, 48046, CM05, 062-13091 A and 062-13101 A
were retested using arithmetic dilutions (0.25 mg/L intervals
between 1 and 4 mg/L), but the values (reported in Table 1)
were always within the corresponding +1 log2 range of the
progression scale of the conventional assay method.
The MICs of ceftaroline for MRSA were lower (1 –2 log2 dilutions) when assayed at pH 5.5, especially for strains with a
higher MIC. Conversely, acid pH caused an increase in the MICs
of daptomycin, no or discordant changes for vancomycin and nonsystematic decreases for linezolid. Of note, strains 062-13091 A
and 062-13101 A, which have previously been reported to
display ceftaroline MICs of 4 mg/L, consistently showed an MIC
of 2 mg/L in our assays.
In this first series of experiments, 24 h concentration –responses
were examined for ceftaroline in comparison with vancomycin,
daptomycin and linezolid against both extracellular and intracellular forms of the reference MRSA strain ATCC 33591. Data are
presented graphically in Figure 1 with the corresponding
pharmacological descriptors and regression parameters shown
in Table 2. In all cases, a single sigmoidal function could be
fitted to the data, in accordance with the pharmacological
model previously described for the fully susceptible MSSA strain
ATCC 25923 and various antibiotics.15 In broth (Figure 1, left
panel), maximal or close to maximal effects (Emax) were
observed for all four antibiotics when their concentration
reached a value corresponding to their serum peak concentrations in patients (Cmax; total drug). Ceftaroline, vancomycin and
daptomycin were highly bactericidal, yielding calculated Emax
values corresponding to the actual limit of detection. In contrast,
linezolid did not achieve a mean 3 log10 cfu/mL decrease. As
expected for this bacteriostatic agent, linezolid was statistically
significantly inferior to the other antibiotics when evaluating
the rate of bacterial kill. Moving to the intracellular forms
(Figure 1, right panel), maximal relative activities (Emax) were
considerably lower (less negative) for all four antibiotics, as
As b-lactams in general, and ceftaroline in particular, are known to
be potentially unstable when exposed to 378C, we checked
for recovery of the antibiotic from broth and culture media after
24 h incubation using both microbiological and analytical
(LC-MS/MS) assays. Recovery was ≏66% using LC-MS/MS
determination, and .75% and ≏50% by the microbiological
method for water or broth and the cell culture medium, respectively.
Ceftaroline
Vancomycin
D Log10 cfu from time 0 (24 h)
(a) 4
Daptomycin
Linezolid
(b)
3
2
2
0
1
–2
0
–4
–6
Broth
–3
–1
THP–1
–2
–1
0
1
2
3
–3
–2
Log10 concentration (mg/L)
D Log10 cfu from time 0 (24 h)
Stability of ceftaroline
–1
0
1
2
3
Figure 1. Concentration-dependent activities of four antistaphylococcal antibiotics against extracellular [MHB broth pH 7.4 (a)] and intracellular
[THP-1 monocytes (b)] forms of S. aureus strain ATCC 33591 (MRSA). For these experiments, broths or infected cells were incubated for 24 h in the
presence of increasing concentrations of antibiotic (total drug; abscissa). The ordinates show the change in the number of cfu (log10) per mL of
medium (broth) or per mg or cell protein (THP-1). Note that because of the marked difference in the amplitude of the change between bacteria in
broth versus bacteria in THP-1 cells, the scale extends from 26 to 4 in panel (a) and from 21 to 3 in panel (b), with the broken horizontal line
showing the zero value (no apparent change from the initial, post-phagocytosis inoculum). All values are means+SD (n¼2 or 3; when not visible,
the SD bars are smaller than the size of the symbols). The lowest limit of detection corresponds to a cfu decrease of 5 log10 units compared with
the original inoculum. The grey zone shows the range of maximal serum concentrations observed in humans for the antibiotics (20– 57 mg/L
based on the following reported Cmax values: ceftaroline, 21 mg/L; vancomycin, 20– 50 mg/L; daptomycin, 57 mg/L; and linezolid, 15– 20 mg/L; see
footnote c in Table 2).
651
Mélard et al.
652
Table 2. Pharmacological descriptors, goodness of fit and statistical analysis of the concentration–response studies of the antibiotics against strain ATCC 33591 (MRSA) in broth and
in THP-1 monocytes (24 h incubation)
Pharmacological descriptor
EC50d
Condition/antibiotic
MH broth (extracellular
ceftaroline
vancomycin
daptomycin
linezolid
Emina
Emaxb
ECmaxc
bacteria)
3.28 A;a (1.91– 4.64)
2.92 A;a (2.08– 3.76)
3.17 A;a (2.27– 4.06)
3.10 A;a (2.40– 3.80)
25.37 A;a (26.50 to 24.23)
25.10 A;a (26.20 to 23.99)
25.09 A;a (25.94 to 24.25)
22.89 B;a (23.48 to 22.28)
25.04
24.55
24.72
22.34
TH P-1 monocytes (intracellular bacteria)
ceftaroline
2.60 A;a (1.92– 3.27)
vancomycin
2.43 A;a (1.70– 3.16)
daptomycin
2.28 A;b (1.76– 2.79)
linezolid
2.33 A;b (2.10– 2.55)
20.56 A;b (20.82 to 20.29)
20.65 A;b (21.08 to 20.21)
20.99 B;b (21.27 to 20.71)
20.32 A,C;b (20.47 to 20.17)
20.53
20.59
20.96
20.26
mg/L
0.82 A;a
2.62 B;a
2.70 B;a
1.74 A,B;a
(0.33 –2.06)
(1.43 –4.81)
(1.34 –5.42)
(0.92 –3.28)
0.16 A;b (0.08 –0.35)
0.67 B;b (0.23 –1.9)
0.61 B;b (0.32 –1.18)
0.42 B;b (0.27 –0.63)
Cse
×MICf
mg/L
×MICf
Goodness of
fit (R 2)
1.64 A;a (0.65 –4.12)
5.24 B;a (2.85 –9.61)
5.39 B;a (2.69 –10.84)
1.74 A;a (0.92 –3.28)
≏0.48
≏1.54
≏1.67
≏1.98
≏1.01
≏3.04
≏3.28
≏1.89
0.934
0.970
0.978
0.970
0.32 A;b (0.14 –0.71)
1.35 B;b (0.47 –3.88)
2.46 B;b (1.28 –4.70)
0.38 A;b (0.23 –0.61)
≏0.76
≏2.61
≏1.40
≏3.55
≏1.45
≏5.22
≏6.02
≏3.41
0.969
0.943
0.963
0.990
Data are from Figure 1.
Statistical analysis: comparison of regression parameters (Emax and EC50). Figures with different letters are significantly different (P≤0.05) from all others in the same group. Uppercase
letters: comparison between antibiotics (i) in broth (upper four rows) or (ii) in THP-1 monocytes (lower four rows) by analysis of variance (with Tukey –Kramer multiple comparisons test
if P,0.05) comparing the four values; lowercase letters: comparison between broth and THP-1 monocytes for the same antibiotic (unpaired two-tailed t-test comparing the two
values).
a
cfu increase (in log10 units) at 24 h from the corresponding initial inoculum as extrapolated for an infinitely low antibiotic concentration.
b
cfu decrease (in log10 units) at 24 h from the corresponding initial inoculum as extrapolated for an infinitely large antibiotic concentration.
c
cfu decrease (in log10 units) at 24 h from the corresponding initial inoculum as intrapolated (using the Hill equation) for a concentration of antibiotic corresponding to the maximal
serum concentration observed in humans receiving conventional therapy (Cmax). Values chosen for this table are: ceftaroline, 21 mg/L; vancomycin, 35 mg/L; daptomycin, 57 mg/L; and
linezolid, 17.5 mg/L (based on mean values in the corresponding US labelling for ceftaroline at its registered dosage of 600 mg every 12 h in the USA and the corresponding rationale
document from EUCAST for the most common registered dosages of the other antibiotics in Europe).
d
Concentration [in mg/L or ×MIC (total drug)] causing a reduction halfway between Emin and Emax, as obtained from the Hill equation (slope factor of 1).
e
Concentration (in mg/L or ×MIC [total drug]) resulting in no apparent bacterial growth, as determined by graphical interpolation.
f
Measured at pH 7.4 in broth (see Table 1; for linezolid, an MIC of 1 mg/L was used for calculations).
JAC
Ceftaroline and intracellular MRSA
D Log cfu from time 0 (24 h)
(a)
MIC = 2 mg/L (n = 4)
Broth
(b)
3
THP-1
(c)
3
3
2
2
1
1
2
0
0
–1
–1
1
–2
–2
–3
–3
0
–4
–5
Broth
–6
–4
–3
–2
–4
–5
THP-1
–1
0
1
2
3
–1
–4
D Log cfu from time 0 (24 h)
MIC = 0.5 mg/L (n = 3–4)
MIC = 1 mg/L (n = 2)
MIC = 0.125 (n = 1)
MIC = 0.25 mg/L (n = 1)
–6
–3
–2
–1
Log10 concentration (mg/L)
0
1
2
3
–4
–3
–2
–1
0
1
2
3
4
Log10 concentration (×MIC)
Figure 2. 24 h concentration-dependent activity of ceftaroline against S. aureus isolates with differing susceptibilities. Left (a) and middle (b) panels:
activity in broth and in THP-1 monocytes, respectively, as a function of drug weight concentration [mg/L (total drug)]: diamonds, MIC¼0.125 mg/L
(strain ATCC 25923; MSSA); squares, MIC¼0.25 mg/L (strain 34843; MSSA); triangles, MIC¼0.5 mg/L [strains ATCC 33591 (MRSA), 19210 (MRSA; not
tested in broth), SA 555 (MRSA/VISA) and SA 19834 (MRSA); all MRSA]; inverted triangles, MIC¼1 mg/L [strains NRS18 (MRSA/VISA) and 35165 (MRSA)];
circles, MIC¼2 mg/L [strains 48046 (MRSA), CM05 (MRSA/LZDR), 062-13101 A (MRSA) and 062-13091 A (MRSA)]; see Table 1 for more details. Right
panel (c): activity in broth (circles) and in THP-1 monocytes (squares) as a function of multiples of MIC (total drug). The ordinates of all graphs show
the change in cfu (log10) per mL of medium (broth) or per mg of cell protein (THP-1) at 24 h compared with the original post-phagocytosis inoculum
(horizontal broken line). Note that because of the marked difference in the amplitude of the change between bacteria in broth versus bacteria in THP-1
cells, the scale extends from 26 to 3 in panel (a) and from 21 to 3 for THP-1 cells in panel (b). All values are means+SD (with each strain tested in
duplicate); when not visible, the SD bars are smaller than the size of the symbols). The lowest limit of detection corresponds to a cfu decrease of
5 log10 units compared with the original inoculum. The vertical continuous line in the left and middle panels indicates the maximal serum
concentration of ceftaroline commonly observed in humans (Cmax).
these achieved only a ≏0.5 to ≏1 log cfu decrease compared
with the original inoculum. Statistically significant but quite
small differences of intracellular Emax were observed, with daptomycin being more active than ceftaroline and vancomycin, and
linezolid being the least active. Considering the relative potencies
(EC50) and the static concentrations (Cs), ceftaroline appeared to
be the most potent, whether in broth or in cells, with values systematically about 2- to 4-fold less than for the other antibiotics,
whether expressed as weight concentration (mg/L) or, except for
linezolid, as multiples of the MIC in broth (pH 7.4). Interestingly
enough, all values of EC50 and Cs were quite similar or lower
for intracellular bacteria compared with bacteria in broth, denoting an unimpaired potency in the intracellular milieu. The numerical values of these parameters were also close to the MICs of
the corresponding antibiotics.
Extracellular and intracellular activity of ceftaroline
against S. aureus isolates with differing susceptibilities
In these experiments we compared the 24 h concentration–
responses of a series of strains of S. aureus with ceftaroline MICs
ranging from 0.125 to 2 mg/L (strains with higher MICs could
not be identified). The results are presented graphically in
Figure 2 with changes in cfu/mL shown as a function of the
drug weight concentration for bacteria in broth (left panel) and
in THP-1 monocytes (middle panel), and as a function of multiples
of MIC for both (right panel). Considering first cfu/mL changes as a
function of weight concentrations (mg/L), there was a gradual
shift of the curves to higher concentrations as a function of the
MIC for the strains, resulting in increases in the EC50 and Cs parameters. This was accompanied by a small (but variable among
strains) decrease in the activity (less-negative values) observed
at a concentration corresponding to the drug Cmax (ECmax) or of
the maximal relative activities (Emax). These changes are illustrated in Figure S1 (available as Supplementary data at JAC
Online). The data were then used for recursive partitioning analysis
of each descriptor on the basis of the MIC (broth; pH 7.4; log2
dilution) of the strains, with the results shown in Table 3 (and
individual graphs shown in Figure S2, available as Supplementary
data at JAC Online). The method used (single-pass decision tree)
yielded a dichotomous split at an MIC of 1 mg/L (split between
,1 and ≥1) for all four descriptors, but yielded statistically significant differences for the EC50 and Cs parameters with bacteria in
broth only (differences were at the limit of statistical significance
for ECmax with bacteria in THP-1 cells). Using MICs determined
by arithmetic dilutions (0.25 mg/L intervals) for strains with MICs
≥1 mg/L (see Table 1) did not significantly change the results of
the analysis and its conclusions.
Lastly, when changes in cfu/mL data were plotted as a function of multiples of the MIC of the corresponding strains, data for
all strains could be analysed as single functions for bacteria in
broth and bacteria in THP-1 cells, respectively, as presented
graphically in the right panel of Figure 2 with the corresponding
653
Mélard et al.
Table 3. Recursive partitioning analysis of the values of the pharmacological descriptors of the concentration-dependent responses of bacterial
strains with increasing MICs of ceftaroline in broth or in THP-1 cells as a function of the corresponding MIC (pH 7.4)
Optimal candidate split
value [MIC (mg/L)]a
Parameter values
(below/above MIC split value)
LogWorthb/P valuec
1. Broth (n ¼11)
Emax (Dlog10 cfu)d
ECmax (Dlog10 cfu)e
EC50 (mg/L)f
Cs (mg/L)g
,1/≥1
,1/≥1
,1/≥1
,1/≥1
25.24+0.10/24.89+0.88
25.04+0.12/24.14+0.79
0.54+0.16/2.41+0.50
0.28+0.12/1.24+0.40
0.09/0.81
0.97/0.11
8.05/,0.01*
3.56/,0.01*
2. THP-1 (n¼12)
Emax (Dlog10 cfu)
ECmax (Dlog10 cfu)
EC50 (mg/L)
Cs (mg/L)
,1/≥1
,1/≥1
,1/≥1
,1/≥1
20.68+0.14/20.55+0.22
20.62+0.16/20.36+0.15
0.38+0.21/1.44+1.11
1.83+1.55/6.51+4.35
0.19/0.64
1.35/0.04*
0.85/0.14
1.00/0.10
Descriptor
Analysis was made on the basis of the data shown in Figure 2 (left and middle panels), but considering the individual values of each strain.
Asterisks indicate results considered statistically significant on the basis of the P value.
a
Values of MIC separating datasets in two categories based on minimization of the sum of squared errors across the whole data as a function of the
MIC (further splitting was unsuccessful because of the limited number of independent values).
b
Node splitting is based on the LogWorth statistic (values .2 indicate that the variable used in the branch is significant and should be included in the
decision tree.
c
Calculated based from LogWorth value [P = 10(−LogWorth) ].
d
cfu decrease (in log10 units) at 24 h from the corresponding initial inoculum, as extrapolated from infinitely large concentrations of antibiotics.
e
cfu decrease (in log10 units) at 24 h from the corresponding initial inoculum, as intrapolated (using the Hill equation) for a concentration of antibiotic
corresponding to the maximal serum concentration observed in humans receiving conventional therapy (Cmax).
f
Concentration (total drug) causing a reduction halfway between Emin and Emax, as obtained from the Hill equation (slope factor of 1).
g
Concentration (total drug) resulting in no apparent bacterial growth, as determined by graphical interpolation.
Table 4. Pharmacological descriptors, goodness of fit and statistical analysis of the concentration– response studies of ceftaroline against strains
with increasing MICs (from 0.125 to 2 mg/L; see list and MICs in the caption of Figure 2) in broth and in THP-1 monocytes (24 h incubation)
Pharmacological descriptor
Condition
Emaxa
MH broth (extracellular bacteria), n¼11
25.09 a (25.34 to 24.84)
THP-1 monocytes (intracellular bacteria), n¼12
20.58 b (20.74 to 20.43)
ECmaxb
EC50 (×MIC)c,d
Cs (×MIC)d,e
Goodness of fit (R 2)
24.60
1.44 a (1.14– 1.83)
0.71
0.938
20.46
0.81 a (0.57– 1.16)
3.62
0.855
Data shown are from Figure 2 (right panel).
Statistical analysis: comparison of parameters (Emax and EC50) between broth and THP-1 monocytes. Figures with different letters are significantly
different (P≤0.05) from each other (unpaired t-test two-tailed).
a
cfu decrease (in log10 units) at 24 h from the corresponding initial inoculum as extrapolated from infinitely large concentrations of antibiotics.
b
cfu decrease (in log10 units) at 24 h from the corresponding initial inoculum as interpolated for an antibiotic concentration (total drug) corresponding
to the reported human Cmax of ceftaroline (21 mg/L).
c
Concentration [multiples of MIC (total drug)] causing a reduction halfway between Emin and Emax, as obtained from the Hill equation (slope factor of 1).
d
Concentration (multiple of MIC [total drug]) resulting in no apparent bacterial growth, as determined by graphical interpolation.
e
MIC determined in broth at pH 7.4.
pharmacological descriptors shown in Table 4. While this further
confirmed the major differences in efficacy (Emax and ECmax) of
ceftaroline against bacteria in broth versus bacteria in THP-1
monocytes, the corresponding relative potencies (EC50) were
654
not markedly different from each other. The static concentration
(Cs) value was close to 1× the MIC for bacteria in broth, as anticipated, and was only about 4-fold higher for bacteria in THP-1
monocytes.
Ceftaroline and intracellular MRSA
Cellular accumulation of ceftaroline
The accumulation of ceftaroline was first measured after 24 h incubation at 20 mg/L in both uninfected and infected (strain ATCC
33591) cells using an LC-MS/MS assay. The apparent cellular to
extracellular concentration ratio was 0.66+0.05 (n¼ 6) and
0.90+0.36 (n¼ 3) in uninfected and infected cells, respectively.
Discussion
There is a clear need to discover and develop novel antibiotics to
meet the increased resistance of bacteria to currently registered
drugs.25,26 Although the most blatant lack of progress is for
anti-Gram-negative agents, the situation with Gram-positive
organisms remains of concern since several new, potentially
promising compounds were not approved by the regulatory
authorities,27 – 29 are facing toxicity and emergence of resistance
issues30 – 33 or may require higher doses than originally
foreseen.34 In this context, ceftaroline fosamil may represent a
useful alternative, especially in light of the decreased susceptibility of contemporary isolates to vancomycin (including heteroresistance),35,36 and the emergence of both chromosomal and
transferable resistance to linezolid.37,38
Ceftaroline in vitro activity against MRSA is related to its high
affinity for PBP2a,39,40 an affinity that is enhanced in the presence of a cell wall structural surrogate.41 In the present study
we confirm the unimpaired in vitro activity of ceftaroline
against MRSA with non-susceptibility or resistance mechanisms
to vancomycin and linezolid, as previously reported by
others.4,42 In this context, the present study adds important information about the intracellular activity of ceftaroline in comparison with vancomycin, linezolid and daptomycin. We first
show that ceftaroline compares in almost every respect to
these antibiotics for a susceptible reference strain (ATCC
33591), but that their relative efficacy is considerably reduced
compared with what is observed in broth. This is based on a
comprehensive pharmacodynamic analysis comparing static
concentrations, relative potencies, effects at concentrations corresponding to the Cmax in humans and maximal effects. Of note,
daptomycin, reported to be highly bactericidal in vitro,43 did not
prove superior to ceftaroline in THP-1 monocytes or in broth.
Thus, for all four agents tested here, it clearly appears that intracellular bacteria are protected to some degree against the antibacterial activities of these different classes of antibiotics (a
similar observation was made for ceftobiprole, another cephalosporin with activity against MRSA).18 This cannot be due to a lack
of bacterial growth in cells (as Emin values show that there is an
≏100-fold growth of bacteria in cells in the absence of antibiotic)
and is not related to a loss of potency (the EC50 and Cs parameters being essentially similar for bacteria in broth and in
THP-1 monocytes). Thus, ceftaroline as well as vancomycin and
daptomycin seem to become essentially bacteriostatic against
intracellular bacteria and compare, in this respect, to linezolid.
This is in contrast to lipoglycopeptides (telavancin and oritavancin), fluoroquinolones or quinupristin/dalfopristin, which show
maximal relative efficacy (Emax) values between 22 and
23 log10 cfu/mL in the same model44 – 48 and may therefore be
considered as exerting a near-to-bactericidal intracellular activity
based on commonly accepted criteria of cidality.19
JAC
In our study we also examined whether a decrease in the susceptibility of S. aureus to ceftaroline (as measured by its MIC in
broth) would be accompanied by a decrease or loss of intracellular activity. This approach was triggered by a previous successful
attempt to define an intracellular breakpoint for moxifloxacin in
the same model.47 With ceftaroline, we see that the intracellular
activity of antibiotics is primarily driven by the MIC for the phagocytosed organism, confirming previous observations made with
other antibiotics.18,48 – 50 However, the changes in the values of
the pharmacodynamic descriptors of ceftaroline over the range
of MICs investigated (0.125– 2 mg/L) were small. Thus, although
the recursive partitioning analysis suggested a breakpoint at an
MIC value of 1 mg/L, this was only significant for bacteria
grown in broth and for parameters related to potency (EC50
and Cs), while efficacy parameters (ECmax and Emax) remained
unaffected. This may be due to the too narrow MIC range investigated (4 log2 dilutions only) or may simply indicate that
changes in efficacy parameters will only become visible at higher
MICs. However, strains displaying increased ceftaroline MICs
seem very difficult to raise51 and, when observed, may show
only limited changes52 as those we used here (mecAindependent high-level ceftaroline-resistant mutants have been
recently generated by serial passage in vitro,53 but these were
not available to us during our study). The fact that the parameters of intracellular activity are even less affected by an increase in MIC may also be related to the fact that the MIC of
ceftaroline is lowered by about 1 –2 log2 dilutions when tested
at pH 5.5 (to mimic the phagolysosomal environment),20,21,44
especially for strains against which ceftaroline is less active
[thus further narrowing the MIC range (3 log2 dilutions only)].
Interestingly enough, this increased activity at acid pH may
partly compensate for the weak cellular accumulation of ceftaroline, and account for its effect on intracellular bacteria as previously observed for other b-lactams.54 This is also consistent with
our observation that acidic pH causes conformational changes of
PBP2a that improve its acylation by b-lactams within a time
frame relevant to the growth rate of MRSA.55 Thus, the present
data suggest that ceftaroline activity against intracellular
S. aureus may remain unimpaired for organisms with an MIC of
2 mg/L, even though it becomes adversely affected for extracellular bacteria when the MIC exceeds 1 mg/L.
The present study also has several limitations that need to be
underlined. First, assessment of intracellular activity was made
with cells (THP-1 monocytes) that, in contrast to fully differentiated macrophages, do not quickly kill phagocytosed S. aureus.
This was by design, as we aimed to analyse the pharmacodynamic properties of ceftaroline without undue interference
from host defence mechanisms. Future studies may need to
examine how and to what extent ceftaroline cooperates with
these mechanisms. Other cell types also capable of harbouring
S. aureus, such as keratinocytes or endothelial cells, could also
be used. Second, we did not examine the effect of time on the
response to ceftaroline (all experiments used a fixed 24 h timepoint), which, again, could be the subject of future studies
similar to those made recently with extracellular bacteria.56
However, we know from previous studies15 that killing of intracellular S. aureus by b-lactams is a slow process. Thus, shorter
exposure time would yield only minimal changes in cfu that
prove non-significant. Third, we did not monitor the expression
of Panton-Valentine leucocidin toxin (PVL), which could affect
655
Mélard et al.
the host cell viability. However: (i) our previous studies failed to
find evidence of an impact of the presence of the PVL-encoding
genes on the intracellular behaviour and antibiotic susceptibility
of S. aureus in the model used;47 (ii) the production of PVL and
other toxins is maximal at the stationary stage,57 which is not
reached for intracellular bacteria under the conditions of our experiment; and (iii) PVL presence was not a primary determinant
of outcome in patients with complicated skin and skin structure
infections due to either MRSA or MSSA in the clinical studies
assessing the efficacy of ceftaroline.58 Lastly, all comparisons
were made using total drug concentrations and using nominal
ones. We know that only free concentrations are usually considered for in vivo activity assessment and for clinical breakpoint
setting.59 However, our culture medium contains only 10%
serum, which means that most drugs will be free, as previously
documented for b-lactams with high protein-binding.16 But this
will not influence much the behaviour of ceftaroline since it
has a low protein binding (≏20%) in 100% serum.1 Conversely,
our model may lead to an overestimation of daptomycin activity,
as it is impaired in human serum in comparison with broth.60
Loss of activity during incubation was also not taken into
account because it is likely to be progressive and did not, over
the 24 h duration of our experiments, exceed 50%, which is
less than the 2-fold MIC change that is considered significant
in conventional susceptibility testing.
In conclusion, we present in vitro evidence that ceftaroline is
capable of controlling the growth of intracellular S. aureus to an
extent similar to that of vancomycin, daptomycin and linezolid,
irrespective of the presence of resistance mechanisms to conventional b-lactams (methicillin resistance), vancomycin (VISA)
or linezolid (cfr), and for strains for which ceftaroline shows an
MIC ≤2 mg/L. These results may now trigger the performance
of in vivo animal and human studies aimed at better delineating
the potential use of ceftaroline in difficult-to-treat infections
where the persistence of an intracellular inoculum may be a critical determinant.12
Acknowledgements
We thank K. Kosowska-Shick, P. C. Appelbaum, J. Quinn, JMI Laboratories
and the Network on Antimicrobial Resistance in S. aureus (NARSA) for the
gift of bacterial strains. We are grateful to M.-C. Cambier for dedicated
technical assistance.
Funding
This work was supported by Belgian Fonds de la Recherche Scientifique
Médicale (FRSM; grant nos. 3.4.597.06 and 3.4530.12), the Belgian
Fonds de la Recherche Scientifique (FRS-FNRS; grant no. 1.5118.11) and
a grant-in-aid from Cerexa, Inc., a wholly owned subsidiary of Forest
Laboratories, Inc. Editorial assistance was funded by Forest Research
Institute, Inc.
L. G. G. is Boursière of the Belgian Fonds pour la Formation à la Recherche dans l’Industrie et dans l’Agriculture (FRIA). D. D. is the recipient of a
post-doctoral fellowship of the Belgian Fonds de la Recherche Scientifique
(FRS-FNRS; grant no. 1.5118.11). S. L. and F. V. B. are Chargé de Recherche
and Maı̂tre de Recherche of the FRS-FNRS, respectively.
656
Transparency declarations
No conflicts of interest to declare.
Cerexa, Inc. was involved in the design and decision to present these
results, but had no involvement in the collection, analysis or interpretation of data.
Editorial assistance was provided by Scientific Therapeutics Information, Inc.
Supplementary data
Figures S1 and S2 are available as Supplementary data at JAC Online
(http://jac.oxfordjournals.org/).
References
1 Biek D, Critchley IA, Riccobene TA et al. Ceftaroline fosamil: a novel
broad-spectrum cephalosporin with expanded anti-Gram-positive
activity. J Antimicrob Chemother 2010; 65 Suppl 4: iv9– 16.
2 Iizawa Y, Nagai J, Ishikawa T et al. In vitro antimicrobial activity of
T-91825, a novel anti-MRSA cephalosporin, and in vivo anti-MRSA
activity of its prodrug, TAK-599. J Infect Chemother 2004; 10: 146–56.
3 Flamm RK, Sader HS, Farrell DJ et al. Summary of ceftaroline activity
against pathogens in the United States, 2010: Report from the
Assessing Worldwide Antimicrobial Resistance Evaluation (AWARE)
Surveillance Program. Antimicrob Agents Chemother 2012; 56: 2933 –40.
4 Zhanel GG, Rossnagel E, Nichol K et al. Ceftaroline pharmacodynamic
activity versus community-associated and healthcare-associated
methicillin-resistant Staphylococcus aureus, heteroresistant vancomycinintermediate S. aureus, vancomycin-intermediate S. aureus and
vancomycin-resistant S. aureus using an in vitro model. J Antimicrob
Chemother 2011; 66: 1301–5.
5 Ishikawa T, Matsunaga N, Tawada H et al. TAK-599, a novel
N-phosphono type prodrug of anti-MRSA cephalosporin T-91825:
synthesis, physicochemical and pharmacological properties. Bioorg Med
Chem 2003; 11: 2427–37.
6 Corrado ML. Integrated safety summary of CANVAS 1 and 2 trials:
Phase III, randomized, double-blind studies evaluating ceftaroline
fosamil for the treatment of patients with complicated skin and skin
structure infections. J Antimicrob Chemother 2010; 65 Suppl 4: iv67–71.
7 Wilcox MH, Corey GR, Talbot GH et al. CANVAS 2: the second Phase III,
randomized, double-blind study evaluating ceftaroline fosamil for the
treatment of patients with complicated skin and skin structure
infections. J Antimicrob Chemother 2010; 65 Suppl 4: iv53–65.
8 Corey GR, Wilcox MH, Talbot GH et al. CANVAS 1: the first Phase III,
randomized, double-blind study evaluating ceftaroline fosamil for the
treatment of patients with complicated skin and skin structure
infections. J Antimicrob Chemother 2010; 65 Suppl 4: iv41–51.
9 Anonymous. TEFLARO Prescribing Information. Forest Pharmaceuticals,
Inc., subsidiary of Forest Laboratories, Inc., St Louis, MO, USA. http://www.
frx.com/pi/Teflaro_pi.pdf (11 October 2012, date last accessed).
9a Anonymous. ZINFORO Summary of Product Characteristics. European
Medicines Agency, London, UK. http://www.ema.europa.eu/ (26 October
2012, date last accessed).
10 Friedland HD, O’Neal T, Biek D et al. CANVAS 1 and 2: analysis of
clinical response at day 3 in two Phase 3 trials of ceftaroline fosamil
versus vancomycin plus aztreonam in treatment of acute bacterial skin
and skin structure infections. Antimicrob Agents Chemother 2012; 56:
2231– 6.
Ceftaroline and intracellular MRSA
11 Lowy FD. Is Staphylococcus aureus an intracellular pathogen? Trends
Microbiol 2000; 8: 341– 3.
12 Garzoni C, Kelley WL. Staphylococcus aureus: new evidence for
intracellular persistence. Trends Microbiol 2009; 17: 59–65.
13 Van Bambeke F, Barcia-Macay M, Lemaire S et al. Cellular
pharmacodynamics and pharmacokinetics of antibiotics: current views
and perspectives. Curr Opin Drug Discov Devel 2006; 9: 218–30.
14 Sandberg A, Lemaire S, Van Bambeke F et al. Intra- and extracellular
activities of dicloxacillin and linezolid against a clinical Staphylococcus
aureus strain with a small-colony-variant phenotype in an in vitro
model of THP-1 macrophages and an in vivo mouse peritonitis model.
Antimicrob Agents Chemother 2011; 55: 1443–52.
15 Barcia-Macay M, Seral C, Mingeot-Leclercq MP et al. Pharmacodynamic evaluation of the intracellular activities of antibiotics against
Staphylococcus aureus in a model of THP-1 macrophages. Antimicrob
Agents Chemother 2006; 50: 841– 51.
16 Lemaire S, Van Bambeke F, Mingeot-Leclercq MP et al. Activity of three
b-lactams (ertapenem, meropenem and ampicillin) against intraphagocytic
Listeria monocytogenes and Staphylococcus aureus. J Antimicrob Chemother
2005; 55: 897–904.
17 Page MG. Emerging cephalosporins. Expert Opin Emerg Drugs 2007;
12: 511–24.
18 Lemaire S, Glupczynski Y, Duval V et al. Activities of ceftobiprole and
other cephalosporins against extracellular and intracellular (THP-1
macrophages and keratinocytes) forms of methicillin-susceptible and
methicillin-resistant Staphylococcus aureus. Antimicrob Agents
Chemother 2009; 53: 2289 –97.
19 Clinical and Laboratory Standards Institute. Performance Standards
for Antimicrobial Susceptibility Testing: Twentieth Informational
Supplement M100-S20. CLSI, Wayne, PA, USA, 2010.
20 Ohkuma S, Poole B. Fluorescence probe measurement of the
intralysosomal pH in living cells and the perturbation of pH by various
agents. Proc Natl Acad Sci USA 1978; 75: 3327– 31.
21 Lemaire S, Van Bambeke F, Mingeot-Leclercq MP et al. Role of acidic
pH in the susceptibility of intraphagocytic methicillin-resistant
Staphylococcus aureus strains to meropenem and cloxacillin. Antimicrob
Agents Chemother 2007; 51: 1627 –32.
22 Tsuchiya S, Yamabe M, Yamaguchi Y et al. Establishment and
characterization of a human acute monocytic leukemia cell line
(THP-1). Int J Cancer 1980; 26: 171– 6.
23 Carryn S, Van d V, Van Bambeke F et al. Impairment of growth of
Listeria monocytogenes in THP-1 macrophages by granulocyte
macrophage colony-stimulating factor: release of tumor necrosis
factor-a and nitric oxide. J Infect Dis 2004; 189: 2101 –9.
24 Lowry OH, Rosebrough NJ, Farr AL et al. Protein measurement with
the Folin phenol reagent. J Biol Chem 1951; 193: 265–75.
25 Livermore DM. Has the era of untreatable infections arrived?
J Antimicrob Chemother 2009; 64 Suppl 1: i29–36.
26 Bush K, Courvalin P, Dantas G et al. Tackling antibiotic resistance. Nat
Rev Microbiol 2011; 9: 894– 6.
27 Arias CA, Mendes RE, Stilwell MG et al. Unmet needs and prospects for
oritavancin in the management of vancomycin-resistant enterococcal
infections. Clin Infect Dis 2012; 54 Suppl 3: S233–8.
28 Sincak CA, Schmidt JM. Iclaprim, a novel diaminopyrimidine for the
treatment of resistant gram-positive infections. Ann Pharmacother
2009; 43: 1107– 14.
29 Bush K, Heep M, Macielag MJ et al. Anti-MRSA b-lactams in
development, with a focus on ceftobiprole: the first anti-MRSA
b-lactam to demonstrate clinical efficacy. Expert Opin Investig Drugs
2007; 16: 419–29.
JAC
30 Rubinstein E, Prokocimer P, Talbot GH. Safety and tolerability of
quinupristin/dalfopristin: administration guidelines. J Antimicrob Chemother
1999; 44 Suppl A: 37–46.
31 Falagas ME, Karageorgopoulos DE, Dimopoulos G. Clinical significance
of the pharmacokinetic and pharmacodynamic characteristics of
tigecycline. Curr Drug Metab 2009; 10: 13 –21.
32 Di Paolo A, Malacarne P, Guidotti E et al. Pharmacological issues of
linezolid: an updated critical review. Clin Pharmacokinet 2010; 49:
439–47.
33 Smith WJ, Drew RH. Telavancin: a new lipoglycopeptide for grampositive infections. Drugs Today (Barc) 2009; 45: 159– 73.
34 Seaton RA. Daptomycin: rationale and role in the management of skin
and soft tissue infections. J Antimicrob Chemother 2008; 62 Suppl 3:
iii15 –23.
35 Sakoulas G, Moise-Broder PA, Schentag J et al. Relationship of MIC and
bactericidal activity to efficacy of vancomycin for treatment of
methicillin-resistant Staphylococcus aureus bacteremia. J Clin Microbiol
2004; 42: 2398– 402.
36 Haque NZ, Zuniga LC, Peyrani P et al. Relationship of vancomycin
minimum inhibitory concentration to mortality in patients with methicillinresistant Staphylococcus aureus hospital-acquired, ventilator-associated, or
health-care-associated pneumonia. Chest 2010; 138: 1356–62.
37 Ikeda-Dantsuji Y, Hanaki H, Sakai F et al. Linezolid-resistant
Staphylococcus aureus isolated from 2006 through 2008 at six
hospitals in Japan. J Infect Chemother 2011; 17: 45–51.
38 Witte W, Cuny C. Emergence and spread of cfr-mediated
multiresistance in staphylococci: an interdisciplinary challenge. Future
Microbiol 2011; 6: 925– 31.
39 Moisan H, Pruneau M, Malouin F. Binding of ceftaroline to penicillinbinding proteins of Staphylococcus aureus and Streptococcus
pneumoniae. J Antimicrob Chemother 2010; 65: 713–6.
40 Kosowska-Shick K, McGhee PL, Appelbaum PC. Affinity of ceftaroline
and other b-lactams for penicillin-binding proteins from Staphylococcus
aureus and Streptococcus pneumoniae. Antimicrob Agents Chemother
2010; 54: 1670– 7.
41 Villegas-Estrada A, Lee M, Hesek D et al. Co-opting the cell wall in
fighting methicillin-resistant Staphylococcus aureus: potent inhibition of
PBP 2a by two anti-MRSA b-lactam antibiotics. J Am Chem Soc 2008;
130: 9212– 3.
42 Vidaillac C, Leonard SN, Rybak MJ. In vitro activity of ceftaroline
against methicillin-resistant Staphylococcus aureus and heterogeneous
vancomycin-intermediate S. aureus in a hollow fiber model. Antimicrob
Agents Chemother 2009; 53: 4712 –7.
43 Stratton CW, Liu C, Weeks LS. Activity of LY146032 compared with
that of methicillin, cefazolin, cefamandole, cefuroxime, ciprofloxacin,
and vancomycin against staphylococci as determined by kill-kinetic
studies. Antimicrob Agents Chemother 1987; 31: 1210– 5.
44 Seral C, Van Bambeke F, Tulkens PM. Quantitative analysis of
gentamicin, azithromycin, telithromycin, ciprofloxacin, moxifloxacin, and
oritavancin (LY333328) activities against intracellular Staphylococcus
aureus in mouse J774 macrophages. Antimicrob Agents Chemother
2003; 47: 2283– 92.
45 Barcia-Macay M, Lemaire S, Mingeot-Leclercq MP et al. Evaluation
of the extracellular and intracellular activities (human THP-1
macrophages) of telavancin versus vancomycin against methicillinsusceptible,
methicillin-resistant,
vancomycin-intermediate
and
vancomycin-resistant Staphylococcus aureus. J Antimicrob Chemother
2006; 58: 1177– 84.
46 Baudoux P, Lemaire S, Denis O et al. Activity of quinupristin/
dalfopristin against extracellular and intracellular Staphylococcus aureus
657
Mélard et al.
with various resistance phenotypes. J Antimicrob Chemother 2010; 65:
1228– 36.
47 Lemaire S, Kosowska-Shick K, Appelbaum PC et al. Activity of
moxifloxacin against intracellular community-acquired methicillinresistant Staphylococcus aureus: comparison with clindamycin, linezolid
and co-trimoxazole and attempt at defining an intracellular
susceptibility breakpoint. J Antimicrob Chemother 2011; 66: 596–607.
48 Vallet CM, Marquez B, Ngabirano E et al. Cellular accumulation of
fluoroquinolones is not predictive of their intracellular activity: studies
with gemifloxacin, moxifloxacin and ciprofloxacin in a pharmacokinetic/
pharmacodynamic model of uninfected and infected macrophages. Int
J Antimicrob Agents 2011; 38: 249– 56.
49 Lemaire S, Van Bambeke F, Tulkens PM. Cellular accumulation and
pharmacodynamic evaluation of the intracellular activity of CEM-101, a
novel fluoroketolide, against Staphylococcus aureus, Listeria monocytogenes, and Legionella pneumophila in human THP-1 macrophages.
Antimicrob Agents Chemother 2009; 53: 3734–43.
50 Lemaire S, Van Bambeke F, Appelbaum PC et al. Cellular
pharmacokinetics and intracellular activity of torezolid (TR-700): studies
with human macrophage (THP-1) and endothelial (HUVEC) cell lines.
J Antimicrob Chemother 2009; 64: 1035– 43.
51 Mushtaq S, Warner M, Ge Y et al. In vitro activity of ceftaroline (PPI0903M, T-91825) against bacteria with defined resistance mechanisms
and phenotypes. J Antimicrob Chemother 2007; 60: 300– 11.
52 Clark C, McGhee P, Appelbaum PC et al. Multistep resistance
development studies of ceftaroline in gram-positive and -negative
bacteria. Antimicrob Agents Chemother 2011; 55: 2344– 51.
53 Chan L, Chambers H. MecA-independent high-level b-lactam
resistance in Staphylococcus aureus strains passaged in ceftobiprole
and ceftaroline. In: Abstracts of the Fifty-second Interscience Conference
658
on Antimicrobial Agents and Chemotherapy, San Francisco, CA, USA,
2012. Abstract C1-1351a, p. 300. American Society for Microbiology,
Washington, DC, USA.
54 Baudoux P, Bles N, Lemaire S et al. Combined effect of pH and
concentration on the activities of gentamicin and oxacillin against
Staphylococcus aureus in pharmacodynamic models of extracellular
and intracellular infections. J Antimicrob Chemother 2007; 59: 246–53.
55 Lemaire S, Fuda C, Van Bambeke F et al. Restoration of susceptibility of
methicillin-resistant Staphylococcus aureus to b-lactam antibiotics by
acidic pH: role of penicillin-binding protein PBP 2a. J Biol Chem 2008;
283: 12769–76.
56 Marconescu P, Graviss EA, Musher DM. Rates of killing of methicillinresistant Staphylococcus aureus by ceftaroline, daptomycin, and
telavancin compared to that of vancomycin. Scand J Infect Dis 2012;
44: 620–2.
57 Graves SF, Kobayashi SD, Braughton KR et al. Relative contribution of
Panton-Valentine leukocidin to PMN plasma membrane permeability and
lysis caused by USA300 and USA400 culture supernatants. Microbes
Infect 2010; 12: 446–56.
58 Tong A, Tong SY, Zhang Y et al. Panton-Valentine leukocidin is not the
primary determinant of outcome for Staphylococcus aureus skin
infections: evaluation from the CANVAS studies. PLoS One 2012; 7:
e37212.
59 Mouton JW, Brown DF, Apfalter P et al. The role of pharmacokinetics/
pharmacodynamics in setting clinical MIC breakpoints: the EUCAST
approach. Clin Microbiol Infect 2012; 18: E37–45.
60 Stratton CW, Weeks LS. Effect of human serum on the bactericidal
activity of daptomycin and vancomycin against staphylococcal and
enterococcal isolates as determined by time-kill kinetic studies. Diagn
Microbiol Infect Dis 1990; 13: 245–52.
Activity of ceftaroline against extracellular (broth) and intracellular (THP-1 monocytes) forms of methicillinresistant Staphylococcus aureus: comparison with vancomycin, linezolid and daptomycin
Mélard et al - J Antimicrob Chemother 2013; 68: 648–658
Supplementary data
Figure S1
broth
THP-1
efficacy
potency
4
-1
3
EC 50 (mg/L)
Emax ( log10 cfu)
0
-2
-3
-4
2
1
-5
-6
0
0.125
0.25
0.5
1
2
0.125
0.25
0.5
1
2
1
2
MIC (mgL)
0
15.0
-1
12.5
-2
10.0
Cs (mg/L)
ECmax ( log10 cfu)
MIC (mgL)
-3
-4
-5
7.5
5.0
2.5
-6
0.0
0.125
0.25
0.5
MIC (mgL)
1
2
0.125
0.25
0.5
MIC (mgL)
Figure S1: Pharmacological descriptors of the concentration-dependent responses of Staphylococcus
aureus strains with increasing MICs to ceftaroline. Circles: bacteria in broth (MHB pH 7.4: 11 strains);
squares: bacteria in THP-1 monocytes (12 strains). Data are from the experiments illustrated in Figure 2
(24 h incubation). Emax (upper left): change in cfu (in log10 units) from the original inoculum for an infinitely
large antibiotic concentration; ECmax (lower left): change in cfu (in log10 units) for a ceftaroline concentration
corresponding to its maximal serum concentration in patients receiving standard therapy (21 mg/L); EC50
(upper right): concentration (in mg/L) yielding a change in cfu half way between Emin (change in cfu from the
original inoculum for an infinitely low antibiotic concentration) and Emax; Cs (lower right): concentration (in
mg/L) resulting in an apparent static effect (no change from the original inoculum). Emax and EC50 are the
parameters of the sigmoidal function (Hill equation; slope factor = 1) fitted to the data (non-linear
regression); ECmax and Cs are determined by graphical intrapolation using the corresponding Hill equations
(each individual point corresponds to the value observed for one strain). Curves are "best fitting" with no
specific underlying model.
Activity of ceftaroline against extracellular (broth) and intracellular (THP-1 monocytes) forms of methicillinresistant Staphylococcus aureus: comparison with vancomycin, linezolid and daptomycin
Mélard et al - J Antimicrob Chemother 2013; 68: 648–658
Figure S2
Figure S2: Graphical representation of the results of the recursive partitioning analysis of the
pharmacological descriptors derived from the concentration-dependent responses experiments using
bacterial strains with increasing MIC to ceftaroline in broth (left) or in THP-1 cells (right). The ordinate of
each graph shows the values of the corresponding pharmacological descriptor (Emax [upper row]: cfu
decrease [in log10 units] at 24 h from the corresponding initial inoculum, as extrapolated from infinitely large
concentrations of antibiotic; ECmax [2d row]: cfu decrease [in log10 units] at 24 h from the corresponding initial
inoculum, as intrapolated (using the Hill equation) for a concentration of antibiotic corresponding to the
maximal serum concentration observed in humans receiving conventional therapy (Cmax [21 mg/L total
drug]); EC50 [3d row]: concentration [in mg/L; total drug]) causing a reduction halfway between Emin (cfu
change at 24 h [in log10 units] from the corresponding initial inoculum as exatrapolated for an infinitely low
antibiotic concentration) and Emax; Cs [lower row]: concentration [in mg/L; total drug] resulting in no apparent
bacterial growth, as determined by graphical interpolation (using the Hill equation). The abscissa separates
the data in two groups according to the MIC values (broth; pH 7.4) of the corresponding strains as falling
below or above the best split value (1 mg/L). The horizontal bars show the mean of the values for each
group. Data points are coloured (from green to black to red) according to the scale on the right of each
graph to highlight the differences between them (points with similar colour are numerically close to each
other).