Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2022 Mar 15;66(3):e02203-21. doi: 10.1128/aac.02203-21

Simulated Intravenous versus Inhaled Tobramycin with or without Intravenous Ceftazidime Evaluated against Hypermutable Pseudomonas aeruginosa via a Dynamic Biofilm Model and Mechanism-Based Modeling

Hajira Bilal a, Jessica R Tait a,b, Yinzhi Lang c, Jieqiang Zhou c, Phillip J Bergen a, Anton Y Peleg d, Jürgen B Bulitta c, Antonio Oliver e, Roger L Nation b, Cornelia B Landersdorfer a,b,
PMCID: PMC8923196  PMID: 35041509

ABSTRACT

Acute exacerbations of chronic respiratory infections in patients with cystic fibrosis are highly challenging due to hypermutable Pseudomonas aeruginosa, biofilm formation and resistance emergence. We aimed to systematically evaluate the effects of intravenous versus inhaled tobramycin (TOB) with and without intravenous ceftazidime (CAZ). Two hypermutable P. aeruginosa isolates, CW30 (MICCAZ, 0.5 mg/liter; MICTOB, 2 mg/liter) and CW8 (MICCAZ, 2 mg/liter; MICTOB, 8 mg/liter), were investigated for 120 h in dynamic in vitro biofilm studies. Treatments were intravenous ceftazidime, 9 g/day (33% lung fluid penetration); intravenous tobramycin, 10 mg/kg of body every 24 h (50% lung fluid penetration); inhaled tobramycin, 300 mg every 12 h; and both ceftazidime-tobramycin combinations. Total and less susceptible planktonic and biofilm bacteria were quantified over 120 h. Mechanism-based modeling was performed. All monotherapies were ineffective for both isolates, with regrowth of planktonic (≥4.7 log10 CFU/ml) and biofilm (>3.8 log10 CFU/cm2) bacteria and resistance amplification by 120 h. Both combination treatments demonstrated synergistic or enhanced bacterial killing of planktonic and biofilm bacteria. With the combination simulating tobramycin inhalation, planktonic bacterial counts of the two isolates at 120 h were 0.47% and 36% of those for the combination with intravenous tobramycin; for biofilm bacteria the corresponding values were 8.2% and 13%. Combination regimens achieved substantial suppression of resistance of planktonic and biofilm bacteria compared to each antibiotic in monotherapy for both isolates. Mechanism-based modeling well described all planktonic and biofilm counts and indicated synergy of the combination regimens despite reduced activity of tobramycin in biofilm. Combination regimens of inhaled tobramycin with ceftazidime hold promise to treat acute exacerbations caused by hypermutable P. aeruginosa strains and warrant further investigation.

KEYWORDS: Pseudomonas aeruginosa, dosage regimen optimization, dynamic infection model, mathematical modeling, pharmacodynamics, pharmacokinetics

TEXT

Respiratory infections caused by Pseudomonas aeruginosa in patients with cystic fibrosis (CF) are a significant challenge (1). In CF, acute infective exacerbations (AIEs) with this pathogen and progressive pulmonary insufficiency are responsible for high morbidity and mortality (2). Over time, extensive use of antibiotics and significant pulmonary environmental pressures promote a phenotypic shift of P. aeruginosa (3). The hypermutator phenotype (with an up to 1,000-fold increased mutation rate) occurs in up to ∼65% of P. aeruginosa isolates from patients with CF (4, 5). These strains are associated with the transition of early (planktonic-phase) infection to a respiratory infection involving biofilm formation (6, 7). The biofilm harbors phenotypic diversity, restricts access of antibiotics to the infecting pathogen, and diminishes their antibacterial activity (2, 8). Furthermore, hypermutable P. aeruginosa strains develop resistance to antibiotics much more rapidly than nonhypermutable strains, are linked to a significant increase in multidrug resistance, and consequently present a substantial clinical challenge (9).

Intravenous ceftazidime and tobramycin are considered first-line treatment options for P. aeruginosa exacerbations, but their use as monotherapy is not recommended and information about rational dosing of this antibiotic combination in CF is limited (1012). P. aeruginosa has a large armamentarium of resistance mechanisms and can rapidly develop resistance following suboptimal antibiotic exposures, increasing the risk of therapeutic failure (13). In addition, for most antibiotics, including ceftazidime and tobramycin, the minimal biofilm inhibitory concentration is usually more than one 2-fold dilution higher than the MIC for planktonic bacteria, further complicating the treatment of AIEs in patients with CF (14).

While inhaled tobramycin is widely used in attempts to eradicate early acquisition of P. aeruginosa and control chronic infections, it is not usually employed in the management of P. aeruginosa AIEs (1517). This mode of administration ensures delivery of high concentrations of the antibiotic into lung fluid (1821). The effect of tobramycin concentration-time profiles that can be achieved with inhalation against hypermutable P. aeruginosa embedded in biofilms and in planktonic form has never been explored. Thus, we utilized a dynamic biofilm model to investigate the effect of clinically relevant pharmacokinetic (PK) profiles of ceftazidime and tobramycin (intravenous versus inhalation), as monotherapy and in combination, against clinical isolates of hypermutable P. aeruginosa. We developed the first mechanism-based mathematical model for the time course of viable counts from the dynamic biofilm model.

RESULTS

Pharmacokinetic validation and microbiological response.

The observed ceftazidime and tobramycin concentrations in the Centers for Disease Control and Prevention biofilm reactor (CBR) were, on average, within 5% of the targeted exposures that are shown in Table 1. Viable cell count profiles for total populations of planktonic and biofilm bacteria are shown in Fig. 1 for both isolates, and the corresponding profiles for less susceptible populations of CW30 and CW8 are in Fig. 2 and 3, respectively. Log changes in viable counts of total bacteria, mutant frequencies, and MICs of colonies from antibiotic-containing agar are in Tables 2, 3, and 4, respectively.

TABLE 1.

Clinically representative lung fluid concentrations, exposures, and PK/PD indices for CAZ and TOB against CW30 and CW8a

Isolate and treatment fCss, fCmax; fCmin (mg/liter) fAUC24 (mg·h/liter) fCss/MIC, fCmax/MIC; fCmin/MIC fT>MIC (%) fAUC24/MIC
CW30
 CAZ 9 g/day CI 18 432 36 100 864
 TOB 10 mg/kg q24h i.v. 12.3; 0.1 64.4 6.1; 0 32.2
 TOB 300 mg q12h INH 100; 9.4 928 50; 4.7 464
CW8
 CAZ 9 g/day CI 18 432 9 100 216
 TOB 10 mg/kg q24h i.v. 12.3; 0.1 64.4 1.5; 0 8.1
 TOB 300 mg q12h INH 100; 9.4 928 12.5; 1.2 116
a

CAZ, ceftazidime; TOB, tobramycin; CI, continuous intravenous infusion; q24h, every 24 h; q12h, every 12 h; i.v., intermittent intravenous infusion; INH, inhalation; fCss, unbound steady-state concentration; fCmax, unbound maximum concentration; fCmin, unbound minimum concentration before next dose; fAUC24, the area under the unbound concentration-time curve over 24 h; fCss/MIC, the ratio of fCss to MIC; fCmax/MIC, the ratio of fCmax to MIC; fCmin/MIC, the ratio of fCmin to MIC; fT>MIC, the cumulative percentage of a 24 h period that unbound concentrations exceeded the MIC; fAUC24/MIC, the ratio of fAUC24 to MIC. The simulated half-life in lung fluid was 3.5 h for tobramycin, as the tobramycin half-life is slightly longer in lung fluid than in plasma (66, 73). The simulated lung fluid penetration was 33% for ceftazidime and 50% for tobramycin (based on area under the concentration-time curve ratios) (66, 7377).

FIG 1.

FIG 1

Total viable counts for growth control and treatments with ceftazidime (CAZ) and/or tobramycin (TOB) with clinically relevant lung fluid concentration-time profiles. CAZ was administered by continuous infusion (CI) and TOB by intermittent intravenous or inhalational (INH) administration. Samples were from the media within the reactor (i.e., planktonic bacteria) and from coupons (i.e., biofilm bacteria). The y axis starts at the limit of counting. The results from treatment C for CW30 and A plus C for both isolates are presented as averages ± SE from two replicates.

FIG 2.

FIG 2

Effect of each dosage regimen on the counts of CW30 able to grow on agar plates containing 2.5 or 10 mg/liter ceftazidime or 5 or 10 mg/liter tobramycin. CAZ was administered by continuous infusion (CI), and TOB administration reflected intermittent intravenous infusion or inhalation (INH). The results for treatment C and treatment A plus C are presented as averages ± SE from of two replicates. Results below the limit of counting are plotted at zero. To differentiate less-susceptible subpopulations from the predominant population, the antibiotic concentrations in agar were based upon Etest MICs, which were 0.064 mg/liter for ceftazidime and 0.75 mg/liter for tobramycin (35).

FIG 3.

FIG 3

Effect of each dosage regimen on the counts of CW8 able to grow on agar plates containing 6 or 10 mg/liter of ceftazidime or 10 or 20 mg/liter of tobramycin. CAZ was administered by continuous infusion (CI), and TOB administration reflected intermittent intravenous infusion or inhalation (INH). The results from treatment A plus C are presented as averages ± SE from two replicates. Results below the limit of counting are plotted at zero. To differentiate less susceptible subpopulations from the predominant population, the antibiotic concentrations in agar were based upon Etest MICs, which were 0.75 mg/liter for ceftazidime and tobramycin (35).

TABLE 2.

Log changes in viable cell counts of total bacteria at various time points with clinically relevant lung fluid concentration exposures of ceftazidime and/or tobramycina

graphic file with name aac.02203-21_t002.jpg

a

CAZ, ceftazidime; TOB, tobramycin; CI, continuous intravenous infusion; Q24 h, every 24 h; Q12 h, every 12 h; IV, intermittent intravenous infusion; INH, inhalation; log10(CFUt), log10 CFU/ml or log10 CFU/cm2 at each sample collection time; log10(CFU0), log10 CFU/ml or log10 CFU/cm2 at time zero. The green background indicates synergy (a ≥2-log10 decrease in the CFU/ml or CFU/cm2 with the combination compared to its most active component and, for planktonic bacteria, a ≥2-log10 decrease in the CFU/ml compared to the initial inoculum); the blue background indicates a 1.0- to <2-log10 decrease in the number of CFU/ml or CFU/cm2 with the combination compared to its most active component.

TABLE 3.

Log10 mutant frequencies at 2.5 or 6 mg/liter and 10 mg/liter ceftazidime and 5 or 10 mg/liter and 10 or 20 mg/liter tobramycina

graphic file with name aac.02203-21_t003.jpg

a

CAZ, ceftazidime; TOB, tobramycin; CI, continuous intravenous infusion; Q24 h, every 24 h; Q12 h, every 12 h; IV, intermittent intravenous infusion; INH, inhalation. The red background indicates a high mutant frequency, i.e., a large proportion of less susceptible bacteria being present in the total population; the green background indicates a low mutant frequency, i.e., a small proportion of less susceptible bacteria being present in the total population. When no colonies were present on antibiotic-containing plates, mutant frequencies reported represent an upper limit based on the total viable count.

TABLE 4.

MIC values of colonies isolated from antibiotic-containing agar plates (10 mg/liter ceftazidime and 10 or 20 mg/liter tobramycin) at 0 and 120 h for each dosage regimena

Isolate and arm Time (h) CAZ, 10 mg/liter
TOB, 10 or 20 mg/liter
Planktonic bacteria Biofilm bacteria Planktonic bacteria Biofilm bacteria
CW30
 Control 0 NC NC NC NC
120 8 4 4 4
 A: CAZ 9 g/day CI 120 64 32
 B: TOB 10 mg/kg q24h i.v. 120 32 16
 C: TOB 300 mg q12h INH 120 16 8
 A + B 120 NC NC NC NC
 A + C 120 NC NC NC NC
CW8
 Control 0 8 8 16 8
120 32 16 32 16
 A: CAZ 9 g/day CI 120 128 128
 B: TOB 10 mg/kg q24h i.v. 120 64 32
 C: TOB 300 mg q12h INH 120 32 16
 A + B 120 16 NC NC NC
 A + C 120 8 NC NC NC
a

CAZ, ceftazidime; TOB, tobramycin; CI, continuous intravenous infusion; q24h, every 24 h; q12h, every 12 h; i.v., intermittent intravenous infusion; INH, inhalation; NC, no colonies grew on the antibiotic-containing agar plates; —, not tested.

Planktonic bacteria.

Tobramycin monotherapy simulating intravenous dosing produced initial killing of ≥2-log10 CFU/ml at 7 h followed by regrowth close to the growth control by 48 h for both isolates (Fig. 1, Table 2). With this treatment, amplification of resistance was observed at 72 and 120 h, with an increase of tobramycin-resistant populations compared to the growth control (Fig. 2 and 3, Table 3). The MIC of colonies recovered from tobramycin-containing agar at 120 h was 32 mg/liter for CW30 and 64 mg/liter for CW8 (Table 4).

For CW30, tobramycin inhalation produced extensive initial killing of ∼4.2 log10 CFU/ml at 7 h and viable counts remained suppressed at ∼4.7 log10 CFU/ml across 72 to 120 h, which was ∼4 log below that with intravenous tobramycin. For CW8, inhaled tobramycin achieved ∼2.7 log10 CFU/ml initial killing at 7 h, followed by slow regrowth to ∼5.9 log10 CFU/ml at 120 h, which was ∼2.5 log below the count for intravenous tobramycin (Fig. 1, Table 2). Less susceptible tobramycin populations at 120 h were slightly lower (CW30) (Fig. 2) or substantially higher (CW8) (Fig. 3) than for the growth control. For both isolates and inhaled tobramycin, MICs of colonies recovered from drug-containing agar were one 2-fold dilution lower than those from the intravenous tobramycin treatment (Table 4).

Ceftazidime monotherapy achieved ≥2.2-log10 CFU/ml initial killing at 7 h for both isolates, followed by regrowth (Fig. 1, Table 2). Less susceptible ceftazidime populations at 120 h had counts ∼2 to 5 log higher than those for the growth control (Fig. 2 and 3, Table 3); the MIC was 64 mg/liter for CW30 and 128 mg/liter for CW8 (Table 4).

For CW30, the combination of intravenous tobramycin with ceftazidime showed enhanced bacterial killing at 72 h and synergy at 96 and 120 h, whereas for CW8 enhanced activity was observed as early as 5 h and synergy was achieved at 48 h and beyond (Fig. 1, Table 2). No colonies were detected at 120 h on tobramycin-containing agar plates for both isolates and on ceftazidime-containing plates for CW30 (Fig. 2 and 3). With CW8, ≤3.2 log10 CFU/ml was observed on ceftazidime-containing plates at 120 h, i.e., ∼1 to 2.5 log10 CFU/ml below the growth control values; the MIC was 16 mg/liter (Fig. 3, Table 4).

The combination of tobramycin inhalation plus ceftazidime resulted in greater and more sustained killing than the combination with intravenous tobramycin; for CW30 and CW8, the counts (in CFU/ml) at 120 h of the combination simulating inhaled tobramycin were 0.47% and 36%, respectively, of those for the combination containing intravenous tobramycin. For CW30, enhanced bacterial killing was observed as early as 5 h, and synergy or enhanced activity was evident at all sampling times from 7 to 120 h. Viable counts of the total population were below the limit of counting at 28, 96, and 120 h (Fig. 1, Table 2), and no colonies were observed on agar containing either antibiotic (Fig. 2). For CW8, the combination simulating tobramycin inhalation produced enhanced activity at 5 h with ∼4 log10 CFU/ml bacterial killing. Enhancement or synergy was evident at all sampling times from 28 to 120 h (Fig. 1, Table 2). No colonies were detected on tobramycin-containing plates for either isolate and on ceftazidime-containing plates for CW30 (Fig. 2 and 3). For CW8, ∼1.2 log10 CFU/ml of bacteria less susceptible to ceftazidime were observed at 120 h; these counts were ∼3 to 4 log10 CFU/ml below those from the growth control and ∼1.5 log below the counts for the combination with intravenous tobramycin (Fig. 3). The MIC at 120 h was 8 mg/liter (Table 4).

Biofilm bacteria.

The intravenous tobramycin and ceftazidime monotherapies were ineffective against biofilm bacteria and produced only modest killing of both isolates. Tobramycin inhalation monotherapy achieved maximal killing of ∼3.3 log10 CFU/cm2 for CW30 and ∼0.9 log10 CFU/cm2 for CW8, i.e., viable counts ∼4.3 and ∼2.6 log10 below those for intravenous tobramycin (Fig. 1, Table 2). Amplification of ceftazidime- and tobramycin-resistant bacteria, compared to corresponding control values, was observed with all intravenous monotherapies (Fig. 2 and 3, Table 3). The MICs at 120 h are presented in Table 4. The combination simulating intravenous tobramycin produced synergy from 72 to 120 h for both isolates; ∼3 log10 CFU/cm2 on 5 mg/liter tobramycin-containing agar was observed at 120 h only for CW30, which was ∼1 log lower than that of the growth control (Fig. 2). The combination with tobramycin inhalation produced more pronounced killing for both isolates, with enhanced or synergistic effects evident beyond 24 to 48 h (Fig. 1, Table 2); for CW30 and CW8, the counts (in CFU/cm2) at 120 h for the combination simulating inhaled tobramycin were 8.2% and 13%, respectively, of those for the combination containing intravenous tobramycin. For CW30, ∼1.8 log10 CFU/cm2 of colonies was retrieved at 96 and 120 h; the viable count at this time was ∼1 log10 CFU/cm2 lower than that observed with the intravenous tobramycin combination. For CW8, the viable count from 48 to 120 h remained below ∼3 log10 CFU/cm2, and bacterial killing was ∼1 to ∼1.2 log10 CFU/cm2 greater than what occurred with the intravenous tobramycin combination. During treatment with the combination containing inhaled tobramycin, no colonies were observed on antibiotic-containing agar at any time (Fig. 2 and 3, Table 3).

Mechanism-based modeling.

The mechanism-based model (MBM) well described the planktonic and biofilm bacterial counts and yielded unbiased and precise curve fits for both isolates (Fig. 4; see also Fig. S1 in the supplemental material). For each mode of growth, the data from all dosage regimens and growth controls across the two different isolates could be described simultaneously by the same model and generally the same parameter estimates. Different parameter estimates were only required to describe differences in the initial biofilm inocula, abundance of preexisting resistant subpopulations, and tobramycin susceptibilities of the planktonic bacteria (Table 5). The lower estimate of KC50,SS (tobramycin concentration causing 50% of the maximum killing rate constant for the susceptible population) for planktonic bacteria of CW8 compared to CW30 was needed to describe the greater extent of initial bacterial killing by intravenous tobramycin of CW8 compared to CW30. The parameters were estimated with sufficient precision, as the standard errors (SE) were below 30% coefficient of variation for the majority of parameters.

FIG 4.

FIG 4

Total viable counts of planktonic and biofilm bacteria of isolates CW30 and CW8 for growth control and treatments with ceftazidime (CAZ) and/or tobramycin (TOB) with mechanism-based model fitted lines. Samples below the limit of counting (1.0 log10 CFU/ml, 1.6 log10 CFU/cm2) are plotted at zero. The results from treatment C for CW30 and A plus C for both isolates (open and filled symbols) are biological replicates that were modeled separately.

TABLE 5.

Parameter estimates for the MBM for CAZ and TOB in monotherapy and combination regimens against planktonic and biofilm bacteria of CW8 and CW30

Parameter Population mean estimate (SE%)
Symbol (unit) Planktonic Biofilm
Bacterial growth and subpopulations
 Initial inoculum Log10CFU0 7.05 (1.8) 5.44a (2.2), 6.28b (2.2)
 Maximum population size Log10CFUmax 8.97 (1.8) 7.62 (2.1)
 Mean generation time (MGT)
  CAZs-TOBs MGTSS (min) 90.3 (13) 457 (15)
  CAZR-TOBI MGTRI (min) 126 (9.2) 700 (13)
  CAZI-TOBR MGTIR (min) 240 (5.8) 2,377 (14)
 Log10 mutation frequency
  CAZ LogMUT,CAZ −4.64a (8.3), −4.52b (6.6) −1.74a (19), −3.19b (8.7)
  TOB LogMUT,TOB −3.36a (6.4), −3.75b (6.0) −1.57a (11), −3.33b (8.1)
Inhibition of successful replication by CAZ
 Maximum inhibition of successful replication I max,REP 0.958 (11) 0.881 (14)
 CAZ concn causing 50% of Imax,REP
  CAZs-TOBs IC50,REP,SS (mg/liter) 0.622 (36) 0.841 (35)
  CAZR-TOBI IC50,REP,RI (mg/liter) 128c (16) 102 (37)
  CAZI-TOBR IC50,REP,IR (mg/liter) 128c (16) 5.75 (26)
Bacterial killing by TOB
 Maximum killing rate constant Kmax,TOB (h−1) 7.37 (16) 0.283 (13)
 TOB concn causing 50% of Kmax,TOB
 CAZs-TOBs KC50,SS (mg/liter) 7.32a (35), 40.1b (24) 7.70c (22)
 CAZR-TOBI KC50,RI (mg/liter) 118a (49), 75.0b (19) 7.70c (22)
 CAZI-TOBR KC50,IR (mg/liter) 15,647 (31) 1,044 (19)
 Hill coefficient HillTOB 2.15a (39), 1b (fixed) 1 (fixed)
Mechanistic synergy
 Maximum fractional decrease of IC50,REP via mechanistic synergy I max,OM,TOB 0.795 (22) 0.661 (35)
 Concn of TOB causing 50% of Imax,OM,TOB IC50,OM,TOB (mg/liter) 1.74 (40) 16.1 (21)
 Hill coefficient HillOM 3.50a (50), 1b (fixed) 1 (fixed)
Residual variability
 SD of residual error on log10 scale SDCFU 0.442 (6.3) 0.306 (7.6)
a

CW8.

b

CW30.

c

The RI and IR subpopulations, or SS and RI subpopulations, were described by the same population mean estimate.

Comparison of the parameter estimates between planktonic and biofilm bacteria revealed a few trends across both isolates. The mean generation times (MGTs) for the three preexisting bacterial populations (i.e., double susceptible, ceftazidime resistant and tobramycin intermediate, and ceftazidime intermediate and tobramycin resistant) in the biofilm states were considerably (5- to 10-fold) higher than those estimated for the planktonic states. The parameter estimates representing inhibition of successful replication by ceftazidime were similar for biofilm and planktonic bacteria, except for a lower IC50,REP,IR (ceftazidime concentration causing 50% of maximum inhibition of successful replication of the ceftazidime-intermediate, tobramycin-resistant population) in biofilm (Table 5). In contrast, the maximum killing rate constant of tobramycin (Kmax,TOB) was estimated to be 26 times lower for biofilm than planktonic bacteria. Despite some KC50 estimates being higher for planktonic than biofilm bacteria, the estimated overall killing effect of tobramycin was greater on planktonic than biofilm bacteria for all three subpopulations. The very high estimates of KC50,IR indicate that the tobramycin-resistant planktonic and biofilm subpopulations were not killed by tobramycin in the model. In addition, the concentration of tobramycin required for 50% of the maximum extent of mechanistic synergy was estimated to be 9-fold higher for biofilm than planktonic bacteria, while the maximum extent of mechanistic synergy was similar for both modes of growth.

DISCUSSION

For β-lactams, including ceftazidime, the PK/pharmacodynamic (PD) index generally considered most predictive of antibacterial activity is the duration of the dosing interval over which the unbound concentration remains above a multiple of the MIC of the infecting pathogen (fT>MIC) (2226). For ceftazidime, favorable microbiological and clinical outcomes were more likely when fT>MIC was >45% (27). Since the maximum rate of bacterial killing was found to occur at an unbound ceftazidime concentration of ∼4× MIC, it was suggested that administration of ceftazidime as a continuous infusion to maintain unbound concentrations 4-fold higher than the MIC should maximize efficacy (25, 28). In the present CBR study, 9 g/day ceftazidime was simulated as a continuous infusion. For isolate CW30, the ceftazidime concentration was 36× MIC across the entire study duration; the corresponding value for CW8 was 9× MIC. Even with these high levels of exposure, ceftazidime monotherapy was unable to suppress regrowth and resistance emergence for both planktonic and biofilm bacteria of each hypermutable isolate. In contrast, an in vitro study using clinical strains of P. aeruginosa with preexisting resistant mutants suggested that an unbound ceftazidime concentration >3.8-fold higher than the MIC was needed to suppress the amplification of resistant subpopulations (29). That study examined nonhypermutable planktonic bacteria in a hollow-fiber infection model, whereas in the present study the interaction and cycling of biofilm and planktonic hypermutable bacteria may have decreased the ability to suppress amplification of resistant subpopulations (13, 30).

For aminoglycosides, antibacterial activity has been correlated with the ratio of unbound exposure across a 24-h period to MIC (fAUC/MIC) and the ratio of unbound maximum concentration to MIC (fCmax/MIC) (31); an fAUC/MIC of >70 and fCmax/MIC of 8 to 10 have been proposed as targets to increase likelihood of clinical success (32). In the present study, tobramycin was administered representing two modes of delivery, intravenous and inhalation. For the regimen simulating intravenous administration (10 mg/kg of body weight every 24 h [q24h]), the fAUC/MIC and fCmax/MIC of CW30 were 32.2 and 6.1, respectively; the corresponding values for CW8 were 8.1 and 1.5. The exposure values for inhaled delivery (300 mg every 12 h) were 464 and 50 for fAUC/MIC and fCmax/MIC for CW30 and 116 and 12.5 for fAUC/MIC and fCmax/MIC for CW8. The fAUC/MIC and fCmax/MIC values achieved with the intravenous regimen did not reach the above-mentioned PK/PD targets for either isolate, but the targets were exceeded with inhaled delivery against both isolates. However, neither of the tobramycin regimens in monotherapy was able to suppress the regrowth of both planktonic and biofilm bacteria. This result agreed with our previous in vitro studies where tobramycin monotherapy failed to inhibit regrowth of planktonic hypermutable P. aeruginosa even with fAUC/MIC exposures substantially higher than 70 (33, 34).

In the current study, the monotherapy regimens of ceftazidime and tobramycin were less effective against biofilm than planktonic bacteria. There are multiple mechanisms that may contribute to the relative resistance of P. aeruginosa biofilm bacteria, some of which are general in nature while others are specific to particular classes of antibiotics, such as β-lactams and aminoglycosides (8). The failure of each of ceftazidime at 9 g/day as a continuous infusion and tobramycin, even when administered by inhalation, to effectively suppress both bacterial growth forms strongly argues against the use of monotherapy for treatment of infections caused by hypermutable and biofilm-forming P. aeruginosa strains.

The combination regimen containing intravenous tobramycin increased bacterial killing and decreased emergence of resistant subpopulations relative to the monotherapy treatments. The increased antibacterial effect culminated in enhanced or synergistic killing of planktonic and biofilm bacteria of the isolates across the last 2 to 4 days of the 5-day study period. The extent of the increased killing of biofilm bacteria, relative to either antibiotic administered as monotherapy, was especially marked. However, the largest antibacterial effect occurred with the combination containing tobramycin inhalation. Against both isolates, that combination regimen produced enhanced or synergistic bacterial killing of planktonic bacteria on each of the 5 days of the study and of biofilm bacteria across the last 3 to 4 days, over which time at least a 1- to 2-log10 reduction in bacterial counts was observed relative to inhaled tobramycin alone. The combination regimen including inhaled tobramycin decreased the planktonic bacteria of both isolates to ≤2 log10 CFU/ml at 28 h (and at 96 to 120 h for CW30). The immune system might eliminate the remaining planktonic cells, which may or may not be the case for remaining biofilm bacteria. An additional notable finding of both combination regimens was resistance suppression, given the isolates were strong hypermutators and at baseline had mutations in genes associated with resistance to β-lactams and aminoglycosides (35).

Previous studies investigated ceftazidime in combination with tobramycin in static and dynamic in vitro studies and reported synergistic outcomes against P. aeruginosa (3639). However, all studies had one or more limitations, including that they were of short duration (24 to 48 h), examined only planktonic bacteria, and did not include hypermutable strains or report whether an isolate was hypermutable (3639). Ceftazidime in combination with tobramycin achieved significant bacterial reductions compared to the components in monotherapy against nonhypermutable P. aeruginosa in vivo (40, 41).

The enhanced and synergistic bacterial killing in the current study may be the result of the difference between the two antibiotics in mechanisms of action and resistance. Ceftazidime inhibits cell wall synthesis via binding to penicillin-binding proteins (PBPs), and major mechanisms of resistance in P. aeruginosa isolates involve chromosomally mediated AmpC β-lactamase overexpression, enzymatic inactivation via β-lactamases, and reduced affinity of PBPs (13, 42). Tobramycin blocks protein synthesis but also disrupts the outer bacterial membrane, very likely resulting in increased ceftazidime concentrations in the periplasmic space where the PBPs are located (4345). Resistance mechanisms of P. aeruginosa against aminoglycosides include increased expression of MexXY-OprM, target site modification, enzymatic cleavage, and reduced outer membrane permeability (4650). The mechanism by which the combination regimens achieved such remarkable activity toward biofilm bacteria in the current study is unknown but may be related to the ability of tobramycin to inhibit adhesion and microcolony formation during cycling of biofilm and planktonic cells (51).

There are a number of potential advantages of administering antibiotics via inhalation for the treatment of lung infections (52). These advantages include the ability to achieve a substantially higher lung-to-plasma concentration ratio than can be achieved with intravenous administration (53). This means that for antibiotics with narrow therapeutic windows (e.g., tobramycin), it is possible to achieve concentrations in lung much higher than can be safely achieved with intravenous administration; for tobramycin, this implies reduced risk of nephrotoxicity. For the last few decades antibiotics have been administered by inhalation for the management of chronic lung infections, including those occurring in patients with CF (54, 55). A recent Cochrane systematic review considered the randomized controlled trials that have been conducted in people with CF experiencing a pulmonary exacerbation in whom treatment with inhaled antibiotics was compared to placebo, standard treatment, or another inhaled antibiotic for between 1 and 4 weeks (15). Four trials with 167 participants were included in the review. Unfortunately, due to heterogeneity in trial design, high risk from lack of blinding, difficulty in assessing risk of bias, and lack of statistical power, the systematic review was unable to demonstrate whether or not one treatment was superior to the other. The authors concluded that further research is needed to establish whether inhaled tobramycin can be used as an alternative to intravenous tobramycin for pulmonary exacerbations (15). The results of the study reported here lend strong support for the conduct of well-designed clinical trials to evaluate the delivery of tobramycin by inhalation in patients having pulmonary exacerbations. In designing clinical trials, it should be recognized that P. aeruginosa can distribute across different airway niches in the lung of patients with cystic fibrosis, some of which can be better reached by intravenous administration (56). Thus, intravenous plus inhaled tobramycin in combination with ceftazidime should be considered for inclusion as a treatment arm in clinical trials. The limited systemic bioavailability of inhaled tobramycin (∼9 to 17%) generates relatively low plasma exposure (53), and limited clinical experience suggests generally good safety of coadministration of intravenous and inhaled tobramycin as a component of antibiotic treatment for pulmonary exacerbations in patients with CF (15, 57).

Our developed MBM well described the antibacterial effects of all monotherapy and combination regimens against planktonic and biofilm bacteria for both isolates. Longer estimated mean generation times for biofilm compared to planktonic bacteria reflected the slower growth of biofilm bacteria (8). The parameter estimates related to inhibition of successful replication by ceftazidime were similar between biofilm and planktonic bacteria, except for the IC50,REP of the tobramycin-resistant subpopulation. However, the slower growth of the biofilm bacteria resulted in an attenuated effect of ceftazidime monotherapy on biofilm compared to planktonic bacteria in the MBM, leading to a lower rate of bacterial killing. This is in agreement with the mechanism of action of ceftazidime affecting replicating bacteria (8). Similarly, the substantially lower maximum killing rate constant of tobramycin and higher tobramycin concentration required for 50% of the maximum mechanistic synergy for biofilm compared to planktonic bacteria reflect a lower activity of aminoglycosides in a biofilm matrix (8).

The strengths of this study include the following: it is the first study to examine the activity of clinically relevant regimens of ceftazidime and tobramycin in monotherapy and in combination against hypermutable isolates of P. aeruginosa in the CBR model; the impact of delivering tobramycin by inhalation versus intermittent intravenous infusions was investigated; the effects on both planktonic and biofilm bacteria were examined in a dynamic model over 5 days; and the time courses of both total and resistant subpopulations were characterized. The study also has some limitations. As with most nonclinical models, the planktonic and biofilm growth modes in the CBR model may not fully recapitulate those in the lungs of patients with CF, but a major advantage of the model we employed is that it enables accurate mimicking of the PK of antibiotics occurring in patients receiving clinical dosage regimens over 5 days. Like other in vitro infection models, the CBR also lacks an immune system and, therefore, the responses observed reflect the effects of the antibiotics only. While our previous studies on these isolates provided data on mutations in resistance genes present prior to treatment (35), we did not undertake genomic studies on emergent resistant populations of failed monotherapy regimens. It may also have been helpful to explore possible changes in biofilm structure in response to the different regimens. As is the case for most published MBM describing dynamic in vitro infection studies (5862), the total viable counts were modeled and bacterial counts on drug-containing agar were not incorporated in the MBM. However, our study is the first to develop an MBM based on data from the dynamic biofilm model; therefore, future studies may investigate an extension of the current MBM to characterize the resistance emergence.

In conclusion, this study has provided evidence that ceftazidime and tobramycin, when administered in monotherapy against hypermutable P. aeruginosa, are unable to provide sustained reduction in total and resistant subpopulations of both planktonic and biofilm bacteria. This supports guideline recommendations that these antibiotics should not be used in monotherapy regimens for treatment of pulmonary exacerbations. Indeed, the study demonstrated the enhanced and synergistic activity of combination therapy, especially with tobramycin administered by inhalation. The latter finding strongly supports the need for further studies to investigate the administration of antibiotics by inhalation in the treatment of infective exacerbations.

MATERIALS AND METHODS

Antibiotics, bacterial isolates, and MIC testing.

Solutions of ceftazidime (lot WS16174; Waterstone Technology, Carmel, IN, USA) and tobramycin (lot LC24138; AK Scientific, Union City, MD, USA) were prepared in sterile Milli-Q water immediately before each experiment. Cation-adjusted Mueller-Hinton agar (CAMHA) and broth (CAMHB) supplemented with 1% tryptic soy broth (TSB; BD, Sparks, MD, USA) were used in all experiments.

Two previously described clinical hypermutable P. aeruginosa isolates (CW30 and CW8) were examined; hypermutability is defined in the supplemental material (35). Agar dilution MICs determined for each isolate in triplicate on three separate days using Clinical and Laboratory Standards Institute guidelines (63) were 0.5 mg/liter and 2 mg/liter for ceftazidime and 2 mg/liter and 8 mg/liter for tobramycin for CW30 and CW8, respectively.

Dynamic biofilm model and antibiotic dosing.

The Centers for Disease Control and Prevention biofilm reactor (CBR), as previously described (64, 65), was used over 120 h to investigate the effects of ceftazidime and tobramycin alone and in combinations. Before each experiment, isolates were subcultured onto CAMHA and incubated at 36°C for 48 h. Colonies (2 to 3) were randomly selected and grown overnight in 10 ml of TSB, from which early-log-phase growth was obtained. Early-log-phase bacterial suspension (1 ml) was inoculated into each reactor containing 350 ml TSB. The 28-h conditioning phase was performed as described previously (64, 65). At 0 h (before starting treatment), the flow of medium (CAMHB–1% TSB) through each CBR was set to 1.16 ml/min to mimic a tobramycin half-life (t1/2) of 3.5 h in lung fluid (6568).

For ceftazidime, the highest recommended daily dose of 9 g/day for intravenous administration, and for tobramycin, doses of 10 mg/kg/day for intravenous administration and 300 mg q12h for inhalation, were simulated in the CBR (12). The PK profiles were based on those expected in lung fluid of patients with CF given the respective antibiotic regimens. These expected concentration-time profiles were simulated in silico using Berkeley Madonna (version 8.3.18) (69) based on clinical studies and population PK models for patients with CF (67, 7072). Information on lung fluid penetration of ceftazidime (33%) and tobramycin (50%) after intravenous administration was from published studies (66, 7377). The lung fluid concentration-time profiles of tobramycin following simulated inhalation were informed by clinical studies that quantified tobramycin by bioassay and high-performance liquid chromatography in epithelial lining fluid (20, 78) or sputum (21).

To simulate intravenous and inhalational dosing in the CBR, tobramycin was delivered via syringe driver over 30 min every 24 h and over 15 min every 12 h, respectively. As tobramycin does not accumulate following multiple intravenous administrations, no loading dose was required to achieve steady-state concentrations; for tobramycin inhalation, a small loading dose to attain fCmin (Table 1) was delivered at 0 h. Continuous infusion dosing of ceftazidime was achieved by administering a loading dose at 0 h directly into the reactor to immediately attain the required steady-state concentration (fCss), which was subsequently maintained by delivering medium containing the same concentration (fCss) of ceftazidime to the reactor (Table 1). Ceftazidime-containing medium was kept in the fridge to avoid thermal degradation and changed every 24 h.

Growth controls were included, and some monotherapy and combination treatments were studied in two biological replicates. Serial samples of planktonic and biofilm bacteria were collected aseptically at the times indicated in Table 2; total viable planktonic and biofilm bacteria were counted as previously described (64, 65). Less susceptible subpopulations were quantified at baseline (0 h) and during treatment at 24, 72, and 120 h on CAMHA supplemented with ceftazidime at 2.5 or 6 mg/liter and 10 mg/liter or tobramycin at 5 or 10 mg/liter and 10 or 20 mg/liter for CW30 and CW8, respectively. MICs of colonies isolated from antibiotic-containing agar at 0 and 120 h were determined to verify phenotypically the presence of stable resistance. Ceftazidime and tobramycin in PK samples were measured using validated liquid chromatography tandem mass spectrometry (LC-MS/MS) assays, as described in the supplemental material.

Pharmacodynamic analysis.

Microbiological responses to monotherapy and combination regimens were examined using the log change method, as described previously (65, 79). Synergy was considered ≥2 log10 CFU/ml or log10 CFU/cm2 more killing for the combination relative to the most active corresponding monotherapy at a specified time and, for planktonic cells, a viable cell count of ≥2 log10 CFU/ml below the initial inoculum (80). Combination regimens achieving bacterial killing of ≥1 to <2 log10 CFU/ml or log10 CFU/cm2 compared to the most active corresponding monotherapy at the same time were considered to represent enhanced antibacterial activity. Mutant frequencies were calculated as previously described (65).

Mechanism-based modeling methods are described in the supplemental material, and all parameters are explained in Table 5.

Data availability.

The figures and tables include data from the reported studies.

ACKNOWLEDGMENTS

This work was supported by the Australian National Health and Medical Research Council (NHMRC) project grant GNT1159579 and Ideas grant GNT1184428 to C.B.L., A.O., and R.L.N.

Footnotes

Supplemental material is available online only.

Supplemental file 1
Supplemental material. Download aac.02203-21-s0001.pdf, PDF file, 0.5 MB (474.3KB, pdf)

REFERENCES

  • 1.Moore JE, Mastoridis P. 2017. Clinical implications of Pseudomonas aeruginosa location in the lungs of patients with cystic fibrosis. J Clin Pharm Ther 42:259–267. 10.1111/jcpt.12521. [DOI] [PubMed] [Google Scholar]
  • 2.Malhotra S, Hayes D, Jr, Wozniak DJ. 2019. Cystic fibrosis and Pseudomonas aeruginosa: the host-microbe interface. Clin Microbiol Rev 32:e00138-18. 10.1128/CMR.00138-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Winstanley C, O'Brien S, Brockhurst MA. 2016. Pseudomonas aeruginosa evolutionary adaptation and diversification in cystic fibrosis chronic lung infections. Trends Microbiol 24:327–337. 10.1016/j.tim.2016.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Oliver A, Cantón R, Campo P, Baquero F, Blázquez J. 2000. High frequency of hypermutable Pseudomonas aeruginosa in cystic fibrosis lung infection. Science 288:1251–1254. 10.1126/science.288.5469.1251. [DOI] [PubMed] [Google Scholar]
  • 5.Oliver A. 2010. Mutators in cystic fibrosis chronic lung infection: prevalence, mechanisms, and consequences for antimicrobial therapy. Int J Med Microbiol 300:563–572. 10.1016/j.ijmm.2010.08.009. [DOI] [PubMed] [Google Scholar]
  • 6.Sousa AM, Pereira MO. 2014. Pseudomonas aeruginosa diversification during infection development in cystic fibrosis lungs–a review. Pathogens (Basel, Switzerland) 3:680–703. 10.3390/pathogens3030680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lujan AM, Macia MD, Yang L, Molin S, Oliver A, Smania AM. 2011. Evolution and adaptation in Pseudomonas aeruginosa biofilms driven by mismatch repair system-deficient mutators. PLoS One 6:e27842. 10.1371/journal.pone.0027842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ciofu O, Tolker-Nielsen T. 2019. Tolerance and resistance of Pseudomonas aeruginosa biofilms to antimicrobial agents-how P. aeruginosa can escape antibiotics. Front Microbiol 10:913. 10.3389/fmicb.2019.00913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Maciá MD, Blanquer D, Togores B, Sauleda J, Pérez JL, Oliver A, 2005. Hypermutation is a key factor in development of multiple-antimicrobial resistance in Pseudomonas aeruginosa strains causing chronic lung infections. Antimicrob Agents Chemother 49:3382–3386. 10.1128/AAC.49.8.3382-3386.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.West NE, Flume PA. 2019. Pulmonary exacerbations clinical care guidelines. https://www.cff.org/Care/Clinical-Care-Guidelines/Respiratory-Clinical-Care-Guidelines/Pulmonary-Exacerbations-Clinical-Care-Guidelines/.
  • 11.Castellani C, Duff AJA, Bell SC, Heijerman HGM, Munck A, Ratjen F, Sermet-Gaudelus I, Southern KW, Barben J, Flume PA, Hodková P, Kashirskaya N, Kirszenbaum MN, Madge S, Oxley H, Plant B, Schwarzenberg SJ, Smyth AR, Taccetti G, Wagner TOF, Wolfe SP, Drevinek P. 2018. ECFS best practice guidelines: the 2018 revision. J Cyst Fibros 17:153–178. 10.1016/j.jcf.2018.02.006. [DOI] [PubMed] [Google Scholar]
  • 12.Anonymous. 2009. Antibiotic treatment for cystic fibrosis. UK Cystic Fibrosis Trust guidelines, 3rd ed. Cystic Fibrosis Foundation, London, United Kingdom. https://www.cysticfibrosis.org.uk/sites/default/files/2020-11/Anitbiotic%20Treatment.pdf. Accessed 15 September 2021. [Google Scholar]
  • 13.Poole K. 2011. Pseudomonas aeruginosa: resistance to the max. Front Microbiol 2:65. 10.3389/fmicb.2011.00065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Macia MD, Rojo-Molinero E, Oliver A. 2014. Antimicrobial susceptibility testing in biofilm-growing bacteria. Clin Microbiol Infect 20:981–990. 10.1111/1469-0691.12651. [DOI] [PubMed] [Google Scholar]
  • 15.Smith S, Rowbotham NJ, Charbek E. 2018. Inhaled antibiotics for pulmonary exacerbations in cystic fibrosis. Cochrane Database Syst Rev 10:CD008319. 10.1002/14651858.CD008319.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Stuart B, Lin JH, Mogayzel PJ, Jr.. 2010. Early eradication of Pseudomonas aeruginosa in patients with cystic fibrosis. Paediatr Respir Rev 11:177–184. 10.1016/j.prrv.2010.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Langton Hewer SC, Smyth AR. 2017. Antibiotic strategies for eradicating Pseudomonas aeruginosa in people with cystic fibrosis. Cochrane Database Syst Rev 4:CD004197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Geller DE, Pitlick WH, Nardella PA, Tracewell WG, Ramsey BW. 2002. Pharmacokinetics and bioavailability of aerosolized tobramycin in cystic fibrosis. Chest 122:219–226. 10.1378/chest.122.1.219. [DOI] [PubMed] [Google Scholar]
  • 19.Bos AC, Mouton JW, van Westreenen M, Andrinopoulou ER, Janssens HM, Tiddens H. 2017. Patient-specific modelling of regional tobramycin concentration levels in airways of patients with cystic fibrosis: can we dose once daily? J Antimicrob Chemother 72:3435–3442. 10.1093/jac/dkx293. [DOI] [PubMed] [Google Scholar]
  • 20.Gibson RL, Emerson J, McNamara S, Burns JL, Rosenfeld M, Yunker A, Hamblett N, Accurso F, Dovey M, Hiatt P, Konstan MW, Moss R, Retsch-Bogart G, Wagener J, Waltz D, Wilmott R, Zeitlin PL, Ramsey B, Cystic Fibrosis Therapeutics Development Network Study Group. 2003. Significant microbiological effect of inhaled tobramycin in young children with cystic fibrosis. Am J Respir Crit Care Med 167:841–849. 10.1164/rccm.200208-855OC. [DOI] [PubMed] [Google Scholar]
  • 21.Ruddy J, Emerson J, Moss R, Genatossio A, McNamara S, Burns JL, Anderson G, Rosenfeld M. 2013. Sputum tobramycin concentrations in cystic fibrosis patients with repeated administration of inhaled tobramycin. J Aerosol Med Pulm Drug Deliv 26:69–75. 10.1089/jamp.2011.0942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Leggett JE, Fantin B, Ebert S, Totsuka K, Vogelman B, Calame W, Mattie H, Craig WA. 1989. Comparative antibiotic dose-effect relations at several dosing intervals in murine pneumonitis and thigh-infection models. J Infect Dis 159:281–292. 10.1093/infdis/159.2.281. [DOI] [PubMed] [Google Scholar]
  • 23.Turnidge JD. 1998. The pharmacodynamics of beta-lactams. Clin Infect Dis 27:10–22. 10.1086/514622. [DOI] [PubMed] [Google Scholar]
  • 24.Tozuka Z, Murakawa T. 2007. Beta-Lactam pharmacodynamics, p 129–146. In Nightingale CH, Ambrose PG, Drusano G, Murakami T (ed), Antimicrobial pharmacodynamics in theory and practice, 2nd ed. Informa Healthcare, New York, NY. [Google Scholar]
  • 25.Mouton JW, Punt N, Vinks AA. 2007. Concentration-effect relationship of ceftazidime explains why the time above the MIC is 40 percent for a static effect in vivo. Antimicrob Agents Chemother 51:3449–3451. 10.1128/AAC.01586-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.McKinnon PS, Paladino JA, Schentag JJ. 2008. Evaluation of area under the inhibitory curve (AUIC) and time above the minimum inhibitory concentration (T>MIC) as predictors of outcome for cefepime and ceftazidime in serious bacterial infections. Int J Antimicrob Agents 31:345–351. 10.1016/j.ijantimicag.2007.12.009. [DOI] [PubMed] [Google Scholar]
  • 27.Muller AE, Punt N, Mouton JW. 2013. Optimal exposures of ceftazidime predict the probability of microbiological and clinical outcome in the treatment of nosocomial pneumonia. J Antimicrob Chemother 68:900–906. 10.1093/jac/dks468. [DOI] [PubMed] [Google Scholar]
  • 28.Mouton JW, Vinks AA. 2007. Continuous infusion of beta-lactams. Curr Opin Crit Care 13:598–606. 10.1097/MCC.0b013e3282e2a98f. [DOI] [PubMed] [Google Scholar]
  • 29.Tam VH, Chang KT, Zhou J, Ledesma KR, Phe K, Gao S, Van Bambeke F, Sánchez-Díaz AM, Zamorano L, Oliver A, Cantón R. 2017. Determining β-lactam exposure threshold to suppress resistance development in Gram-negative bacteria. J Antimicrob Chemother 72:1421–1428. 10.1093/jac/dkx001. [DOI] [PubMed] [Google Scholar]
  • 30.Mah TF. 2012. Biofilm-specific antibiotic resistance. Future Microbiol 7:1061–1072. 10.2217/fmb.12.76. [DOI] [PubMed] [Google Scholar]
  • 31.Drusano GL. 2004. Antimicrobial pharmacodynamics: critical interactions of “bug and drug.” Nat Rev Microbiol 2:289–300. 10.1038/nrmicro862. [DOI] [PubMed] [Google Scholar]
  • 32.Roberts JA, Abdul-Aziz MH, Lipman J, Mouton JW, Vinks AA, Felton TW, Hope WW, Farkas A, Neely MN, Schentag JJ, Drusano G, Frey OR, Theuretzbacher U, Kuti JL, International Society of Anti-Infective Pharmacology and the Pharmacokinetics and Pharmacodynamics Study Group of the European Society of Clinical Microbiology and Infectious Diseases. 2014. Individualised antibiotic dosing for patients who are critically ill: challenges and potential solutions. Lancet Infect Dis 14:498–509. 10.1016/S1473-3099(14)70036-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Rees VE, Bulitta JB, Oliver A, Tsuji BT, Rayner CR, Nation RL, Landersdorfer CB. 2016. Resistance suppression by high-intensity, short-duration aminoglycoside exposure against hypermutable and non-hypermutable Pseudomonas aeruginosa. J Antimicrob Chemother 71:3157–3167. 10.1093/jac/dkw297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Landersdorfer CB, Rees VE, Yadav R, Rogers KE, Kim TH, Bergen PJ, Cheah S-E, Boyce JD, Peleg AY, Oliver A, Shin BS, Nation RL, Bulitta JB. 2018. Optimization of a meropenem-tobramycin combination dosage regimen against hypermutable and nonhypermutable Pseudomonas aeruginosa via mechanism-based modeling and the hollow-fiber infection model. Antimicrob Agents Chemother 62:e02055-17. 10.1128/AAC.02055-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Rees VE, Deveson Lucas DS, López-Causapé C, Huang Y, Kotsimbos T, Bulitta JB, Rees MC, Barugahare A, Peleg AY, Nation RL, Oliver A, Boyce JD, Landersdorfer CB. 2019. Characterization of hypermutator Pseudomonas aeruginosa isolates from patients with cystic fibrosis in Australia. Antimicrob Agents Chemother 63:e02538-18. 10.1128/AAC.02538-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.den Hollander JG, Horrevorts AM, van Goor ML, Verbrugh HA, Mouton JW. 1997. Synergism between tobramycin and ceftazidime against a resistant Pseudomonas aeruginosa strain, tested in an in vitro pharmacokinetic model. Antimicrob Agents Chemother 41:95–100. 10.1128/AAC.41.1.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zelenitsky SA, Karlowsky JA, Hoban DJ, Kabani A, Zhanel GG. 1998. Once versus thrice daily tobramycin alone and in combination with ceftazidime, ciprofloxacin and imipenem in an in vitro pharmacodynamic model. Chemotherapy 44:1–6. 10.1159/000007083. [DOI] [PubMed] [Google Scholar]
  • 38.Zelenitsky SA, Iacovides H, Harding GK, Ariano RE. 2004. Effect of antibiotic sequence on combination regimens against Pseudomonas aeruginosa in a multiple-dose, in vitro infection model. Diagn Microbiol Infect Dis 49:67–70. 10.1016/j.diagmicrobio.2003.10.016. [DOI] [PubMed] [Google Scholar]
  • 39.Tait JR, Bilal H, Kim TH, Oh A, Peleg AY, Boyce JD, Oliver A, Bergen PJ, Nation RL, Landersdorfer CB. 2021. Pharmacodynamics of ceftazidime plus tobramycin combination dosage regimens against hypermutable Pseudomonas aeruginosa isolates at simulated epithelial lining fluid concentrations in a dynamic in vitro infection model. J Glob Antimicrob Resist 26:55–63. 10.1016/j.jgar.2021.04.021. [DOI] [PubMed] [Google Scholar]
  • 40.Gordin FM, Rusnak MG, Sande MA. 1987. Evaluation of combination chemotherapy in a lightly anesthetized animal model of Pseudomonas pneumonia. Antimicrob Agents Chemother 31:398–403. 10.1128/AAC.31.3.398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Croisier D, Martha B, Piroth L, Chavanet P. 2008. In vivo efficacy of humanised intermittent versus continuous ceftazidime in combination with tobramycin in an experimental model of pseudomonal pneumonia. Int J Antimicrob Agents 32:494–498. 10.1016/j.ijantimicag.2008.07.010. [DOI] [PubMed] [Google Scholar]
  • 42.Bulitta JB, Ly NS, Yang JC, Forrest A, Jusko WJ, Tsuji BT. 2009. Development and qualification of a pharmacodynamic model for the pronounced inoculum effect of ceftazidime against Pseudomonas aeruginosa. Antimicrob Agents Chemother 53:46–56. 10.1128/AAC.00489-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kadurugamuwa JL, Clarke AJ, Beveridge TJ. 1993. Surface action of gentamicin on Pseudomonas aeruginosa. J Bacteriol 175:5798–5805. 10.1128/jb.175.18.5798-5805.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bulitta JB, Ly NS, Landersdorfer CB, Wanigaratne NA, Velkov T, Yadav R, Oliver A, Martin L, Shin BS, Forrest A, Tsuji BT. 2015. Two mechanisms of killing of Pseudomonas aeruginosa by tobramycin assessed at multiple inocula via mechanism-based modeling. Antimicrob Agents Chemother 59:2315–2327. 10.1128/AAC.04099-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yadav R, Bulitta JB, Schneider EK, Shin BS, Velkov T, Nation RL, Landersdorfer CB. 2017. Aminoglycoside concentrations required for synergy with carbapenems against Pseudomonas aeruginosa determined via mechanistic studies and modeling. Antimicrob Agents Chemother 61:e00722-17. 10.1128/AAC.00722-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Poole K. 2005. Aminoglycoside resistance in Pseudomonas aeruginosa. Antimicrob Agents Chemother 49:479–487. 10.1128/AAC.49.2.479-487.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Islam S, Oh H, Jalal S, Karpati F, Ciofu O, Høiby N, Wretlind B. 2009. Chromosomal mechanisms of aminoglycoside resistance in Pseudomonas aeruginosa isolates from cystic fibrosis patients. Clin Microbiol Infect 15:60–66. 10.1111/j.1469-0691.2008.02097.x. [DOI] [PubMed] [Google Scholar]
  • 48.Doi Y, Arakawa Y. 2007. 16S ribosomal RNA methylation: emerging resistance mechanism against aminoglycosides. Clin Infect Dis 45:88–94. 10.1086/518605. [DOI] [PubMed] [Google Scholar]
  • 49.Delcour AH. 2009. Outer membrane permeability and antibiotic resistance. Biochim Biophys Acta 1794:808–816. 10.1016/j.bbapap.2008.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Morita Y, Tomida J, Kawamura Y. 2012. MexXY multidrug efflux system of Pseudomonas aeruginosa. Front Microbiol 3:408. 10.3389/fmicb.2012.00408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Olivares E, Badel-Berchoux S, Provot C, Jaulhac B, Prevost G, Bernardi T, Jehl F. 2017. Tobramycin and amikacin delay adhesion and microcolony formation in Pseudomonas aeruginosa cystic fibrosis isolates. Front Microbiol 8:1289. 10.3389/fmicb.2017.01289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bos AC, Passé KM, Mouton JW, Janssens HM, Tiddens HAWM. 2017. The fate of inhaled antibiotics after deposition in cystic fibrosis: how to get drug to the bug? J Cyst Fibros 16:13–23. 10.1016/j.jcf.2016.10.001. [DOI] [PubMed] [Google Scholar]
  • 53.Magreault S, Roy C, Launay M, Sermet-Gaudelus I, Jullien V. 2021. Pharmacokinetic and pharmacodynamic optimization of antibiotic therapy in cystic fibrosis patients: current evidences, gaps in knowledge and future directions. Clin Pharmacokinet 60:409–445. 10.1007/s40262-020-00981-0. [DOI] [PubMed] [Google Scholar]
  • 54.Quon BS, Goss CH, Ramsey BW. 2014. Inhaled antibiotics for lower airway infections. Ann Am Thorac Soc 11:425–434. 10.1513/AnnalsATS.201311-395FR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Maselli DJ, Keyt H, Restrepo MI. 2017. Inhaled antibiotic therapy in chronic respiratory diseases. Int J Mol Sci 18:1062. 10.3390/ijms18051062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Bhagirath AY, Li Y, Somayajula D, Dadashi M, Badr S, Duan K. 2016. Cystic fibrosis lung environment and Pseudomonas aeruginosa infection. BMC Pulm Med 16:174. 10.1186/s12890-016-0339-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Stephens D, Garey N, Isles A, Levison H, Gold R. 1983. Efficacy of inhaled tobramycin in the treatment of pulmonary exacerbations in children with cystic fibrosis. Pediatr Infect Dis 2:209–211. 10.1097/00006454-198305000-00007. [DOI] [PubMed] [Google Scholar]
  • 58.Onufrak NJ, Smith NM, Satlin MJ, Bulitta JB, Tan X, Holden PN, Nation RL, Li J, Forrest A, Tsuji BT, Bulman ZP. 2020. In pursuit of the triple crown: mechanism-based pharmacodynamic modelling for the optimization of three-drug combinations against KPC-producing Klebsiella pneumoniae. Clin Microbiol Infect 26:1256–1256. 10.1016/j.cmi.2020.04.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Rao GG, Ly NS, Bulitta JB, Soon RL, San Roman MD, Holden PN, Landersdorfer CB, Nation RL, Li J, Forrest A, Tsuji BT. 2016. Polymyxin B in combination with doripenem against heteroresistant Acinetobacter baumannii: pharmacodynamics of new dosing strategies. J Antimicrob Chemother 71:3148–3156. 10.1093/jac/dkw293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Yadav R, Bergen PJ, Rogers KE, Kirkpatrick CMJ, Wallis SC, Huang Y, Bulitta JB, Paterson DL, Lipman J, Nation RL, Roberts JA, Landersdorfer CB. 2019. Meropenem-tobramycin combination regimens combat carbapenem-resistant Pseudomonas aeruginosa in the hollow-fiber infection model simulating augmented renal clearance in critically ill patients. Antimicrob Agents Chemother 64:e01679-19. 10.1128/AAC.01679-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Smith NM, Lenhard JR, Boissonneault KR, Landersdorfer CB, Bulitta JB, Holden PN, Forrest A, Nation RL, Li J, Tsuji BT. 2020. Using machine learning to optimize antibiotic combinations: dosing strategies for meropenem and polymyxin B against carbapenem-resistant Acinetobacter baumannii. Clin Microbiol Infect 26:1207–1213. 10.1016/j.cmi.2020.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Lin YW, Yu HH, Zhao J, Han ML, Zhu Y, Akter J, Wickremasinghe H, Walpola H, Wirth V, Rao GG, Forrest A, Velkov T, Li J. 2018. Polymyxin B in combination with enrofloxacin exerts synergistic killing against extensively drug-resistant Pseudomonas aeruginosa. Antimicrob Agents Chemother 62:e00028-18. 10.1128/AAC.00028-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.CLSI. 2020. Performance standards for antimicrobial susceptibility testing. M100. 30th ed. CLSI, Wayne, PA. [Google Scholar]
  • 64.Bilal H, Bergen PJ, Tait JR, Wallis SC, Peleg AY, Roberts JA, Oliver A, Nation RL, Landersdorfer CB. 2020. Clinically relevant epithelial lining fluid concentrations of meropenem with ciprofloxacin provide synergistic killing and resistance suppression of hypermutable Pseudomonas aeruginosa in a dynamic biofilm model. Antimicrob Agents Chemother 64:e00469-20. 10.1128/AAC.00469-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Bilal H, Bergen PJ, Kim TH, Chung SE, Peleg AY, Oliver A, Nation RL, Landersdorfer CB. 2019. Synergistic meropenem-tobramycin combination dosage regimens against clinical hypermutable Pseudomonas aeruginosa at simulated epithelial lining fluid concentrations in a dynamic biofilm model. Antimicrob Agents Chemother 63:e01293-19. 10.1128/AAC.01293-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Carcas AJ, Garcia-Satue JL, Zapater P, Frias-Iniesta J. 1999. Tobramycin penetration into epithelial lining fluid of patients with pneumonia. Clin Pharmacol Ther 65:245–250. 10.1016/S0009-9236(99)70103-7. [DOI] [PubMed] [Google Scholar]
  • 67.Hennig S, Standing JF, Staatz CE, Thomson AH. 2013. Population pharmacokinetics of tobramycin in patients with and without cystic fibrosis. Clin Pharmacokinet 52:289–301. 10.1007/s40262-013-0036-y. [DOI] [PubMed] [Google Scholar]
  • 68.Touw DJ, Jacobs FA, Brimicombe RW, Heijerman HG, Bakker W, Briemer DD. 1997. Pharmacokinetics of aerosolized tobramycin in adult patients with cystic fibrosis. Antimicrob Agents Chemother 41:184–187. 10.1128/AAC.41.1.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Macey RI. 2010. Berkeley Madonna version 8.3.18 441. University of California, Berkeley, CA. [Google Scholar]
  • 70.Bulitta JB, Landersdorfer CB, HüTtner SJ, Drusano GL, Kinzig M, Holzgrabe U, Stephan U, SöRgel F, 2010. Population pharmacokinetic comparison and pharmacodynamic breakpoints of ceftazidime in cystic fibrosis patients and healthy volunteers. Antimicrob Agents Chemother 54:1275–1282. 10.1128/AAC.00936-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Bensman TJ, Wang J, Jayne J, Fukushima L, Rao AP, D'Argenio DZ, Beringer PM. 2017. Pharmacokinetic-pharmacodynamic target attainment analyses to determine optimal dosing of ceftazidime-avibactam for the treatment of acute pulmonary exacerbations in patients with cystic fibrosis. Antimicrob Agents Chemother 61:e00988-17. 10.1128/AAC.00988-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Vinks AA, Brimicombe RW, Heijerman HG, Bakker W. 1997. Continuous infusion of ceftazidime in cystic fibrosis patients during home treatment: clinical outcome, microbiology and pharmacokinetics. J Antimicrob Chemother 40:125–133. 10.1093/jac/40.1.125. [DOI] [PubMed] [Google Scholar]
  • 73.Rodvold KA, George JM, Yoo L. 2011. Penetration of anti-infective agents into pulmonary epithelial lining fluid: focus on antibacterial agents. Clin Pharmacokinet 50:637–664. 10.2165/11594090-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 74.Boselli E, Breilh D, Rimmelé T, Poupelin J-C, Saux M-C, Chassard D, Allaouchiche B. 2004. Plasma and lung concentrations of ceftazidime administered in continuous infusion to critically ill patients with severe nosocomial pneumonia. Intensive Care Med 30:989–991. 10.1007/s00134-004-2171-2. [DOI] [PubMed] [Google Scholar]
  • 75.Dimelow R, Wright JG, MacPherson M, Newell P, Das S. 2018. Population pharmacokinetic modelling of ceftazidime and avibactam in the plasma and epithelial lining fluid of healthy volunteers. Drugs R D 18:221–230. 10.1007/s40268-018-0241-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Nicolau DP, Siew L, Armstrong J, Li J, Edeki T, Learoyd M, Das S. 2015. Phase 1 study assessing the steady-state concentration of ceftazidime and avibactam in plasma and epithelial lining fluid following two dosing regimens. J Antimicrob Chemother 70:2862–2869. 10.1093/jac/dkv170. [DOI] [PubMed] [Google Scholar]
  • 77.Cousson J, Floch T, Guillard T, Vernet V, Raclot P, Wolak-Thierry A, Jolly D. 2015. Lung concentrations of ceftazidime administered by continuous versus intermittent infusion in patients with ventilator-associated pneumonia. Antimicrob Agents Chemother 59:1905–1909. 10.1128/AAC.04232-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Rosenfeld M, Gibson R, McNamara S, Emerson J, McCoy KS, Shell R, Borowitz D, Konstan MW, Retsch-Bogart G, Wilmott RW, Burns JL, Vicini P, Montgomery AB, Ramsey B. 2001. Serum and lower respiratory tract drug concentrations after tobramycin inhalation in young children with cystic fibrosis. J Pediatr 139:572–577. 10.1067/mpd.2001.117785. [DOI] [PubMed] [Google Scholar]
  • 79.Albur M, Noel A, Bowker K, MacGowan A. 2012. Bactericidal activity of multiple combinations of tigecycline and colistin against NDM-1-producing Enterobacteriaceae. Antimicrob Agents Chemother 56:3441–3443. 10.1128/AAC.05682-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Pillai SK, Moellering RC, Eliopoulos GM. 2005. Antimicrobial combinations. In Lorian V, ed, Antibiotics in laboratory medicine. Lippincott Williams & Wilkins, Philadelphia, PA. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental file 1

Supplemental material. Download aac.02203-21-s0001.pdf, PDF file, 0.5 MB (474.3KB, pdf)

Data Availability Statement

The figures and tables include data from the reported studies.


Articles from Antimicrobial Agents and Chemotherapy are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES