Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2005 May;49(5):1943–1948. doi: 10.1128/AAC.49.5.1943-1948.2005

Pharmacodynamic Activity of Telithromycin at Simulated Clinically Achievable Free-Drug Concentrations in Serum and Epithelial Lining Fluid against Efflux (mefE)-Producing Macrolide- Resistant Streptococcus pneumoniae for Which Telithromycin MICs Vary

George G Zhanel 1,2,3,*, Christel Johanson 1, Nancy Laing 1, Tamiko Hisanaga 1, Aleksandra Wierzbowski 1, Daryl J Hoban 1,2
PMCID: PMC1087669  PMID: 15855517

Abstract

The present study, using an in vitro model, assessed telithromycin pharmacodynamic activity at simulated clinically achievable free-drug concentrations in serum (S) and epithelial lining fluid (ELF) against efflux (mefE)-producing macrolide-resistant Streptococcus pneumoniae. Two macrolide-susceptible (PCR negative for both mefE and ermB) and 11 efflux-producing macrolide-resistant [PCR-positive for mefE and negative for ermB) S. pneumoniae strains with various telithromycin MICs (0.015 to 1 μg/ml) were tested. The steady-state pharmacokinetics of telithromycin were modeled, simulating a dosage of 800 mg orally once daily administered at time 0 and at 24 h (free-drug maximum concentration [Cmax] in serum, 0.7 μg/ml; half-life [t1/2], 10 h; free-drug Cmax in ELF, 6.0 μg/ml; t1/2, 10 h). Starting inocula were 106 CFU/ml in Mueller-Hinton Broth with 2% lysed horse blood. Sampling at 0, 2, 4, 6, 12, 24, and 48 h assessed the extent of bacterial killing (decrease in log10 CFU/ml versus initial inoculum). Free-telithromycin concentrations in serum achieved in the model were Cmax 0.9 ± 0.08 μg/ml, area under the curve to MIC (AUC0-24 h) 6.4 ± 1.5 μg · h/ml, and t1/2 of 10.6 ± 0.6 h. Telithromycin-free ELF concentrations achieved in the model were Cmax 6.6 ± 0.8 μg/ml, AUC0-24 h 45.5 ± 5.5 μg · h/ml, and t1/2 of 10.5 ± 1.7 h. Free-telithromycin S and ELF concentrations rapidly eradicated efflux-producing macrolide-resistant S. pneumoniae with telithromycin MICs up to and including 0.25 μg/ml and 1 μg/ml, respectively. Free-telithromycin S and ELF concentrations simulating Cmax/MIC ≥ 3.5 and AUC0-24 h/MIC ≥ 25 completely eradicated (≥4 log10 killing) macrolide-resistant S. pneumoniae at 24 and 48 h. Free-telithromycin concentrations in serum simulating Cmax/MIC ≥ 1.8 and AUC0-24 h/MIC ≥ 12.5 were bacteriostatic (0.1 to 0.2 log10 killing) against macrolide-resistant S. pneumoniae at 24 and 48 h. In conclusion, free-telithromycin concentrations in serum and ELF simulating Cmax/MIC ≥ 3.5 and AUC0-24 h/MIC ≥ 25 completely eradicated (≥4 log10 killing) macrolide-resistant S. pneumoniae at 24 and 48 h.


Macrolide (azithromycin, clarithromycin, and erythromycin) resistance in Streptococcus pneumoniae is presently ∼25% in the United States and approximately 13% in Canada (1, 3, 28, 31). Macrolide resistance in S. pneumoniae involves alteration of the ribosomal target site or production and utilization of an efflux mechanism (6, 9, 29, 33). The production of ribosomal methylase, which alters the ribosomal target site of the macrolide, is usually coded for by the ermB gene and confers broad macrolide, lincosamide, and streptogramin B resistance (6, 9, 29, 33). The second mechanism, which results in macrolide efflux, is coded by the mefA or mefE genes (6, 9, 29, 33). Efflux is macrolide specific (14- and 15-membered macrolides only) and does not affect the lincosamide or streptogramins (28, 32). Note that ermB-positive S. pneumoniae strains generally exhibit high-level (MIC90 ≥ 64 μg/ml) macrolide resistance, while mefA- or mefE-positive S. pneumoniae strains exhibit low- to moderate-level resistance (MIC90 4 μg/ml) (6, 9, 29, 33). Both of these mechanisms are transmissible to other isolates (6, 9, 29, 33). Presently, in North America, mefE-positive S. pneumoniae is more common than ermB-positive S. pneumoniae and mefE strains make up the majority of macrolide-resistant S. pneumoniae strains (6, 9). In many European countries, ermB-positive S. pneumoniae strains are more prevalent (28, 32).

Although reports associating macrolide-resistant S. pneumoniae with macrolide clinical failure in the treatment of community-acquired respiratory infections are available, they are not that common (24).

Ketolides are a new class of semisynthetic agents derived from erythromycin A and are designed specifically to combat respiratory tract pathogens that have acquired resistance to macrolides (5, 7, 8, 11, 22, 26, 32). The main structural difference between ketolides and the macrolides is the lack of l-cladinose sugar at position 3 of the erythronolide A ring and its replacement with a 3-keto group (28, 33). Telithromycin and cethromycin (formerly ABT-773) have excellent in vitro activity against many pathogens causing community-acquired respiratory infections, including penicillin and macrolide-resistant strains (5-9, 22, 26, 28, 32). Ketolides demonstrate potent activity against most macrolide-resistant streptococci, including ermB- and mefA- or mefE-positive Streptococcus pneumoniae (5-9, 22, 26, 28, 32). Their pharmacokinetics display a long half-life (t1/2) as well as extensive tissue distribution and uptake into respiratory tissues and fluids, allowing for once-daily (OD) dosing (4, 12, 14, 15, 16, 19, 27). Presently only limited data are available on the pharmacodynamic activity of ketolides against macrolide-resistant S. pneumoniae in comparison to macrolides (13, 34).

The purpose of this study was to assess the pharmacodynamic activity of the ketolide telithromycin at simulated clinically achievable free-drug concentrations in serum (S) and epithelial lining fluid (ELF) against efflux-producing mefE macrolide-resistant S. pneumoniae.

MATERIALS AND METHODS

Bacterial strains and culture conditions.

Two macrolide-susceptible and 11 efflux-producing mefE macrolide-resistant strains of S. pneumoniae were evaluated (Table 1). As the mef gene in S. pneumoniae occurs as two variants, discrimination between mefA and mefE was performed by PCR-restriction fragment length polymorphism analysis according to a previously described protocol (2). Isolates were obtained from the Canadian Respiratory Organism Susceptibility Study (CROSS) (31). Telithromycin and azithromycin MICs are depicted in Table 1. The wild-type strains 11771 and 11888 were PCR-negative for mefA, mefE, and ermB and were macrolide-susceptible (azithromycin MIC ≤ 0.5 μg/ml). Macrolide-resistant (azithromycin MIC ≥ 2 μg/ml) strains were PCR-positive for mefE and PCR-negative for ermB (Table 1). Isolates were chosen to represent a variety of telithromycin MICs (0.015 to 1 μg/ml). The method and conditions used for PCR detection of mefE and ermB genotypes have been previously described (9).

TABLE 1.

Telithromycin susceptibilities of macrolide-susceptible and macrolide-resistant mefE S. pneumoniae

Isolate Result
mefE ermB Azithromycin MIC (μg/ml)a Clarithromycin MIC (μg/ml)b Clindamycin MIC (μg/ml)c Telithromycin MIC (μg/ml)d
11771 0.06 0.03 ≤0.12 0.008
11888 0.06 0.03 ≤0.12 0.008
12629 + 8 4 ≤0.12 0.015
35168 + 2 1 ≤0.12 0.03
8086 + 8 4 ≤0.12 0.06
16218 + 8 1 ≤0.12 0.06
11183 + 2 1 ≤0.12 0.12
18701 + 8 4 ≤0.12 0.12
17258 + 8 4 ≤0.12 0.25
1333 + 8 8 ≤0.12 0.5
UK185 + 16 8 ≤0.12 0.5
3543 + 16 8 ≤0.12 1
1217 + 16 16 ≤0.12 1
a

Susceptible, ≤0.5 μg/ml; intermediate, 1.0 μg/ml; resistant, ≥2 μg/ml.

b

Susceptible, ≤0.25 μg/ml; intermediate, 0.5 μg/ml; resistant, ≥1 μg/ml.

c

Susceptible, ≤0.25 μg/ml; intermediate, 0.5 μg/ml; resistant, ≥1 μg/ml.

d

Susceptible, ≤1 μg/ml; intermediate, 2 μg/ml; resistant, ≥4 μg/ml (17, 31, 33).

Antibiotic preparation and susceptibility testing.

Antibiotics were obtained as laboratory grade powders from their respective manufacturers. Stock solutions were prepared, and dilutions were made according to previously described methods (17). Following two subcultures from frozen stock, antibiotic MICs were determined by the NCCLS broth microdilution method (17, 18). All MIC determinations were performed in triplicate on separate days.

In vitro pharmacodynamic model.

The in vitro pharmacodynamic model used in this study has been previously described (21). Logarithmic phase cultures were prepared using a 0.5 McFarland (108 CFU/ml) standard by suspending several colonies in cation-supplemented Mueller-Hinton broth with 2% lysed horse blood (Oxoid, Nepean, Ontario, Canada) (pH 7.1). This suspension was diluted 1:100, and 20 μl of the diluted suspension was further diluted in 60 ml of cation-supplemented Mueller-Hinton broth with 2% lysed horse blood. The resulting suspension was allowed to grow overnight at 35°C in ambient air (21, 30, 34). After a maximum of 17 h, the suspension was further diluted to 1:10 and 60 ml of the diluted suspension was added to the in vitro pharmacodynamic model. Viable bacterial counts consistently yielded a starting inoculum of approximately 106 CFU/ml (21, 30, 34). This final inoculum was introduced into the central compartment (volume, 610 ml) of the in vitro pharmacodynamic model.

Pharmacokinetics and pharmacodynamics simulated.

Telithromycin was modeled based upon data obtained from previous publications (our target or simulated concentrations), simulating steady-state pharmacokinetics after a dosage of 800 mg orally (p.o.) OD (4, 12, 16, 28, 32). Thus, if after the administration of telithromycin at 800 mg, the maximum serum concentration (Cmax) was ∼2.2 μg/ml (and the serum protein binding was ∼70%) (4, 28, 32), it was assumed that the free Cmax in serum was ∼0.7 μg/ml. Thus, in serum (S) we simulated the maximum concentration [Cmax] at 0.7 μg/ml, t1/2 10 h. For epithelial lining fluid (ELF), it has been reported that the Cmax of telithromycin after 800 mg is ∼15 μg/ml (12). As the protein binding of telithromycin in ELF was not known, it was assumed to be similar to that of serum (70%) and thus only the likely concentration of free drug in ELF (Cmax ∼ 6.0 μg/ml) was simulated. Not knowing what the exact t1/2 of telithromycin was in ELF, we chose to simulate a t1/2 for telithromycin of ∼10 h for both serum and ELF and to simulate a slightly higher free-drug concentration in ELF (Cmax ∼ 6.0 μg/ml) knowing this would result in a larger AUC0-24 h. Telithromycin was administered once at time 0 and as a second dose at 24 h. Thus, two doses were administered every 24 h for 48 h. Pharmacodynamic experiments were performed in ambient air at 37°C. Samples were collected at 0, 1, 2, 4, 6, 12, 24, and 48 h for both pharmacokinetic and pharmacodynamic assessment (21, 30, 34). Telithromycin concentrations in the pharmacodynamic model were determined microbiologically with a bioassay (21, 30, 34). Actual or achieved telithromycin concentrations were determined in quadruplicate using Bacillus subtilis ATCC 6633 as the test organism with lower limits of quantification of 0.03 μg/ml. The plates were incubated aerobically for 18 h at 37°C. Concentrations were determined in relation to the diameters of the inhibition zones caused by the known concentrations from the standard series. The correlation coefficient of this assay was 0.80. Intra- and interrun variability of quality control samples were ≤6.5% and ≤5.8%, respectively. The actual or achieved concentrations of telithromycin and not the target or simulated concentrations were used in pharmacodynamic interpretations (e.g., Cmax/MIC and AUC0-24 h/MIC). Pharmacodynamic parameters of Cmax/MIC, AUC0-24 h/MIC, and time above the MIC (T > MIC) were derived from the actual or achieved telithromycin concentrations obtained in the model relative to the MIC of the strain in question.

Pharmacodynamic sampling was performed over 48 h with viable bacterial counts assessed by plating serial 10-fold dilutions onto cation-supplemented Mueller-Hinton agar with 2.0% lysed horse blood. Plates were incubated for 24 h at 37°C in ambient air. The lowest dilution plated was 0.1 ml of undiluted sample, and the lowest level of detection was 200 CFU/ml (2.0 log10) (21, 30, 34).

RESULTS

Table 1 shows the MICs of telithromycin and azithromycin against the clinical isolates utilized in this study. Strains were chosen to include macrolide-susceptible (wild-type) as well as low-level (MIC 2 to 4 μg/ml), intermediate (MIC 8 μg/ml), and high-level (MIC 16 μg/ml) macrolide-resistant mefE strains and ermB-positive S. pneumoniae. As well, isolates were chosen to represent a wide distribution of telithromycin MICs ranging from 0.008 μg/ml to 1 μg/ml. As shown in Table 1, all mefE strains were susceptible to clindamycin.

Pharmacokinetics.

Target (simulated) and actual (achieved) pharmacokinetic parameters of telithromycin after simulating a dosage of 800 mg p.o. OD (free serum and free epithelial lining fluid) achieved in the model were similar (Table 2). Target (simulated) and actual (achieved) pharmacokinetic parameters of telithromycin achieved in serum were as follows: free drug Cmax, 0.7 μg/ml (occurring at t = 0); AUC0-24 h, 4.5 μg · h/ml; t1/2, 10 h; and Cmax, 0.9 ± 0.08 (± standard deviation [SD]) μg/ml (occurring at t = 0); AUC0-24 h, 6.4 ± 1.5 (± SD) μg · h/ml; t1/2, 10.6 ± 0.6 (± SD) h, respectively. Telithromycin target (simulated) and actual (achieved) pharmacokinetic parameters achieved under free-drug conditions in ELF were CELF-free maximum, 6.0 μg/ml (occurring at t = 0); AUC0-24 h, 38.6 μg · h/ml; t1/2, 10 h; and CELF-free maximum, 6.6 ± 0.8 (± SD) μg/ml (occurring at t = 0); AUC0-24 h, 45.5 ± 5.5 (± SD) μg·h/ml; t1/2, 10.5 ± 1.7 (± SD) h, respectively.

TABLE 2.

Simulated (target) and achieved (actual) telithromycin pharmacokinetics

Pharmacokinetic parameter Resulta
Simulated (serum) Achieved (Serum) Simulated (ELF)b Achieved (ELF)
Cmax (μg/ml) ± SD 0.7 0.9 ± 0.08 6.0 6.6 ± 0.8
AUC0-24 h (μg · h/ ml) ± SD 4.5 6.4 ± 1.5 38.6 45.5 ± 5.5
t1/2 (h) ± SD 10 10.6 ± 1.6 10 10.5 ± 1.7
a

Target (simulated) and actual (achieved) pharmacokinetic parameters of telithromycin after simulating a dosage of 800 mg PO OD.

b

ELF, epithelial lining fluid.

Pharmacodynamics.

Table 3 describes the killing of S. pneumoniae with achieved telithromycin free-drug concentrations in serum. Free-telithromycin concentrations in serum resulted in bactericidal (≥3.0 log10 CFU/ml decrease versus initial inoculum) activity as early as 4 h for strains with telithromycin MICs ≤ 0.12 μg/ml (Table 3). This bactericidal activity was maintained for the entire 48 h of the experimental period. For strain 17258 with a telithromycin MIC of 0.25 μg/ml, free-telithromycin concentrations in serum were bacteriostatic (≤3.0 log10 CFU/ml decrease versus initial inoculum) for the first 12 h followed by complete bacterial eradication (≥4.0 log10 CFU/ml decrease versus initial inoculum) at 24 and 48 h (Table 3). Free-telithromycin concentrations in serum resulted in bacteriostatic (≤3.0 log10 CFU/ml decrease versus initial inoculum) activity over the entire 48 h period for strains with telithromycin MIC 0.5 μg/ml (Table 3). For strains with telithromycin MIC of 1 μg/ml, free-telithromycin concentrations in serum were bacteriostatic (≤3.0 log10 CFU/ml decrease versus initial inoculum) over the first 6 to 12 h followed by rapid regrowth at 24 and 48 h (Table 3).

TABLE 3.

Telithromycin killing of S. pneumoniae at simulated free-drug concentrations in serum and epithelial lining fluid

Strain (MIC [μg/ml]) Mean log10 cfu/ml killing at indicated h (serum result/epithelial lining fluid result)a
2 4 6 12 24 48
11771 (0.008) 1.9/3.1 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0
11888 (0.008) 1.7/3.3 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0
12629 (0.015) 1.7/3.1 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0
35168 (0.03) 1.1/3.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0
8086 (0.06) 1.0/3.0 3.2/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0
16218 (0.06) 0.9/3.0 3.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0
11183 (0.12) 0.9/3.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0
18701 (0.12) 1.1/3.2 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0
17258 (0.25) 0.5/0.2 0.6/1.2 1.6/2.0 2.0/≥4.0 ≥4.0/≥4.0 ≥4.0/≥4.0
1333 (0.5) 0.1/0.2 0.2/1.0 0.4/2.1 1.0/≥4.0 0.2/≥4.0 0.1/≥4.0
UK185 (0.5) 0/0.2 0.1/1.1 0.3/1.9 1.2/≥4.0 0.1/≥4.0 0.1/≥4.0
3543 (1) 0/0.3 0/0.7 0.4/1.0 0/3.2 0/≥4.0 0/≥4.0
1217 (1) 0/0.5 0.2/1.7 0.4/3.0 0.1/≥4.0 0/≥4.0 0/≥4.0
a

Growth reduction relative to initial inoculum. Growth controls started at 106 cfu/ml, reached 108 cfu/ml at 6 hours, and maintained this inoculum over the 48-hour study period.

Table 3 also describes the killing of S. pneumoniae with achieved free-telithromycin concentrations in epithelial lining fluid (ELF). Free-telithromycin concentrations in ELF resulted in bactericidal (≥3.0 log10 CFU/ml decrease versus initial inoculum) activity as early as 2 h for strains with telithromycin MICs ≤ 0.12 μg/ml (Table 3). This bactericidal activity was maintained for the entire 48 h of the experimental period. For strains with telithromycin MICs of 0.25 μg/ml and 0.5 μg/ml, free-telithromycin concentrations in ELF were bacteriostatic (≤3.0 log10 CFU/ml decrease versus initial inoculum) for the first 6 h followed by complete bacterial eradication (≥4.0 log10 CFU/ml decrease versus initial inoculum) at 12, 24, and 48 h (Table 3). For strains with telithromycin MICs of 1 μg/ml, free-telithromycin concentrations in ELF were bacteriostatic (≤3.0 log10 CFU/ml decrease versus initial inoculum) for the first 4 to 6 h followed by complete bacterial eradication (≥4.0 log10 CFU/ml decrease versus initial inoculum) at 12 to 24 h (Table 3).

The pharmacodynamic parameters associated with bacterial inhibition (decrease in log10 CFU/ml at 24 h versus initial inoculum) by telithromycin at simulated achieved free-drug concentrations in serum as well as in ELF are depicted in Tables 4, 5, and 6. Free-telithromycin concentrations in serum and ELF simulating Cmax/MIC ≥ 3.5 and AUC0-24 h/MIC ≥ 25 (time above the MIC [T > MIC] of 84%, shown for comparative purposes only) completely eradicated (≥4 log10 killing) macrolide-resistant S. pneumoniae at 24 and 48 h. Free-telithromycin concentrations in serum simulating Cmax/MIC ≥ 1.8 and AUC0-24 h/MIC ≥ 12.5 (time above the MIC [T > MIC] of 42%, shown for comparative purposes only) were bacteriostatic (0.1 to 0.2 log10 killing) against macrolide-resistant S. pneumoniae at 24 and 48 h. Free-telithromycin concentrations in serum simulating Cmax/MIC ≤ 0.9 and AUC0-24  h/MIC ≤ 6.3 (time above the MIC [T > MIC] of 0%, shown for comparative purposes only) resulted in regrowth of macrolide-resistant S. pneumoniae at 24 and 48 h.

TABLE 4.

Pharmacodynamics of telithromycin versus macrolide-susceptible and macrolide-resistant S. pneumoniae (T > MIC)a

Isolate/MIC (μg/ml) Free-drug result in:
Serum
ELF
T > MIC (% of dosing interval) Outcome at 24 hb T > MIC (% of dosing interval) Outcome at 24 h
11771/0.008 100 Ec 100 E
11888/0.008 100 E 100 E
12629/0.015 100 E 100 E
35168/0.03 100 E 100 E
8086/0.06 100 E 100 E
16218/0.06 100 E 100 E
11183/0.12 100 E 100 E
18701/0.12 100 E 100 E
17258/0.25 84 E 100 E
1333/0.5 42 0.2 100 E
UK185/0.5 42 0.1 100 E
3543/1 0 0 100 E
1217/1 0 0 100 E
a

Assumption made that protein binding in ELF was same as in serum (fraction unbound, 0.30).

b

Log10 killing at 24 hours (0 represents regrowth relative to the initial inoculum).

c

E, eradicated.

TABLE 5.

Pharmacodynamics of telithromycin versus macrolide-susceptible and macrolide-resistant S. pneumoniae (Cmax/MIC)a

Isolate/MIC (μg/ml) Free-drug result in:
Serum
ELF
Cmax/MIC Outcomeb Cmax/MIC Outcome
11771/0.008 113 Ec 825 E
11888/0.008 113 E 825 E
12629/0.015 56.3 E 413 E
35168/0.03 28.1 E 206 E
8086/0.06 14.1 E 103 E
16218/0.06 14.1 E 103 E
11183/0.12 7.0 E 51.6 E
18701/0.12 7.0 E 51.6 E
17258/0.25 3.5 E 25.8 E
1333/0.5 1.8 0.2 12.9 E
UK185/0.5 1.8 0.1 12.9 E
3543/1 0.9 0 6.4 E
1217/1 0.9 0 6.4 E
a

Assumption made that protein binding in ELF was same as in serum (fraction unbound, 0.30).

b

Log10 killing at 24 hours (0 represents regrowth relative to the initial inoculum).

c

E, eradicated.

TABLE 6.

Pharmacodynamics of telithromycin vs. macrolide-susceptible and macrolide-resistant S. pneumoniae (AUC0-24 h/MIC)

Isolate/MIC (μg/ml) Free-drug result in:
Serum
ELF
AUC0-24 h: MIC Outcomeb AUC0-24 h: MIC Outcome
11771/0.008 800 Ec 5688 E
11888/0.008 800 E 5688 E
12629/0.015 400 E 2844 E
35168/0.03 200 E 1422 E
8086/0.06 100 E 711 E
16218/0.06 100 E 711 E
11183/0.12 50 E 355 E
18701/0.12 50 E 355 E
17258/0.25 25 E 178 E
1333/0.5 12.5 0.2 89 E
UK185/0.5 12.5 0.1 89 E
3543/1 6.3 0 44 E
1217/1 6.3 0 44 E
a

Assumption made that protein binding in ELF was same as in serum (fraction unbound, 0.30).

b

Log10 killing at 24 hours (0 represents regrowth relative to the initial inoculum).

c

E, eradicated.

DISCUSSION

In a previous study, using this same in vitro model, we assessed telithromycin pharmacodynamic activity (against macrolide-susceptible and macrolide-resistant S. pneumoniae) at simulated clinically achievable free-drug concentrations in serum (S) and epithelial lining fluid (ELF) against strains with telithromycin MICs of 0.008 to 0.03 μg/ml. Against these very susceptible isolates, telithromycin serum and epithelial lining fluid concentrations resulted in eradication from the model in 4 h with no regrowth over 48 h (34). The purpose of this study was to assess the pharmacodynamic activity of telithromycin at simulated clinically achievable free-drug concentrations in serum and epithelial lining fluid (ELF) against efflux-producing macrolide-resistant S. pneumoniae with various telithromycin MICs (from 0.008 to 1 μg/ml). In this study, we modeled telithromycin based upon data obtained from previous publications of simulations of steady-state pharmacokinetics after a dosage of 800 mg p.o. OD (4, 12, 16, 28, 32). Thus, if after the administration of telithromycin 800 mg, the maximum concentration (Cmax) in serum was ∼2.2 μg/ml (and the serum protein binding was ∼70%) (4, 28, 32), it was assumed that the free Cmax in serum was ∼0.7 μg/ml. As our pharmacodynamic model contains no high molecular weight protein such as albumin, no protein binding occurs in the model and thus all drug that is added is non-protein-bound or free drug capable of crossing bacterial membranes and exerting a microbiological or pharmacological response. Assuming that, after administration of 800 mg of telithromycin, only the free drug in serum (Cmax, 0.7 μg/ml) is active and not the entire protein-bound and free drug (Cmax, 2.2 μg/ml) may underestimate the pharmacodynamic activity of telithromycin in serum. However, we chose in this study to study only the pharmacodynamic potential of the free drug. Thus, in serum (S) we simulated the maximum concentration at a Cmax of 0.7 μg/ml and a t1/2 of 10 h. For epithelial lining fluid (ELF), it has been reported that the Cmax of telithromycin after 800 mg is ∼15 μg/ml (12). As the protein binding of telithromycin in ELF was not known, it was assumed to be similar to that of serum (70%) and thus only the likely concentration of free drug in ELF (Cmax ∼ 6.0 μg/ml) was simulated. We chose to simulate a Cmax in ELF of ∼6.0 μg/ml and not 4.5 μg/ml because it has been reported that the t1/2 of telithromycin in ELF is longer than in serum (16). Not knowing what the exact t1/2 of telithromycin was in ELF, we chose to simulate a t1/2 for telithromycin of ∼10 h for both serum and ELF and to simulate a slightly higher free-drug concentration in ELF (Cmax ∼ 6.0 μg/ml) knowing this would result in a larger AUC0-24 h. As with serum, it was assumed that only the free drug in the ELF (Cmax ∼ 6.0 μg/ml) is active and not the entire protein-bound and free drug (Cmax ∼ 15 μg/ml). It is true that these assumptions may underestimate the pharmacodynamic activity of telithromycin in ELF; however, we chose in this study only to examine the pharmacodynamic potential of the free drug. It should be mentioned that the exact methods of how best to model ELF concentrations using an in vitro model are under debate.

Using the above-described pharmacodynamic model, we clearly showed that free-telithromycin concentrations in ELF rapidly eradicated macrolide-resistant S. pneumoniae with telithromycin MICs ranging from 0.015 μg/ml to 1 μg/ml (Table 3). Free-telithromycin concentrations in serum rapidly eradicated macrolide-resistant S. pneumoniae with telithromycin MICs up to and including 0.25 μg/ml (Table 3). Pharmacodynamically, free telithromycin concentrations in serum and ELF simulating Cmax/MIC ≥ 3.5 and AUC0-24 h/MIC ≥ 25 (time above the MIC [T > MIC] of 84%) completely eradicated (≥4 log10 killing) macrolide-resistant S. pneumoniae at 24 and 48 h (Tables 4, 5, and 6). It should be clear that although the pharmacodynamics of telithromycin correlate with Cmax/MIC and AUC0-24 h/MIC, we also showed the T > MIC for comparative purposes only and not to imply that the pharmacodynamics of telithromycin correlate with T > MIC.

Comparing the ketolide telithromycin to the macrolide azithromycin, we previously reported that azithromycin serum and epithelial lining fluid concentrations rapidly eradicated macrolide-susceptible S. pneumoniae but did not eradicate macrolide-resistant S. pneumoniae regardless of resistance phenotype (30). It should, however, be mentioned that our model simulates an immunocompromised host as no component of the immune system is added to the model. Thus, whether in an immunocompetent host, azithromycin can eradicate macrolide-resistant S. pneumoniae is not known. As the majority of S. pneumoniae in North America are macrolide-susceptible (∼75% in the United States and ∼87% in Canada), this may help to explain the excellent bacteriological and clinical outcomes obtained with macrolides (such as azithromycin) versus comparator antibiotics in clinical studies of community-acquired respiratory infections, such as community-acquired pneumonia, acute exacerbations of chronic bronchitis, acute sinusitis, and otitis media, where S. pneumoniae is a key pathogen (28). However, the rapid and extensive eradication of macrolide-resistant S. pneumoniae by telithromycin, when simulating clinically achievable free-drug concentrations in serum and epithelial ling fluid in this study, suggests that ketolides offer an advantage compared to macrolides such as azithromycin which are not able to eradicate macrolide-resistant S. pneumoniae (whether mefE or ermB) in serum, epithelial lining fluid, or middle ear fluid (30). These differences may help explain why ketolides compared to macrolides may result in reductions in hospitalization rates when treating community-acquired pneumonia (20, 25).

Only limited data are available regarding the pharmacodynamic properties of ketolides (4, 10, 13, 23, 34). Jacobs et al. demonstrated that against gram-positive cocci such as S. pneumoniae, telithromycin demonstrated postantibiotic effects of 0.3 to 3.8 h and postantibiotic sub-MIC effects of 0.8 to 4.6 h (10). It has been reported that telithromycin is a concentration-dependent bacterial killer with eradication being related to AUC/MIC and Cmax/MIC (4, 13, 14, 19, 34). Odenholt et al. reported that against S. pneumoniae, telithromycin demonstrated extremely fast (∼1 h) bactericidal (≥3 log10 killing) activity with Cmax/MIC ≥ 37.5 (23). Kim et al. using a murine pneumococcal pneumonia model reported that free Cmax/MIC and AUC0-24 h/MIC best explained the relationship between ketolide (cethromycin) drug exposure and reductions in viable bacterial counts (13). These authors documented free-drug Cmax/MIC of 1 and AUC0-24 h/MIC of 50 as resulting in bacteriostatic effects and maximal survival at free-drug Cmax/MIC and AUC0-24 h/MICs twice these amounts (13). In this study, we also observed very rapid bactericidal activity (within 4 h) against S. pneumoniae in simulations of free-telithromycin concentrations in serum and ELF, with pharmacodynamics of Cmax/MIC ≥ 7 and area under the curve to MIC (AUC0-24 h/ MIC) ≥ 50 (Tables 5 and 6).

In conclusion, telithromycin concentrations in serum and epithelial lining fluid rapidly eradicated efflux-producing macrolide-resistant S. pneumoniae with telithromycin MICs up to and including 0.25 and 1 μg/ml, respectively. Free-telithromycin concentrations in serum and ELF simulating Cmax/MIC ≥ 3.5 and area under the curve to MIC (AUC0-24 h/MIC) ≥ 25 completely eradicated (≥4 log10 killing) macrolide-resistant S. pneumoniae at 24 and 48 h. Finally, it should once again be mentioned that the exact methods of how best to model in vivo ELF concentrations using an in vitro model are under debate.

Acknowledgments

The expert secretarial assistance of M. Tarka is appreciated.

This study was supported in part by the University of Manitoba.

REFERENCES

  • 1.Butler, J. C., J. Hofmann, M. S. Cetron, J. A. Elliot, R. R. Facklam, and R. F. Breiman. 1996. The continued emergence of drug-resistant Streptococcus pneumoniae in the United States: an update from the Centers for Disease Control and Prevention's Pneumococcal Sentinel Surveillance System. J. Infect. Dis. 174:986-993. [DOI] [PubMed] [Google Scholar]
  • 2.Del Grosso, M., F. Iannelli, C. Messina, M. Santagati, N. Petrosillo, S. Stefani, G. Pozzi, and A. Pantosti. 2002. Macrolide efflux genes mefA and mefE are carried by different genetic elements in Streptococcus pneumoniae. J. Clin. Microbiol. 40:774-778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Doern, G. V., K. P. Heilmann, H. K. Huynh, P. R. Rhomberg, S. L. Coffam, and A. B. Brueggemann. 2001. Antimicrobial resistance among clinical isolates of Streptococcus pneumoniae in the United States during 1999-2000, including a comparison of rates since 1994-1995. Antimicrob. Agents Chemother. 45:1721-1729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Drusano, G. 2001. Pharmacodynamic and pharmacokinetic considerations in antimicrobial selection: Focus on telithromycin. Clin. Microbiol. Infect. 7(Suppl. 3):24-29. [PubMed] [Google Scholar]
  • 5.Farrell, D., and D. Felmingham. 2004. Activities of telithromycin against 13,874 Streptococcus pneumoniae isolates collected between 1999-2003. Antimicrob. Agents Chemother. 48:1882-1884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Farrell, D. J., and S. G. Jenkins. 2004. Distribution across the USA of macrolide resistance and macrolide resistance mechanism among Streptococcus pneumoniae isolates collected from patients with respiratory tract infections: PROTEKT US 2001-2002. J. Antimicrob. Chemother. 54(Suppl. 1):17-22. [DOI] [PubMed] [Google Scholar]
  • 7.Farrell, D. J., I. Morrissey, S. Bakker, S. Buckridge, and D. Felmingham. 2004. In vitro activities of telithromycin linezolid quinupristin-dalfopristin against Streptococcus pneumoniae with macrolide resistance due to ribosomal mutations. Antimicrob. Agents Chemother. 48:3169-3171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hoban, D. J., G. G. Zhanel, and J. A. Karlowsky. 1999. In vitro activity of the novel ketolide HMR 3647 and comparative oral antibiotics against Canadian respiratory tract isolates of Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis. Diagn. Microbiol. Infect. Dis. 35:37-44. [DOI] [PubMed] [Google Scholar]
  • 9.Hoban, D. J., A. Wierzbowski, K. A. Nichol, and G. G. Zhanel. 2001. Macrolide-resistant Streptococcus pneumoniae in Canada from 1998-1999: prevalence of mefA and ermB and susceptibility to ketolides. Antimicrob. Agents Chemother. 45:2147-2150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Jacobs, M. R., S. Bajaksouzian, and P. C. Appelbaum. 2003. Telithromycin post-antibiotic and post-antibiotic sub-MIC effects for 10 gram-positive cocci. J. Antimicrob. Chemother. 52(5):809-812. [DOI] [PubMed] [Google Scholar]
  • 11.Jorgensen, J. H., S. A. Crawford, M. L. McElmeel, and C. G. Whitney. 2004. Activities of cethromycin and telithromycin against recent North American isolates of Streptococcus pneumoniae. Antimicrob. Agents Chemother. 48:605-607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Khair, O. A., J. M. Andrews, D. Honeybourne, G. Jevons, F. Vacheron, and R. Wise. 2001. Lung concentrations of telithromycin after oral dosing. J. Antimicrob. Chemother. 47:837-840. [DOI] [PubMed] [Google Scholar]
  • 13.Kim, M., W. Zhou, P. R. Tessier, D. Xuan, M. Ye, C. H. Nightingale, and D. P. Nicolau. 2002. Bactericidal effect and pharmacodynamics of cethromycin (ABT-773) in a murine pneumococcal pneumonia model. Antimicrob. Agents Chemother. 46:3185-3192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Maglio, D., D. P. Nicolau, and C. H. Nightingale. 2003. Impact of pharmacodynamics on dosing of macrolides, azalides, ketolides. Infect. Dis. Clin. North Am. 17:563-577. [DOI] [PubMed] [Google Scholar]
  • 15.Muller-Serieys, C., P. Soler, C. Cantalloube, F. Lemaitre, H. P. Gia, F. Brunner, and A. Andremont. 2001. Bronchopulmonary disposition of the ketolide telithromycin (HMR 3647). Antimicrob. Agents Chemother. 45:3104-3108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Muller-Serieys, C., J. Andrews, F. Vacheron, and C. Cantalloube. 2004. Tissue kinetics of telithromycin, the first ketolide antibacterial. J. Antimicrob. Chemother. 53:149-157. [DOI] [PubMed] [Google Scholar]
  • 17.National Committee for Clinical Laboratory Standards. 2000. Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically. Approved standard M7-A5, 5th ed. National Committee for Clinical Laboratory Standards, Wayne, Pa.
  • 18.National Committee for Clinical Laboratory Standards. 2002. Performance standards for antimicrobial susceptibility testing: supplemental tables. M100-S12. National Committee for Clinical Laboratory Standards, Wayne, Pa.
  • 19.Nicolau, D. P. 2004. Clinical use of antimicrobial pharmacodynamic profiles to optimize treatment outcomes in community-acquired bacterial respiratory tract infections: Applications to telithromycin. Exp. Opin. Pharmacother. 5:229-235. [DOI] [PubMed] [Google Scholar]
  • 20.Niederman, M. S., J. R. Chang, J. Stewart, R. Nusrat, and R. B. Nieman. 2004. Comparison of hospitalization rates in patients with community acquired pneumonia treated with 10 days of telithromycin or clarithromycin. Curr. Med. Res. Opin. 20:740-756. [DOI] [PubMed] [Google Scholar]
  • 21.Noreddin, A., D. Roberts, K. Nichol, A. Wierzbowski, D. J. Hoban, and G. G. Zhanel. 2002. Pharmacodynamic modeling of clarithromycin against macrolide-resistant [PCR-positive mef(A) or erm(B)] Streptococcus pneumoniae simulating clinically achievable serum and epithelial lining fluid free-drug concentrations. Antimicrob. Agents Chemother. 46:4029-4034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Novotny, G. W., L. Jakobsen, N. M. Andrewsen, J. Poehlsgaard, and S. Douthwaite. 2004. Ketolide antimicrobial activity persists after disruption of interactions with domain II of 23S rRNA. Antimicrob. Agents Chemother. 48:3677-3683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Odenholt, I., E. Lowdin, and O. Cars. 2001. Pharmacodynamics of telithromycin in vitro against respiratory tract pathogens. Antimicrob. Agents Chemother. 45:23-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Rzeszutek, M., A. Wierzbowski, J. Conly, W. Bishai, D. Hoban, and G. G. Zhanel. 2004. A review of clinical failures involving macrolide-resistant Streptococcus pneumoniae. Int. J. Antimicrob. Agents 24:95-104. [DOI] [PubMed] [Google Scholar]
  • 25.Tellier, G., J. R. Chang, C. V. Asche, B. Lavin, J. Stewart, and S. D. Sullivan. 2004. Comparison of hospitalization rates in patients with community acquired pneumonia treated with telithromycin for 5 or 7 days or with clarithromycin for 10 days. Curr. Med. Res. Opin. 20:739-747. [DOI] [PubMed] [Google Scholar]
  • 26.Walsh, F., F. Carnegy, J. Willcock, and S. Amyes. 2004. Comparative in vitro activity of telithromycin against macrolide resistant and susceptible Streptococcus pneumoniae, Moraxella catarrhalis, and Haemophilus influenzae. J. Antimicrob. Chemother. 53:793-796. [DOI] [PubMed] [Google Scholar]
  • 27.Zhanel, G. G. 2001. Influence of pharmacokinetic and pharmacodynamic principles on antibiotic selection. Curr. Infect. Dis. Rep. 3:29-34. [DOI] [PubMed] [Google Scholar]
  • 28.Zhanel, G. G., M. Dueck, D. J. Hoban, L. Vercaigne, J. Embil, S. S. Gin, and J. A. Karlowsky. 2001. Macrolides and ketolides: a review focusing on respiratory infections. Drugs 61:443-498. [DOI] [PubMed] [Google Scholar]
  • 29.Zhanel, G. G., and J. A. Karlowsky. 2001. Ribosomal resistance: emerging problems and potential solutions. Curr. Infect. Dis. Rep. 1:459-463. [DOI] [PubMed] [Google Scholar]
  • 30.Zhanel, G. G., M. DeCorby, A. Noreddin, C. Mendoza, A. Cumming, K. Nichol, A. Wierzbowski, and D. J. Hoban. 2003. Pharmacodynamic activity of azithromycin against macrolide-susceptible and macrolide-resistant (PCR positive mefA or ermB) Streptococcus pneumoniae simulating clinically achievable free serum and epithelial lining fluid (ELF) and middle ear fluid (MEF) concentrations. J. Antimicrob. Chemother. 52:83-88. [DOI] [PubMed] [Google Scholar]
  • 31.Zhanel, G. G., L. Palatnick, K. Nichol, T. Bellyou, D. Low, and D. J. Hoban. 2003. Antimicrobial resistance in respiratory tract Streptococcus pneumoniae isolates: results of the Canadian Respiratory Organism Susceptibility Study, 1997 to 2002. Antimicrob. Agents Chemother. 47:1867-1874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zhanel, G. G., A. Wierzbowski, T. Hisanaga, and D. J. Hoban. 2004. The use of ketolides in the treatment of upper respiratory tract infections. Clin. Infect. Dis. Rep. 6:191-199. [DOI] [PubMed] [Google Scholar]
  • 33.Zhanel, G. G., T. Hisanaga, K. Nichol, A. Wierzbowski, and D. J. Hoban. 2003. Emerging treatments for macrolide resistant bacteria. Exp. Opin. Emerg. Drugs 8:297-321. [DOI] [PubMed] [Google Scholar]
  • 34.Zhanel, G.G., C. Johanson, T. Hisanaga, C. Mendoza, N. Laing, A. Noreddin, A. Wierzbowski, and D. J. Hoban. Pharmacodynamic activity of telithromycin against macrolide-susceptible and macrolide-resistant Streptococcus pneumoniae simulating clinically achievable free serum and epithelial lining fluid concentrations. J. Antimicrob. Chemother. 54:1072-1077. [DOI] [PubMed]

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

RESOURCES