147
Views
3
CrossRef citations to date
0
Altmetric
Review

Moxifloxacin in the treatment of skin and skin structure infections

Pages 417-434 | Published online: 24 Dec 2022

Abstract

Moxifloxacin is a recent addition to the fluoroquinolone class, differing from ciprofloxacin and other older agents in having much better in vitro activity against Gram-positive aerobes while retaining potent activity against Gram-negative aerobes. It is also active against the pathogens of human and animal bite wounds and those species of atypical mycobacteria associated with dermatologic infections. Its activity against anaerobes is quite variable. Moxifloxacin penetrates well into inflammatory blister fluid and muscle and subcutaneous adipose tissues. Moxifloxacin should thus be a reasonable option for the treatment of skin and skin structure infections (SSSIs). In 3 randomized controlled trials (RCTs), oral moxifloxacin was as effective as cephalexin in the treatment of uncomplicated SSSIs in adults while in 2 RCTs, intravenous/oral moxifloxacin was as effective as intravenous/oral β-lactam/β-lactamase inhibitor therapy in the treatment of complicated SSSIs in adults. Moxifloxacin does not inhibit cytochrome P450 enzymes and thus interact with warfarin or methylxanthines. However, multivalent cations can reduce its oral bioavailability substantially. Dosage adjustment is not required in the presence of renal or hepatic impairment. The clinical relevance of its electrophysiologic effects (QTc prolongation) remains unresolved.

Introduction

Moxifloxacin is one of most-recently marketed of the fluoroquinolone (hereafter called quinolone) antimicrobials (Avelox®, Bayer, Leverkusen, Germany). Of this group (gatifloxacin, gemifloxacin, moxifloxacin), only moxifloxacin has regulatory approval for use in skin and skin structure infections (SSSIs) (CitationAnonymous 2005). Until recently, this approval was limited to uncomplicated SSSIs in adults caused by Staphylococcus aureus or Streptococcus pyogenes (CitationAnonymous 2005). In June of 2005, it also received approval for use in complicated SSSIs caused by methicillin-susceptible S. aureus, Escherichia coli, Klebsiella pneumoniae, and Enterobacter cloacae (CitationAnonymous 2005). Complicated SSSIs are defined as those which involve deeper layers such as fascia or muscle, require significant surgical interventions, or arise in the presence of significant comorbidities such as diabetes or HIV infection (CitationRaghavan and Linden 2004). The purpose of this paper is to review the in vitro antimicrobial activity, pharmacokinetics, clinical and bacteriological efficacy, safety, tolerability, and drug–drug interaction potential of moxifloxacin, concentrating on its potential role in the treatment of complicated (and uncomplicated) SSSIs.

Mechanisms of action and resistance

Moxifloxacin acts by the same mechanism as other quinolones: inhibition of bacterial DNA gyrase (a Type II topoisomerase composed of two gyr A and two gyr B subunits) and topoisomerase IV (a heterotetramer made up of two par C and two par E subunits (CitationRuiz 2003; CitationMitscher 2005). Topoisomerase IV is felt to be the preferred target, especially in Gram-positive aerobes (CitationInce et al 2003). Resistance is mediated in all quinolones by either changes in the target site (DNA gyrase and/or topoisomerase IV) or changes that alter intracellular drug accumulation (ie, enhanced efflux).

In vitro antimicrobial activity

Unlike earlier quinolones like ciprofloxacin, moxifloxacin has significant activity against Gram-positive aerobes, including potential dermatologic pathogens such as S. aureus, coagulase-negative staphylococci, and groups A/B/C/F/G streptococci () (CitationDalhoff et al 1996; CitationBauernfeind 1997; CitationFass 1997; CitationGoldstein et al 1997, Citation2000; CitationWoodcock et al 1997; CitationBarry et al 1999; CitationMalathum et al 1999; Citationvon Eiff et al 1999; CitationBlondeau et al 2000; CitationFung-Tomc et al 2000; CitationHardy et al 2000; CitationHoogkamp-Korstanje et al 2000; CitationMilatovic et al 2000; CitationAmabile-Cuevas et al 2001; CitationBlondeau 2002; CitationSpeciale et al 2002; CitationBowker et al 2003; CitationHsueh et al 2003; CitationNoviello et al 2003; CitationEdmiston et al 2004, Citation2005; CitationJacobs et al 2004; CitationPatel et al 2004; CitationWenzler et al 2004; CitationBogdanovich et al 2005; CitationStratchounski et al 2005). Methicillin-resistant strains of staphylococci are much less susceptible and enterococci are only moderately susceptible to moxifloxacin. Moxifloxacin still retains significant activity against Gram-negative aerobes with the exception of variable activity against Pseudomonas aeruginosa, other pseudomonads, and Stenotrophomonas maltophilia (). Among anaerobes, moxifloxacin is quite active against the majority of pathogens of animal and human bite wounds. Activity against Bacteroides, Fusobacterium, and Prevotella species is variable () (CitationDalhoff et al 1996; CitationAldridge and Ashcraft 1997; CitationBauernfeind 1997; CitationFass 1997; CitationGoldstein et al 1997; CitationMacGowen et al 1997; CitationWoodcock et al 1997; CitationEdlund et al 1998; CitationBetriu et al 1999; CitationAckermann et al 2000; CitationFung-Tomc et al 2000; CitationGoldstein et al 2000, Citation2002, Citation2006; CitationHoogkamp-Korstanje et al 2000; CitationMilatovic et al 2000; CitationSchaumann et al 2000; CitationHoellman et al 2001; CitationSnydman et al 2002; CitationSpeciale et al 2002; CitationEdnie et al 2003; CitationHedberg et al 2003; CitationEdmiston et al 2004, Citation2005; CitationPeric et al 2004; CitationSillerstrom et al 2004; CitationLion et al 2006).

Table 1 In vitro activity of moxifloxacin against potential Gram-positive and Gram-negative aerobic pathogens in SSSIs

Table 2 In vitro activity of moxifloxacin against potential bite wound pathogens and Gram-positive and Gram-negative anaerobic pathogens in SSSIs

Moxifloxacin has significant activity against many strains of atypical mycobacteria, microorganisms of significance in dermatological infections. For example, the following minimum inhibitory concentrations of 90% of isolates (MIC90) values (in mg/L) have been reported for a) Mycobacterium marinum, b) M. fortuitum, c) M. chelonae, d) M. abscessus, e) M. kansasii, and f) M. gordonae: a) 8.0 (43 isolates) (CitationBraback et al 2002), 1.0 (53 isolates) (CitationAubry et al 2000); b) 16.0 (69 isolates) (CitationYang et al 2003), 1.0 (7 isolates) (CitationGillespie and Billington 1999), 0.5 (16 isolates) (CitationRodriguez Diaz et al 2003); c) 8.0 (39 isolates) (CitationYang et al 2003), 16.0 (4 isolates) (CitationRodriguez Diaz et al 2003); d) 16.0 (92 isolates) (CitationYang et al 2003); e) 0.06 (16 isolates) (CitationGillespie and Billington 1999), 0.06 (148 isolates) (CitationAlcaide et al 2004), 2.0 (8 isolates) (CitationRodriguez Diaz et al 2003); f) 0.5 (23 isolates) (CitationRodriguez Diaz et al 2003).

Important microorganisms in the context of skin and skin structure infections related to foreign devices are the coagulase-negative staphylococci (CNS), especially those strains able to produce biofilm (an extracellular glycocalyx material also called “slime”). Moxifloxacin’s activity has been evaluated in vitro against 19 biofilm-producing (MIC90 = 2 mg/L) and 22 biofilm-nonproducing (MIC90 = 0.25 mg/L) strains of CNS. SubMIC concentrations of moxifloxacin did not modify biofilm production. It had poor activity against stationary phase microorganisms but had some activity against microorganisms in the biofilm. However, high drug concentrations were necessary for this activity (50- and 100-fold MIC concentrations produced mean 1.10 and 1.69 log reductions in bacterial counts, respectively) (CitationPerez-Giraldo et al 2004). In these regards, moxifloxacin unfortunately behaved like many other antimicrobials evaluated previously.

In another study with potential application to foreign device-related infections, an in vitro study was performed evaluating the bactericidal activity of moxifloxacin monotherapy and combination moxifloxacin plus vancomycin/teicoplanin therapy in 9 S. aureus isolates (1 being methicillin- and quinolone-sensitive and 8 being resistant to both). These isolates came from patients unresponsive to glycopeptide therapy or relapsing after completion of glycopeptide therapy despite device removal. Moxifloxacin MICs were 0.125 mg/L (in the 1 methicillin-sensitive isolate) and 2.0 mg/L (in all of the methicillin-resistant isolates). All isolates were tolerant to vancomycin, teicoplanin, and moxifloxacin at medium concentrations 2 X MIC. However, moxifloxacin plus glycopeptide combination therapy was bactericidal at the glycopeptide MIC and 0.5 X MIC of moxifloxacin (CitationTarasi et al 2003). Thus, combination therapy may be worthwhile considering in difficult-to-treat patients.

Characteristic of quinolones, moxifloxacin has concentration-dependent bactericidal activity (CitationDalhoff et al 1996). This has been shown with methicillin-sensitive and -resistant S. aureus and coagulase-negative staphylococci (CitationLister 2001, CitationSpeciale et al 2002; CitationNoviello et al 2003; CitationBogdanovich et al 2005), S. pyogenes (CitationEsposito et al 2000; CitationOdenholt et al 2002; CitationSpeciale et al 2002; CitationNoviello et al 2003), Enterococcus faecalis (CitationSpeciale et al 2002), and a variety of anaerobes (CitationCredito et al 2003). Bactericidal activity using serum bactericidal titer technology has been demonstrated for S. aureus, S. epidermidis, S. pyogenes, E. coli, and Klebsiella pneumoniae (CitationLemmen et al 2003; CitationDan et al 2004). Also, bactericidal activity using synovial fluid bactericidal titer technology has been demonstrated for S. aureus, S. pyogenes, and K. pneumoniae (CitationDan et al 2004). Activity against S. pyogenes is independent of the presence/absence of macrolide resistance determinants (CitationCritchley et al 2002).

Two recent studies have evaluated the activity of moxifloxacin in in vitro pharmacokinetic/pharmacodynamic models (IVP/PM) of mixed aerobic-anaerobic infection. CitationHermsen and colleagues (2005) demonstrated bactericidal activity against Escherichia coli and Bacteroides fragilis in a study using a limited number of isolates of both microorganisms. Using different strains of the same microorganisms, Schaumann and colleagues (2005) demonstrated that the presence of B. fragilis did not affect the activity of moxifloxacin against E. coli but the presence of E. coli reduced the activity against B. fragilis significantly.

Another recent study evaluated the pharmacodynamics of moxifloxacin against anaerobes in an IVP/PM. B. fragilis, Clostridium perfringens, and Gram-positive anaerobic cocci (GPAC) were studied. Dose-ranging using area under the concentration-time curve/minimum inhibitory concentration (AUC/MIC) ratios of 6.7 to 890 (for the reference aerobe E. coli) and 9 to 216 (for the anaerobes) revealed differing AUC/MIC antibacterial effect patterns for anaerobes versus the aerobe. For E. coli, the AUC/MIC ratios for 50%/90% effect were 34/59 while the corresponding values for B. fragilis, C. perfringens, and GPAC were 11/26, 9/16, and 7/17, respectively. At similar AUC/MIC ratio values, the activity against anaerobes was less than that against the aerobe. Using Monte Carlo simulations of human pharmacokinetic data and a target anaerobic AUC/MIC ratio value of 7.5, over 90% achievement of this target was possible at MICs < 2 mg/L. The clinical implications of this “pharmacokinetic breakpoint” of 2 mg/L for anaerobes deserves further study (CitationNoel et al 2005).

Despite 10- to 20-fold accumulation of drug intracellularly (vida infra), the bactericidal effect of moxifloxacin intracellularly develops slowly, with only a 1.51 log reduction in post-phagocytosis S. aureus inoculum within 24 h (CitationSeral et al 2003). This may be explainable, in part, by the deleterious effect of acid pH on activity. For example, at pH 5 (mimicking phagolysosome pH), moxifloxacin activity falls at least 4-fold compared with physiologic pH (CitationSeral et al 2003). However, over a pH range of 6 to 8, moxifloxacin activity is unaffected for S. aureus and S. pyogenes (CitationDalhoff et al 2005).

The presence of 10%, 30%, and 50% albumin and human serum had no significant effect on the in vitro activity of moxifloxacin (CitationWoodcock et al 1997; CitationRubinstein et al 2000). The presence of dead bacteria, sterile pus under aerobic conditions, sterile pus under anaerobic conditions, and anaerobic conditions alone had no significant effect on the in vitro activity of moxifloxacin against methicillin-sensitive and -resistant staphylococci, S. pyogenes, and E. coli (CitationBoswell et al 1997; CitationRubinstein et al 2000; CitationWright et al 2002; CitationNoel et al 2005). Anaerobic conditions also do not alter the AUC/MIC ratios for 50% and 90% of effect for E. coli and moxifloxacin (CitationNoel et al 2005). Mixed effects have been noted when testing for an inoculum effect (CitationBoswell et al 1997; CitationWoodcock et al 1997).

Moxifloxacin has been evaluated in two mouse models relevant to skin and skin structure infections (systemic infection induced by intraperitoneal inoculation and cellulitis induced by subcutaneous (SC) inoculation). In the systemic infection model using 2 strains of methicillin-sensitive S. aureus (both having a moxifloxacin MIC of 0.06 mg/L), the ED50 and ED90 values for SC dosed drug were 0.6 mg/kg, 1.0 mg/kg and 1.7 mg/kg, 2.3 mg/kg, respectively, while corresponding ED values for orally dosed drug were 3.2 mg/kg (for both) and 9.6 mg/kg (for both) (CitationPatel et al 2004). The term ED50/90 refers to the doses effective in producing survival rates of 50% and 90%, respectively. In the mouse cellulitis model, one methicillin-sensitive strain (MIC 0.03mg/L) and two methicillin-resistant strains (MICs 1 mg/L and 4 mg/L) of S. aureus were used. For the methicillin-sensitive strain, 2.5 mg/kg and 5 mg/kg SC doses of moxifloxacin were administered 1 h, 4 h, 24 h, and 27 h post-inoculation while for the methicillin-resistant strains, 25 mg/kg and 50 mg/kg SC doses were administered 1 h, 3 h, and 5 h post-inoculation. With the methicillin-sensitive strain, moxifloxacin proved to be bacteriostatic (even 5 mg/kg doses led to a <2 log colony forming unit (CFU) reduction/lesion). With one methicillin-resistant strain, there was a marginal fall in CFU/lesion while with the other, it had no effect (CFU increased) (CitationPatel et al 2004).

Two recent studies have evaluated the activity of moxifloxacin in animal models of osteomyelitis (bone infection). Bone infection can occur as a result of hematogenous seeding or contiguous spread from an area of soft tissue infection. One study compared moxifloxacin and vancomycin in the treatment of bone infections due to methicillin-sensitive S. aureus in rats. Moxifloxacin (10 mg/kg) and vancomycin (15 mg/kg) were both dosed twice daily by the intraperitoneal route. Moxifloxacin was significantly more effective in reducing bacterial colony counts in femoral bone than were placebo and vancomycin (both p<0.001; placebo vs vancomycin, p=0.53). However, no treatment sterilized the infected bone, probably due to the presence of foreign material (ie, the hollow needle maintained in the cavity) (CitationKalteis et al 2006a). The second study extended the findings of the first to include bacterial counts in infected capsular soft tissue and biofilm adherent to the implanted needle. Moxifloxacin was significantly superior to vancomycin and placebo in reducing bacterial counts in capsular soft tissue (both p<0.001; placebo vs vancomycin, p=0.09) and biofilm (p=0.009 and p<0.001, respectively; placebo vs vancomycin, p=0.20) (CitationKalteis et al 2006b). Further investigation of moxifloxacin in osteomyelitis appears warranted.

Another atypical mycobacterial infection with dermatological manifestations is leprosy (due to M. leprae). The classic test for evaluating the activity of antimicrobials against this microorganism is the mouse footpad model wherein the microorganism is injected into the footpad. In this model, where all drugs were administered orally, moxifloxacin proved superior to the quinolone used most frequently to date in combination regimens for leprosy, ofloxacin (percentages killed of 92.1% vs 60.2%, respectively; p<0.05). In addition, the combination of minocycline plus moxifloxacin was superior to that of minocycline plus ofloxacin (percentages killed of 93.7% vs 74.9%, respectively; p<0.05). Moxifloxacin was equipotent with rifampin, one of the key anti-leprosy drugs (percentage killed by both drugs was 92.1%, p=NS). The most potent combination tested was rifapentine (a rifamycin) plus minocycline plus moxifloxacin which produced a 99.9% killing rate (CitationJi and Grosset 2000). Moxifloxacin is an important agent to evaluate in vivo in human leprosy.

Performing large-scale surveillance of antimicrobial susceptibility can assist in deciding upon the selection of empiric antimicrobial therapy. In one such study, performed between 1994 and 2001, 12 US hospitals collected 4434 Bacteroides isolates for susceptibility testing. The overall moxifloxacin resistance rate rose from 30% to 43% (p<0.001) from 1998 to 2001 (2001 MIC90 = 16 mg/L). Increased moxifloxacin resistance over time was noted in all species, all infection sites, and in 11 of 12 hospitals. For blood isolates, the resistance rate rose from 38% in 1998 to 52% in 2001. Resistance rates were especially high in the species ovatus, uniformis, and vulgatus. The largest increase in resistance occurred with the species distasonis (22% in 1999 to 37% in 2001). Of all infection sites, decubitus ulcer isolates had the highest resistance rates. On multivariate analysis, the association between the year of microorganism isolation and increased resistance rates remained significant after adjusting for hospital, species, and infection site (adjusted odds ratio=1.33, p<0.001) (CitationGolan et al 2003).

In the LIBRA surveillance study in the US in 1999, 324 laboratories participated in an examination of resistance rates in S. pyogenes isolates. Isolates came from the respiratory tract (n=2039), skin and skin structure (n=405) and blood (n=148). The modal moxifloxacin MIC was 0.12 mg/L and the MIC90 was 0.25 mg/L. One hundred percent of isolates were susceptible (based on a resistance breakpoint of ≥4 mg/L) (CitationCritchley et al 2002).

In 2003, 37 laboratories in 19 countries participated in a survey of resistance rates among B. fragilis group isolates. Among the 1284 isolates (the majority coming from abdominal infections and wounds), 9% were resistant to moxifloxacin (breakpoint = 8 mg/L) (range 0%–22%). Moxifloxacin resistance was commonest in Mediterranean Europe (15%) and the UK (22%). In the various infection types, the rates of intermediate (decreased susceptibility) plus resistant isolates were as follows: abdominal infections (5%), wounds (13%), abscesses (4%), blood (11%), and external ulcers/skin infections (16%) (CitationHedberg et al 2003).

In 2005, results of a six-year (1997–2002) surveillance study of moxifloxacin susceptibility among B. fragilis group organisms at a single hospital in Spain were published. Resistance rates to moxifloxacin rose from 6% (1997) to 16.5% (2002), using an MIC breakpoint of 8 mg/L (p<0.005 for trend). Examining resistance rates by species, these rates rose over six years from 1.5% to 12% (B. fragilis), 10.3% to 25% (B. thetaiotaomicron), and 15.4% to 36.8% (B. uniformis) (CitationBetriu et al 2005).

Results of the SENTRY Antimicrobial Surveillance Program (1997–2004) have revealed the emergence in North America and Europe of quinolone resistance among β-hemolytic streptococci (groups A, B, C, G). Although rates are low at present (0.51% and 0.14% in North America and Europe, respectively), further dissemination and expansion of resistance determinants is likely (CitationBiedenbach et al 2006).

Pharmacokinetics

Moxifloxacin is well-absorbed after oral administration, with absolute oral bioavailabilities of 92% (mean) (range 77%–120%) and 86.2 ± 1.11% (mean ± standard deviation [SD]) being noted in 2 healthy-volunteer studies (CitationBallow et al 1999; CitationStass and Kubitza 1999). Oral administration in the fed state (high fat breakfast) had nonsignificant effects on moxifloxacin peak plasma concentration (Cmax) and area under the plasma concentration-versus-time curve (AUC) but did prolong the median time to Cmax (Tmax) from 1 to 2.5 hours (not clinically significant) (CitationLettieri et al 2001). Milk (240–300 mL) and yogurt (250–300 mL) coadministration did not significantly alter moxifloxacin bioavailability (CitationGuay 2005) nor did enteral feeding coadministration (CitationBurkhardt et al 2005). Minor accumulation occurs during multiple dosing and its pharmacokinetics are linear and dose-proportional (CitationStass et al 1998, Citation2001; CitationSullivan et al 1999).

Mean ± SD plasma protein binding values of moxifloxacin were 48% ± 2.5% (CitationStass et al 1998) and 39.4% ± 2.4% (CitationStass and Kubitza 1999). Plasma protein binding of the M1 sulfocompound metabolite was 89.5% ± 0.9% and that of the M2 glucuronide metabolite was 4.8% ± 4.1% (CitationStass and Kubitza 1999). Penetration of moxifloxacin into body compartments relevant to skin and skin structure infections has been the subject of several studies. In one healthy volunteer study, the pharmacokinetics of moxifloxacin in plasma and inflammatory blister fluid (induced by cantharides plasters) were evaluated after single 400 mg oral and intravenous doses. The mean ± SD (range) penetration of moxifloxacin into inflammatory blister fluid after oral and intravenous dosing were 83.5% ± 14.9% (61%–103%) and 93.7% ± 8.3% (81%–104%), respectively, as measured by AUC0–∞ (CitationWise et al 1999). In another study, the serum and synovial fluid concentrations of moxifloxacin after the last of three once-daily oral moxifloxacin 400 mg doses were assessed in 20 patients undergoing knee arthroscopy (mean age of 71.2 years). At 2 h, 6 h, 12 h, and 24 h after the last dose, the mean ± SD serum/synovial fluid concentrations were 2.42 ± 0.4/2.76 ± 0.24 mg/L, 3.46 ± 0.78/3.42 ± 0.51 mg/L, 1.74 ± 0.26/1.61 ± 0.5 mg/L, and 1.5 ± 0.21/1.23 ± 0.14mg/L, respectively (CitationDan et al 2004).

In yet another healthy volunteer study, the penetration of moxifloxacin into the interstitial fluid compartments of muscle and SC adipose fat tissue and inflammatory blister fluid were evaluated after single 400 mg oral and intravenous doses. Mean ± SD penetration into muscle interstitial fluid was 55% ± 12% (using AUCmuscle/AUCtotal plasma data) and 86% ± 17% (using AUCmuscle/AUCfree plasma data data). Corresponding SC adipose fat interstitial fluid penetration was 38% ± 9% and 81% ± 19%. These results suggest a virtually total equilibration between these two body compartments and free (unbound) drug in plasma. The concentration over time profiles in these two compartments were virtually superimposable. Mean ± SD penetration into inflammatory blister fluid (BF) was 64% ± 21% (using AUCBF/AUCtotal plasma data) (CitationMuller et al 1999).

The penetration of moxifloxacin into healthy and inflamed SC adipose fat tissues was assessed in 11 patients with skin and skin structure infections (n=5 had diabetes and peripheral occlusive arterial disease (POAD), n=6 did not). A single 400 mg IV dose of moxifloxacin was administered over one hour. In the aggregate analysis (n=11), drug concentrations in healthy and inflamed tissues were consistently below total plasma concentrations and approximately equal to free plasma concentrations. Moxifloxacin total plasma concentration over time profiles were similar in patients with and without diabetes/POAD. Penetration into tissue did not statistically differ in the two groups. In patients with diabetes/POAD, the concentrations in healthy tissue exceeded those in inflamed tissue. Only the healthy tissue concentrations were similar to the free concentrations in plasma (the inflamed tissue concentrations were about 1/2 of the free concentrations in plasma). In the patients without diabetes, exactly the opposite findings were noted. Thus, the mean ± SD AUC0–8h for inflamed tissue/AUC0−∞ for free drug in plasma was 0.5 ± 0.4 in patients with diabetes/POAD and 1.2 ± 0.8 in the other group. Mean ± SD inflamed tissue Cmax values were 0.8 ± 0.5 mg/L in patients with diabetes/POAD and 2.3 ± 1.2 mg/L in the other group. The reduced concentrations in inflamed tissues in patients with versus without diabetes/POAD was perhaps due to the vascular compromise associated with POAD. None of these differences between patients with/without diabetes/POAD were significant due to small sample sizes and large interpatient variability. In all cases, tissue and plasma drug concentrations exceeded the MICs of most dermatologic pathogens, suggesting a role for moxifloxacin in the treatment of skin and skin structure infections (CitationJoukhadar et al 2003).

Thirty patients undergoing total knee arthroplasty participated in a study investigating the penetration of moxifloxacin into bone. Ten patients received oral moxifloxacin 2 h (range 1.5–2.5 h) preoperatively and mean plasma, cancellous bone, and cortical bone drug concentrations were 3.45 mg/L, 1.89 mg/kg, and 1.43 mg/kg, respectively. In the ten patients receiving moxifloxacin 4 h (range 3.5–4.5 h) preoperatively, corresponding mean drug concentrations were 3.73 mg/L, 1.81 mg/kg, and 1.56 mg/kg. In the ten patients receiving the drug 14 h and 2 h (range 1.5–2.5 h) preoperatively, corresponding drug concentrations were 6.26 mg/L, 2.97 mg/kg, and 2.54 mg/kg (CitationMalincarne et al 2006).

As with other quinolones, moxifloxacin penetrates well into polymorphonuclear leukocytes (mean ± SD internal concentration = 54 ± 0.5 mg/L with an external medium concentration of 4.5 mg/L) (CitationMandell and Coleman 2001). Moxifloxacin also penetrates well into macrophages (with an external concentration of 4 mg/L, the mean internal–external concentration ratio = 13.4 (in cells infected with S. aureus) and 11.4 (in non-infected cells), p=0.02) (CitationSeral et al 2003).

Moxifloxacin elimination occurs via a combination of hepatic metabolism, biliary secretion, renal excretion, and perhaps, transintestinal secretion. It is metabolized to 2 inactive metabolites, M1 (N-sulfate) and M2 (acylglucuronide) (CitationStass et al 2004). Renal clearance (CLR) of the parent compound involves glomerular filtration and partial renal tubular reabsorption (CitationStass et al 1998; CitationStass and Kubitza 1999). Fecal drug concentrations upon multiple oral dosing are quite high, a result of nonabsorption, biliary secretion, and, possibly, transintestinal secretion (mean ± SD days 2, 4, and 7 values during 400 mg daily oral dosing were 87.3 ± 2.4 mcg/g, 303.3 ± 1.8 mcg/g, and 573.3 ± 1.9 mcg/g, respectively) (CitationBurkhardt et al 2002). Mass balance studies using oral drug have demonstrated that (mean ± SD) 44.8% ± 3.4%, 37.9% ± 3.6%, and 13.6% ± 2.8% of the dose is eliminated as moxifloxacin (19.4% ± 1.2% in urine and 25.4% ± 3.1% in feces), M1 metabolite (2.5% ± 0.6% in urine and 35.5% ± 3.2% in feces) and M2 metabolite (13.6% ± 2.8% in urine and none in feces), respectively (CitationStass and Kubitza 1999). Thus, the urinary and fecal routes account for 35.4% ± 1.8% and 60.9% ± 5.1% of the dose, respectively (CitationStass and Kubitza 1999). The M1 metabolite is a high clearance compound eliminated mainly via hepatic metabolism and biliary secretion into the feces. Renal secretion occurs via a net active tubular secretion process (CitationStass and Kubitza 1999). The M2 metabolite is the main metabolite seen in plasma, undergoing some presystemic clearance via glucuronidation and excretion by the kidneys by active tubular secretion. Biliary/fecal elimination is negligible (CitationStass and Kubitza 1999). A summary of the pharmacokinetic parameters for moxifloxacin in healthy volunteers is provided in (CitationStass et al 1998, Citation2001, Citation2002, Citation2004; CitationBallow et al 1999; CitationStass and Kubitza 1999; CitationSullivan et al 1999, Citation2001; CitationWise et al 1999; CitationLubasch et al 2000; CitationLettieri et al 2001; CitationBurkhardt et al 2002).

Table 3 Mean ± SD pharmacokinetic parameters of moxifloxacin in healthy adult volunteers

When pharmacokinetic parameters are weight-normalized, there are no significant age- or gender-based effects on moxifloxacin pharmacokinetics (CitationSullivan et al 2001; CitationPea et al 2006). Thirty-two individuals participated in a single 400 mg oral dose study of the effects of renal impairment on the pharmacokinetics of moxifloxacin and its M1 and M2 metabolites. With moxifloxacin, there was no significant relationship of AUC with creatinine clearance (CrCl) (p=0.59). However, Cmax did fall as CrCl fell (p=0.03). Significant correlations existed between moxifloxcin Cmax (r=0.438, p=0.012), fractional renal excretion (Fe) (r=0.613, p=0.0002) and CLR (r=0.6405, p=0.0001) and CrCl. The M1 metabolite was little affected by changes in renal function although significant correlations existed between M1 metabolite Fe (r=0.620, p=0.0002) and CLR (r=0.703, p<0.0001) and CrCl. For the M2 metabolite, AUC rose as CrCl fell (p=0.0167) while max did not change (p=0.3042). Significant correlations existed between M2 metabolite AUC (r=−0.565, p=0.0011), CLR (r=0.736, p<0.0001) and Fe (r=0.514, p=0.0026) and CrCl (CitationStass et al 2002).

Single and multiple dose pharmacokinetics of moxifloxacin have also been evaluated in end-stage renal disease patients undergoing hemodialysis (HD) and continuous ambulatory peritoneal dialysis (CAPD). Following a single oral 400 mg dose, the AUC’s of parent compound in these patients were statistically similar to those found generally in healthy volunteers. However, Cmax values were reduced by about 45% (HD) and 33% (CAPD) compared with historical healthy control subject data. The systemic exposure to the M1 metabolite rose by 1.4- to 1.5-fold (based on AUC) while that to the M2 metabolite rose 7.5-fold (based on AUC) and 2.5- to 3-fold (based on Cmax) compared with historical healthy control subject data. The possible consequences of increased metabolite exposure are not known. Multiple once-daily oral dosing of 400 mg for 7 days in these patients produced systemic exposure (AUC) similar to that seen in healthy volunteers. Steady-state Cmax values in HD patients were approximately 22% lower than those in healthy volunteers while they were comparable for CAPD patients. Dialysis removal of moxifloxacin was low (9% by HD, 3% by CAPD) (CitationAnonymous 2005). Continuous renal replacement therapy (continuous venovenous hemodiafiltration) does not affect moxifloxacin disposition to a clinically-meaningful extent (CitationFuhrmann et al 2004).

Clinical efficacy

This section will deal only with the clinical efficacy of moxifloxacin in skin and skin structure infections (SSSIs). illustrates the results of moxifloxacin clinical trials in uncomplicated and complicated SSSIs (CitationLeal del Rosal, Fabian, et al 1999; CitationLeal del Rosal, Martinez, et al 1999; CitationParish et al 2000; CitationAnonymous 2005; CitationGiordano et al 2005). All have been active-controlled in design, with cephalexin as the control in the three uncomplicated SSSI studies, unspecified β-lactam/β-lactamase-inhibitor combinations in the 2 unpublished complicated SSSI studies, and piperacillin-tazobactam followed by amoxicillin-clavulanate as the control in the one published complicated SSSI study. As may be seen, there were no significant or substantial inter-group differences in clinical or bacteriological efficacy or tolerability.

Table 4 Clinical efficacy and tolerability of moxifloxacin in skin and skin structure infections (SSSIs)

These results should be interpreted while keeping in mind potential study limitations. In general, these studies did not provide evidence of power analyses conducted to establish proper group sizes. Thus, they were underpowered to avoid type II statistical errors (ie, false negative intergroup differences), especially with respect to subgroup analyses based on comorbidities or individual pathogens. Three of five studies remain unpublished, available only as abstracts or in the product information. The limitations of this should be obvious to the reader. The extensive number and breadth of exclusion criteria generated study populations much different from the “real world” patients who would require treatment. Hence, the generalizability of study results can be questioned. Clinical trials of SSSIs have long been difficult to design in a way that renders them rigorous and reproducible. Although the publication of SSSI study design criteria by the Infectious Diseases Society of America (IDSA) have helped in this regard (Calandra et al 1992), deficient study designs are, unfortunately, still far too frequently seen. The ability of investigators to add anti-anaerobic coverage (eg, metronidazole) for presumed mixed SSSIs makes it impossible to judge whether or not the anaerobic coverage of moxifloxacin is clinically-relevant. Lastly, the definitions of clinical endpoints can vary between SSSI studies, making it difficult to obtain a global perspective of drug efficacy.

A recent case report detailed a case of a patient with an infected cat-bite wound failing therapy with IV cefuroxime, ciprofloxacin, and metronidazole which responded to the addition of IV moxifloxacin (CitationDraenert et al 2005). Another case report documented successful therapy of a M. thermoresistibile infection following knee replacement surgery with long-term therapy using moxifloxacin and doxycycline (CitationLabombardi et al 2005).

A recent review of potential antitubercular drugs included encouraging in vitro, in vivo, (animal) and pilot human clinical data regarding moxifloxacin use in infections due to M. tuberculosis (CitationDuncan and Barry 2004). These data, in conjunction with in vitro susceptibility data reviewed earlier in this paper, suggest that moxifloxacin may play an important role in the treatment of SSSIs caused by atypical mycobacteria. Clinical trials in this area are warranted.

Safety

As with other quinolones, moxifloxacin is generally well-tolerated, with the majority of adverse events referable to the gastrointestinal tract (nausea, diarrhea, vomiting, constipation, abdominal pain, vaginitis, taste alteration) or the central nervous system (headache, dizziness) (CitationAnonymous 2005). There were only three adverse events, judged by investigators to be at least possibly drug-related, occurring in at least 2% of over 8600 moxifloxacin-treated patients in the drug development process: nausea (6%), diarrhea (5%), and dizziness (2%) (CitationAnonymous 2005). Premature study discontinuation rates for drug-related adverse events were 2.9% (oral) and 4.6% (IV → oral) (CitationAnonymous 2005). Moxifloxacin is not associated with the phototoxicity characteristic of lomefloxacin and sparfloxacin (CitationMan et al 1999) and has only been associated with the “temafloxacin hypersensitivity syndrome” in 1 patient (CitationNori et al 2004). This patient exhibited toxic epidermal necrolysis and fulminant hepatic failure leading to death. Moxifloxacin is not associated with the hypo or hyperglycemia seen with gatifloxacin therapy (CitationPark-Wyllie et al 2006).

In 2004, a cumulative safety review of oral and intravenous moxifloxacin from the drug development program and postmarketing surveillance studies was published. For the oral agent, the database comprised 30 phase II/III active comparator studies (moxifloxacin n=7368, comparators n=5687), one phase IV study (n=18374), and 4 postmarketing observation studies (n=27756). For the intravenous agent, the database comprised two phase III active comparator studies (moxifloxacin n=550, comparators n=579). With both formulations, the commonest adverse events involved the gastrointestinal tract (for the oral agent, nausea in 7.1% and diarrhea in 5.2%; for the intravenous agent, nausea in 3.1% and diarrhea in 6.2%). Premature discontinuation rates due to adverse events to moxifloxacin were 2.7% (oral) and 6.0% (intravenous) while mortality rates were 2.7% (oral) and 4% (intravenous). Cardiovascular adverse events, including surrogate markers for arrhythmias such as tachycardia, palpitations, syncope, and hypotension, occurred at rates far below 1%. Blood sugar-related events were practically nonexistent (CitationBall et al 2004).

In the same year, a similar retrospective review of the phase II/III clinical trial database (32 trials, n=14731 patients (moxifloxacin n=8474, active comparators n=6257)) and postmarketing surveillance studies (n=46130) were reviewed with a focus on adverse events involving glucose metabolism. In the clinical trials, there were 0 and 3 drug-related hypoglycemic adverse events with moxifloxacin and active comparator patients, respectively (latter were 2 cases with levofloxacin and 1 case with trovafloxacin). Drug-related hypoglycemic events occurred in 7 moxifloxacin and 1 active comparator patients. No evidence was found supporting the presence of drug–drug interactions with oral antidiabetic drugs. In the postmarketing studies, no cases of drug-related hypoglycemia were noted. This same paper described results of in vivo (rat) studies utilizing single 30 mg/kg and 100 mg/kg oral doses of moxifloxacin, gatifloxacin, and levofloxacin (positive control was glibenclamide 10 mg/kg). Blood was sampled predosing and at 0.5 h and 1.5 h postdosing. In fasted rates, glibenclamide had the expected effect (blood glucose fell a mean of 24% at 1.5 h postdose which was preceded by a rise in serum insulin). Blood glucose was not significantly affected by either moxifloxacin or levofloxacin but fell means of 8% and 18% with 30 mg/kg and 100 mg/kg doses of gatifloxacin, respectively. No significant effects on serum insulin were noted with any quinolone. In fed rats, glibenclamide increased serum insulin and reduced blood glucose approximately 40% at 1.5 h postdosing. Again, moxifloxacin and levofloxacin had no significant effect on blood glucose or serum insulin. Gatifloxacin 30 mg/kg and 100 mg/kg reduced blood glucose by means of 17% and 26%, respectively. This was preceded by a reduction in serum insulin (only significant at 0.5 h with 100 mg/kg dose). Thus, the gatifloxacin effect was felt to be insulin-independent (CitationGavin et al 2004).

Results of the last of the large retrospective database reviews for safety purposes were published in 2005 and focused on the safety of oral moxifloxacin in the clinical trial database as a function of aging. The database involved 27 phase II/III clinical trials analyzed by age grouping (<65, 65–74, ≥75 years old) and treatment (moxifloxacin and active comparators). Valid data were available from 12231 subjects (moxifloxacin n=6270, active comparators n=5961; comparators were primarily clarithromycin and cefuroxime axetil). The subject distribution by age was as follows: n=9671 were <65, n=1631 were 65–74, and n=924 were ≥75 years old. The distribution of drug-related adverse events comparing moxifloxacin with active comparators was not affected by increasing age (p=0.43) nor were premature discontinuations due to adverse events (p=0.552). The number of deaths were also similar (moxifloxacin n=17, active comparator n=19). No arrhythmias due to QTc interval prolongation were noted in the entire population. The differences in adverse event profiles across the 3 age groups were not significant for moxifloxacin (p=0.599) with 1 exception. For drug-related adverse events with moxifloxacin, the profiles did differ across the 3 age groups (p=0.001) but, unexpectedly most rates increased as age fell (eg, nausea, vomiting, dyspepsia, and vaginal moniliasis rates rose as age fell but insomnia rates rose as age rose). There were no clinically-significant differences between the treatments in the prevalence of serious drug-related events (≤1%) and these were not related to age. In 787 patients who had pre and 3 day-post 12-lead electrocardiograms (n=651 were <65, n=136 were ≥65 years old), the mean ± SD QTC interval prolongation was 7 ± 25msec in the <65 group and 2 ± 28 msec in the ≥65 group (p=0.055, trend) (CitationAndriole et al 2005).

Pharmacokinetic drug–drug interactions

Based on the significant role of metabolism in the elimination of moxifloxacin, a study was conducted to evaluate the effect of concurrent use of a potent cytochrome P450 isozyme 3A4 inhibitor (itraconazole) on moxifloxacin pharmacokinetics. Nine days of itraconazole 200 mg orally daily had no significant effect on the single-dose kinetics of oral moxifloxacin 400 mg (administered on day 7 of the 9-day itraconazole regimen). Moxifloxacin also had no significant effect on itraconazole kinetics (CitationStass et al 2004). Moxifloxacin 400 mg orally daily had no significant effect on theophylline total body clearance and terminal disposition half-life in 2 studies (CitationGuay 2005). Moxifloxacin 400 mg orally once daily had no significant effect on the pharmacokinetics or pharmacodynamics of warfarin in healthy volunteers (CitationGuay 2005). However, case reports have documented moxifloxacin-associated increases in INR (international normalized ratio of prothrombin time) in warfarin recipients (CitationElbe and Chang 2005; CitationGuay 2005). Pending additional information, patients receiving long-term warfarin therapy in whom moxifloxacin is to be used should be monitored for changes in INR. Moxifloxacin does not significantly affect low-dose oral contraceptive or digoxin pharmacokinetics (CitationGuay 2005). Unlike the case with ciprofloxacin, norfloxacin, levofloxacin, and gatifloxacin, probenecid does not significantly alter the systemic pharmacokinetics of moxifloxacin (CitationGuay 2005).

Like all quinolones, moxifloxacin is susceptible to interactions with multivalent cations, wherein the reductions in bioavailability may be clinically-relevant and lead to a loss of therapeutic benefit. For example, calcium carbonate or calcium lactate-gluconate given immediately before and 12 h and 24 h after single-dose moxifloxacin resulted in a mean 15% reduction in Cmax (p≤0.05) and a mean 2% reduction in AUC (p=NS) (CitationGuay 2005). Simultaneous administration of single-dose ferrous sulfate lead to mean reductions of 59 and 39% in single-dose moxifloxacin Cmax and AUC, respectively (both, p≤0.05) (CitationGuay 2005). Single-dose magnesium-aluminum antacid administered simultaneously with single-dose moxifloxacin resulted in mean 61% and 59% reductions in Cmax and AUC, respectively (both, p≤0.05). When the antacid administration was delayed until 2 h after moxifloxacin administration, the mean 7% reduction in Cmax was non-significant but the mean 26% reduction in AUC was significant (p≤0.05). Even administering the antacid 4 h before moxifloxacin administration still resulted in a significant reduction in quinolone AUC (mean 23%, p≤0.05) although the mean 1% fall in Cmax was not (CitationGuay 2005). Sucralfate administered simultaneously with and then 5 h, 10 h, 15 h, and 24 h following single-dose moxifloxacin administration resulted in mean 71% and 60% reductions in quinolone Cmax and AUC, respectively (both, p≤0.05) (CitationGuay 2005). Ranitidine, a histamine-2 receptor antagonist, does not interact with moxifloxacin (2 studies) and can be recommended as a noninteracting antiulcer therapy alternative to sucralfate and antacids (CitationGuay 2005).

Pharmacodynamic drug–drug interactions

The only potentially clinically-relevant pharmacodynamic drug–drug interaction with moxifloxacin involves additive electrophysiological effects with other drugs that can also prolong the QTc interval on the electrocardiogram () (CitationGuay 2005). Drugs which prolong cardiac repolarization may, on rare occasions, be associated with the development of polymorphous ventricular tachycardia (“torsades de pointes”) which, in turn, may degenerate into ventricular fibrillation. Quinolones cause a drug-specific, dose-dependent prolongation of QTc interval by inhibiting outward potassium currents in myocytes. In numerous in vitro models examining the mechanism(s) underlying this arrhythmogenic effect, sparfloxacin has been more potent than moxifloxacin (= grepafloxacin) which, in turn, has been more potent than the other tested quinolones (CitationGuay 2005). In in vivo (animal) models, sparfloxacin again was most potent followed by the trio of moxifloxacin–gatifloxacin– grepafloxacin (latter agent has been withdrawn from the market) (CitationGuay 2005). Concentration-response relationships have been demonstrated in both in vitro and in vivo preclinical models (CitationChen et al 2005).

Table 5 Drugs prolonging the QTcinterval that may potentially interact pharmacodynamically with moxifloxacin. Modified from CitationGuay 2005

In one healthy volunteer study, single 400 mg and 800 mg oral doses of moxifloxacin caused mean ± SD 4.0% ± 5.1% and 4.5% ± 3.8% QTc interval prolongation at rest compared with baseline, respectively (both, p≤0.05). Significant QTc interval prolongation occurred at all heart rates and across the entire RR interval range (400–1000 msec). There were no gender-dependent differences in results. The correlation of moxifloxacin plasma concentrations and QT interval was significant but weak (r=0.35) (CitationGuay 2005).

Only two comparative studies of the effect of quinolones on QTc intervals have been published. In the first, single oral doses of levofloxacin (1000 mg), ciprofloxacin (1500 mg) and moxifloxacin (800 mg) were evaluated in a placebo-controlled, crossover trial in healthy volunteers. Mean QT and QTc interval prolongation was significantly greater for moxifloxacin compared with placebo for all end points, but it was generally not for the other two quinolones. The proportion of subjects with QTc interval prolongation of 30 msec. or higher was greater with moxifloxacin (72%– 81%) compared with levofloxacin (33%–38%) and ciprofloxacin (34%–40%) (CitationGuay 2005). In the second, 13 healthy volunteers received three 7-day regimens in random order: ciprofloxacin 500 mg twice daily, levofloxacin 500 mg once daily, and moxifloxacin 400 mg once daily. Washout periods between treatments were one week in length. Twelve-lead electrocardiograms were obtained before, 2 h after the first dose, and at the end of each regimen. At the end of 7 days, moxifloxacin prolonged QTc by a mean of 6 msec compared with baseline (p=0.022) and a mean of 11 msec from the 2 h baseline value (p=0.003). Ciprofloxacin and levofloxacin had no significant effect on QTc. No quinolone had a significant effect on QT dispersion, a parameter felt to be a more selective marker for risk of polymorphous ventricular tachycardia (CitationTsikouris et al 2006). Further studies of QT dispersion, especially in patients receiving quinolone therapy, with/without concurrent use of other drugs which also prolong QTc, are warranted.

The lack of multiple dose studies and well-done epidemiologic studies do not allow an assessment of the clinical relevance of the above-cited findings. In a recent randomized trial comparing the cardiac safety of IV/oral moxifloxacin and levofloxacin in elderly patients hospitalized for community-acquired pneumonia, the two agents had similar cardiac rhythm safety profiles (CitationMorganroth et al 2005). However, it is prudent to use caution when using moxifloxacin (and other quinolones) in patients already receiving potentially arrhythmogenic drugs () (CitationGuay 2005). Similarly, caution is warranted when use is contemplated in patients with abnormal pretreatment QTc intervals, electrolyte abnormalities (especially hypokalemia, hypomagnesemia, rarely hypocalcemia), starvation/liquid protein fast diets, and prior/current history of coronary artery disease or arrhythmias.

Dosing

Moxifloxacin is available as 400 mg oral tablets and premixed moxifloxacin 400 mg in 250 mL 0.8% sodium chloride IV bags (CitationAnonymous 2005). For SSSIs, the recommended daily dose is 400 mg and the recommended durations of therapy are 7 days (uncomplicated infections) and 7–21 days (complicated infections). Oral doses of moxifloxacin should be administered at least 4 h before or 8 h after magnesium/aluminum antacids, sucralfate, metal cations (eg, iron), multivitamin + mineral supplements (especially with zinc), and didenosine chewable/buffered tablets or pediatric powder for solution (CitationAnonymous 2005). No dosage adjustment is needed with any level of renal impairment or mild or moderate (Child-Pugh Class A or B) hepatic impairment (CitationAnonymous 2005). In severe (Child-Pugh Class C) hepatic impairment, there are no data to guide the clinician (CitationAnonymous 2005).

Conclusions

Optimal therapy for SSSIs requires careful patient evaluation, including obtaining a thorough medical and social history, performing a meticulous examination of the lesion(s), and examination of Gram-stained smears of lesion exudates. Early empirical antimicrobial therapy is the next step after obtaining culture specimens, with subsequent tailoring of therapy based on culture and sensitivity results. Local measures, including debridement, are important adjuncts to antimicrobial therapy. Selection of empiric therapy for SSSIs depends on a number of factors, including type of infection (uncomplicated vs complicated), location of acquisition (community versus hospital), and local sensitivity patterns for the commonest dermatologic pathogens.

Older quinolones have been demonstrated to be efficacious in the treatment of uncomplicated and complicated SSSIs over the past 20 years (CitationBlondeau 2002; CitationSable and Murakawa 2003). In addition, more limited data support the efficacy of the newer quinolones in the treatment of uncomplicated SSSIs (gatifloxacin, levofloxacin 500 mg/d, and moxifloxacin) (CitationLeal del Rosal, Fabian, et al 1999; CitationLeal de Rosal, Martinez, et al 1999; CitationParish et al 2000; CitationRaghavan and Linden 2004). To these data may be added those documenting the efficacy of high-dose levofloxacin (750 mg/d) and moxifloxacin in the treatment of complicated SSSIs (CitationRaghavan and Linden 2004; CitationAnonymous 2005).

What have other reviews recently said regarding the role of quinolones in the treatment of SSSIs? In a recent review of the diagnosis and treatment of facial bite wounds, moxifloxacin was felt to be a reasonable monotherapy for infected human facial bite wounds, especially in patients who are penicillin-allergic. The suggested duration of a prophylactic regimen ranged from 3–5 days (10–14 days if bone was involved). For treatment, if bones or joints were involved, a duration of 21+ days was recommended (CitationStefanopoulos and Tarantzopoulou 2005).

In a review of the role of quinolones in the treatment of SSSIs, Blondeau felt that both older and newer quinolones had roles to play. The older ciprofloxacin-type quinolones were recommended only in SSSIs where Gram-negative aerobic bacilli were known pathogens, adding clindamycin or metronidazole in suspected mixed infections. The newer quinolones (including moxifloxacin) were recommended in SSSIs where non-pseudomonal Gram-negative aerobic bacilli were known pathogens, where there was a mixed infection with anaerobes within the quinolone’s spectrum of activity, where non-methicillin-resistant S. aureus gram-positive aerobes were involved, and where Gram-positive and Gram-negative aerobic pathogens were coexisting (CitationBlondeau 2002).

The first organization-based guidelines for the diagnosis and management of SSSIs were published by IDSA in November 2005 (CitationStevens et al 2005). Quinolones are mentioned in the context of several general and organism-specific infections. Quinolones are one of many empirical choices in human and animal bite wounds, especially in patients with severe penicillin allergies. Ciprofloxacin, gatifloxacin, and moxifloxacin with/without clindamycin or metronidazole are those specifically mentioned. Again, quinolones are one of many empirical choices in SSSIs in the immunocompromised host. A quinolone (probably ciprofloxacin) plus an extended-spectrum β-lactam is the specific quinolone mentioned. Quinolones are one of several choices for the empiric management of each of three defined types of surgical wound infections. In community-acquired mixed necrotizing fasciitis, one option is a combination of ciprofloxacin plus a β-lactam-β-lactamase inhibitor. In SSSIs due to community-acquired MRSA, the newer quinolone group (including moxifloxacin) is one option (author’s comment: based on 20 years of experience with quinolones in infections due to MRSA, the likelihood of the development of resistance to quinolones during such use is quite high. caution is warranted). Lastly, in infected wounds after intestinal or genital tract surgeries, a combination of a quinolone plus one of clindamycin/metronidazole/chloramphenicol/β-lactam-β-lactamase inhibitor is one option. Quinolones are mentioned in the context of the patient with severe penicllin allergies. In cutaneous anthrax, ciprofloxacin is the drug-of-choice (newer quinolones are also considered likely effective). In erysipeloid, quinolones are options only in patients with severe penicillin allergies. Lastly, among many options, levofloxacin and ciprofloxacin are considered options for tularemia but only in mild to moderate illness.

Evolving resistance trends, especially amongst staphylococci and enterococci, and sporadic intolerance to conventional β-lactam therapy have led to the need for novel options with proven efficacy and tolerability. Moxifloxacin is comparable in both efficacy and tolerability with conventional agents for SSSIs and is also effective against many organisms resistant to these agents (exception: methicillin-resistant staphylococci). It also possesses activity against atypical mycobacteria associated with SSSIs (moxifloxacin may develop into a first-line therapy for these types of SSSIs but much work remains to be done before this can be recommended). Despite its proven activity against dermatologic pathogens, one could argue that its use be carefully considered and, in general, restricted to serious, life-threatening infections caused by resistant pathogens or those patients with serious intolerances to conventional therapy. In this way, the development and progression of resistance might be blunted and its “lifespan” as an antimicrobial for dermatologic infections prolonged.

References

  • AckermannGSchaumannRPlessBComparative activity of moxifloxacin in vitro against obligately anerobic bacteriaEur J Clin Microbiol Infect Dis2000192283210795599
  • AlcaideFCalatayudLSantinMComparative in vitro activities of linezolid, telithromycin, clarithromycin, levofloxacin, moxifloxacin, and four conventional antimycobacterial drugs against Mycobacterium kansasiiAntimicrob Agents Chemother2004484562515561826
  • AldridgeKEAshcraftDSComparison of the in vitro activities of Bay 12–8039, a new quinolone, and other antimicrobials against clinically important anaerobesAntimicrob Agents Chemother199741709119056020
  • Amabile-CuevasCFHermida-EscobedoCVivarRComparative in vitro activity of moxifloxacin by E-test against Streptococcus pyogenesClin Infect Dis200132Suppl 1S30211249826
  • AndrioleVTHaverstockDCChoudhriSHRetrospective analysis of the safety profile of oral moxifloxacin in elderly patients enrolled in clinical trialsDrug Safety2005284435215853445
  • AnonymousAvelox® (moxifloxacin) product information2005West Haven, CTBayer HealthCare June 2005
  • AubryAJarlierVEscolanoSAntibiotic susceptibility pattern of Mycobacterium marinumAntimicrob Agents Chemother2000443133611036036
  • BallPStahlmannRKubinRSafety profile of oral and intravenous moxifloxacin: cumulative data from clinical trials and postmarketing studiesClin Ther2004269405015336463
  • BallowCLettieriJAgarwalVAbsolute bioavailability of moxifloxacinClin Ther1999215132210321420
  • BarryALFuchsPCBrownSDAntibacterial activity of moxifloxacin (Bay 12–8039) against aerobic clinical isolates, and provisional criteria for disk susceptibility testsEur J Clin Microbiol Infect Dis199918305910385024
  • BauernfeindAComparison of the antibacterial activities of the quinolones Bay 12–8039, gatifloxacin (AM 1155), trovafloxacin, clinafloxacin, levofloxacin and ciprofloxacinJ Antimicrob Chemother199740639519421311
  • BetriuCGomezMPalauMLActivities of new antimicrobial agents (trovafloxacin, moxifloxacin, sanfetrinem, and quinupristin-dalfopristin) against Bacteroides fragilis group: comparison with the activities of 14 other agentsAntimicrob Agents Chemother1999432320210471590
  • BetriuCRodriguez-AvialIGomezMChanging patterns of fluoroquinolone resistance among Bacteroides fragilis group organisms over a 6-year period (1997–2002)Diagn Microbiol Infect Dis200553221316243476
  • BiedenbachDJTolemanMAWalshTRCharacterization of fluoroquinolone-resistant β-hemolytic Streptococcus spp. isolated in North America and Europe including the first report of fluoroquinolone-resistant Streptococcus dysgalactiae subspecies equisimilis: report from the SENTRY Antimicrobiol Surveillance Program (1997–2004)Diagn Microbiol Infect Dis2006551192716530373
  • BlondeauJMLaskowskiRBjarnasonJComparative in vitro activity of gatifloxacin, grepafloxacin, levofloxacin, moxifloxacin and trovafloxacin against 4151 gram-negative and gram-positive organismsInt J Antimicrob Agents200014455010717500
  • BlondeauJMThe role of fluoroquinolones in skin and skin structure infectionsAm J Clin Dermatol20023374611817967
  • BogdanovichTEselDKellyLMAntistaphylococcal activity of DX-169, a new des-F(6)-quinolone, compared with those of other agentsAntimicrob Agents Chemother20054933253316048943
  • BoswellFJAndrewsJMWiseRPharmacodynamic properties of Bay 12-8039 on gram-positive and gram-negative organisms as demonstrated by studies of time-kill kinetics and postantibiotic effectAntimicrob Agents Chemother199741137799174203
  • BowkerKENoelARMacGowanAPIn vitro activities of nine peptide deformylase inhibitors and five comparator agents against respiratory and skin pathogensInt J Antimicrob Agents2003225576114659651
  • BrabackMRiesbeckKForsgrenASusceptibilities of Mycobacterium marinum to gatifloxacin, gemifloxacin, levofloxacin, linezolid, moxifloxacin, telithromycin, and quinupristin-dalfopristin (Synercid) compared with its susceptibilities to reference macrolides and quinolonesAntimicrob Agents Chemother20024611141611897601
  • BurkhardtOBornerKStassHSingle- and multiple-dose pharmacokinetics of oral moxifloxacin and clarithromycin, and concentrations in serum, saliva and faecesScand J Infect Dis20023489890312587622
  • BurkhardtOStassHThussUEffects of enteral feeding on the oral bioavailability of moxifloxacin in healthy volunteersClin Pharmacokinet2005449697616122283
  • ChenXCassJDBradleyJAQT prolongation and proarrhythmia by moxifloxacin: concordance of preclinical models in relation to clinical outcomeBr J Pharmacol2005146792916158069
  • CreditoKLJacobsMRAppelbaumPCTime-kill studies of the antianaerobe activity of garenoxacin compared with those of nine other agentsAntimicrob Agents Chemother200347139940212654677
  • CritchleyIASahmDFThornsberryCAntimicrobial susceptibilities of Streptococcus pyogenes isolated from respiratory and skin and soft tissue infections: United States LIBRA surveillance data from 1999Diagn Microbiol Infect Dis2002421293511858909
  • DalhoffAPetersenUEndermannRIn vitro activity of Bay 12-8039, a new 8-methoxyquinoloneChemotherapy199642410258957575
  • DalhoffASchubertSUllmannUEffect of pH on the in vitro activity of and propensity for the emergence of resistance to fluoroquinolones, macrolides, and a ketolideInfection200533suppl 2364316518710
  • DanMKeynanOFeldbrinZConcentrations of moxifloxacin in serum and synovial fluid, and ex vivo bactericidal activity against arthritis-causing pathogensDiagn Microbiol Infect Dis200448283615062922
  • DraenertRKunzelmannMRoggenkampAInfected cat-bite wound treated successfully with moxifloxacin after failure of parenteral cefuroxime and ciprofloxacinEur J Clin Microbiol Infect Dis2005242889015902538
  • DuncanKBarryCE3rdProspects for new antitubercular drugsCurr Opin Microbiol20047460515451500
  • EdlundCSabouriSNordCEComparative in vitro activity of BAY 12-8039 and five other antimicrobial agents against anaerobic bacteriaEur J Clin Microbiol Infect Dis19981719359665302
  • EdmistonCEKrepelCJSeabrookGRIn vitro activities of moxifloxacin against 900 aerobic and anaerobic surgical isolates from patients with intra-abdominal and diabetic foot infectionsAntimicrob Agents Chemother20044810121614982797
  • EdmistonCEKrepelCJKehlKSComparative in vitro antimicrobial activity of a novel quinolone, garenoxacin, against aerobic and anaerobic microbial isolates recovered from general, vascular, cardiothoracic, and otolaryngologic surgical patientsJ Antimicrob Chemother200556872816186167
  • EdnieLMRattanAJacobsMRAntianaerobe activity of RBX 7644 (ranbezolid), a new oxazolidinone, compared with those of eight other agentsAntimicrob Agents Chemother2003471143712604558
  • ElbeDHChangSWMoxifloxacin-warfarin interaction: a series of five case reportsAnn Pharmacother200539361415632222
  • EspositoSNovielloSIannielloFBactericidal activity of moxifloxacin compared with grepafloxacin and clarithromycin against Streptococcus pneumoniae and Streptococcus pyogenes investigated using an in vitro pharmacodynamic modelJ Chemother2000124758111154028
  • FassRJIn vitro activity of Bay 12–8039, a new 8-methoxyquinoloneAntimicrob Agents Chemother1997411818249257770
  • FuhrmannVSchenkPJaegerWPharmacokinetics of moxifloxacin in patients undergoing continuous venovenous hemodiafiltrationJ Antimicrob Chemother200454780415347636
  • Fung-TomcJCMinassianBKolekBAntibacterial spectrum of a novel des-fluoro(6) quinolone, BMS-284756Antimicrob Agents Chemother2000443351611083639
  • GavinJR3rdKubinRChoudhriSMoxifloxacin and glucose homeostasis: a pooled-analysis of the evidence from clinical and postmarketing studiesDrug Safety2004276718615230648
  • GillespieSHBillingtonOActivity of moxifloxacin against mycobacteriaJ Antimicrob Chemother199944393510511409
  • GiordanoPSongJPentelPSequential intravenous/oral moxifloxacin versus intravenous piperacillin-tazobactam followed by oral amoxicillin-clavulanate for the treatment of complicated skin and skin structure infectionInt J Antimicrob Agents2005263576516229991
  • GolanYMcDermottLAJacobusNVEmergence of fluoroquinolone resistance among Bacteroides speciesJ Antimicrob Chemother2003522081312865399
  • GoldsteinEJCitronDMHudspethMIn vitro activity of Bay 12–8039, a new 8-methoxyquinolone, compared with the activities of 11 other oral antimicrobial agents against 390 aerobic and anaerobic bacteria isolated from human and animal bite wound skin and soft tissue infections in humansAntimicrob Agents Chemother199741155279210683
  • GoldsteinEJCitronDMMerriamCVComparative in vitro activities of GAR-936 against aerobic and anaerobic animal and human bite wound pathogensAntimicrob Agents Chemother20004427475110991855
  • GoldsteinEJCitronDMMerriamCVIn vitro activities of garenoxacin (BMS-284756) against 170 clinical isolates of nine Pasteurella speciesAntimicrob Agents Chemother20024630687012183274
  • GoldsteinEJCitronDMWarrenYAIn vitro activity of moxifloxacin against 923 anaerobes isolated from human intra-abdominal infectionsAntimicrob Agents Chemother2006501485516377680
  • GuayDRPPiscitelliSCRodvoldKAChapter 8. QuinolonesInfectious disease: drug interactions in infectious diseases20052nd edTotowa, NJHumana Pr.21554
  • HardyDAmsterdamDMandellLAComparative in vitro activities of ciprofloxacin, gemifloxacin, grepafloxacin, moxifloxacin, ofloxacin, sparfloxacin, trovafloxacin, and other antimicrobial agents against bloodstream isolates of gram-positive cocciAntimicrob Agents Chemother200044802510681365
  • HedbergMNordCEESCMID Study Group on Antimicrobial Resistance in Anaerobic BacteriaAntimicrobial susceptibility of Bacteroides fragilis group isolates in EuropeClin Microbiol Infect200394758812848722
  • HermsenEDHovdeLBSprandelKALevofloxacin plus metronidazole administered once daily versus moxifloxacin monotherapy against a mixed infection of Escherichia coli and Bacteroides fragilis in an in vitro pharmacodynamic modelAntimicrob Agents Chemother200549685915673752
  • HoellmanDBKellyLMJacobsMRComparative antianaerobic activity of BMS 284756Antimicrob Agents Chemother2001455899211158759
  • Hoogkamp-KorstanjeJARoelofs-WillemseJComparative in vitro activity of moxifloxacin against gram-positive clinical isolatesJ Antimicrob Chemother20004531910629010
  • HsuehPRTengLJLeeCMTelithromycin and quinupristin-dalfopristin resistance in clinical isolates of Streptococcus pyogenes: SMART Program 2001 DataAntimicrob Agents Chemother2003472152712821461
  • InceDZhangXHooperDCActivity of and resistance to moxifloxacin in Staphylococcus aureusAntimicrob Agents Chemother20034714101512654680
  • JacobsMRBajaksouzianSWindauAIn vitro activity of the new quinolone WCK 771 against staphylococciAntimicrob Agents Chemother20044833384215328094
  • JiBGrossetJCombination of rifapentine-moxifloxacin-minocycline (PMM) for the treatment of leprosyLeprosy Rev200071SupplS81711201894
  • JoukhadarCStassHMuller-ZellenbergUPenetration of moxifloxacin into healthy and inflamed subcutaneous adipose tissues in humansAntimicrob Agents Chemother200347309910314506015
  • KalteisTBeckmannJSchroderH-JMoxifloxacin superior to vancomycin for treatment of bone infections - a study in ratsActa Orthoped2006a7731519
  • KalteisTBeckmannJSchroderH-JTreatment of implant-associated infections with moxifloxacin: an animal studyInt J Antimicrob Agents2006b27444816621461
  • LabombardiVJShastryLTischlerHMycobacterium thermoresistibile infection following knee-replacement surgeryJ Clin Microbiol2005435393416208028
  • Leal del RosalPFabianGVick-FragasoREfficacy and safety of moxifloxacin (MXF) vs. cephalexin (with or without metronidazole) in the treatment of mild to moderate uncomplicated skin and skin structure infections (uSSSI) [abstract]1999Proceedings of the 39th Interscience Conference on Antimicrobial Agents and ChemotherapySept. 26–29San Diego, CA, USA1076
  • Leal del RosalPMartinezRFabianGEfficacy and safety of moxifloxacin vs. cephalexin in the treatment of mild to moderate uncomplicated skin and skin structure infections (USSSI)J Antimicrob Chemother199944suppl A148. P493
  • LemmenSWHafnerHKlikSComparison of the bactericidal activity of moxifloxacin and levofloxacin against Staphylococcus aureus, Staphylococcus epidermidis, Escherichia coli and Klebsiella pneumoniaeChemotherapy20034933512714807
  • LettieriJVargasRAgarwalVEffect of food on the pharmacokinetics of a single oral dose of moxifloxacin 400 mg in healthy male volunteersClin Pharmacokinet200140Suppl 1192511352438
  • LionCConroyMCCarpentierAMAntimicrobial susceptibilities of Pasturella strains isolated from humansInt J Antimicrob Agents200627290316564680
  • ListerPDPharmacodynamics of moxifloxacin and levofloxacin against Staphylococcus aureus and Staphylococcus epidermidis in an in vitro pharmacodynamic modelClin Infect Dis200132Suppl 1S33811249827
  • LubaschAKellerIBornerKComparative pharmacokinetics of ciprofloxacin, gatifloxacin, grepafloxacin, levofloxacin, trovafloxacin, and moxifloxacin after single oral administration in healthy volunteersAntimicrob Agents Chemother2000442600310991830
  • MacGowanAPBowkerKEHoltHABay 12-8039, a new 8-methoxy-quinolone: comparative in-vitro activity with nine other antimicrobials against anaerobic bacteriaJ Antimicrob Chemother19974050399372419
  • MalathumKSinghKVMurrayBEIn vitro activity of moxifloxacin, a new 8-methoxyquinolone, against gram-positive bacteriaDiagn Microbiol Infect Dis1999351273310579093
  • MalincarneLGhebregzabherMMorettiMVPenetration of moxifloxacin into bone in patients undergoing total knee arthoplastyJ Antimicrob Chemother200657950416551691
  • ManIMurphyJFergusonJFluoroquinolone phototoxicity: a comparison of moxifloxacin and lomefloxacin in normal volunteersJ Antimicrob Chemother199943Suppl B778210382879
  • MandellGLColemanEUptake, transport, and delivery of antimicrobial agents by human polymorphonuclear neutrophilsAntimicrob Agents Chemother2001451794811353627
  • MilatovicDSchmitzFJBrisseSIn vitro activities of sitafloxacin (DU-6859a) and six other fluoroquinolones against 8,796 clinical bacterial isolatesAntimicrob Agents Chemother20004411027
  • MitscherLBacterial topoisomerase inhibitors: quinolone and pyridone antibacterial agentsChem Rev20051055599215700957
  • MorganrothJDiMarcoJPAnzuetoACAPRIE Study GroupA randomized trial comparing the cardiac rhythm safety of moxifloxacin vs. levofloxacin in elderly patients hospitalized with community-acquired pneumoniaChest2005128339840616304291
  • MullerMStassHBrunnerMPenetration of moxifloxacin into peripheral compartments in humansAntimicrob Agents Chemother1999432345910508004
  • NoelARBowkerKNMacGowanAPPharmacodynamics of moxifloxacin against anaerobes studied in an in vitro pharmacokinetic modelAntimicrob Agents Chemother2005494234916189103
  • NoriSNebesioCBrashearRMoxifloxacin-associated drug hypersensitivity syndrome with toxic epidermal necrolysis and fulminant hepatic failure (letter)Arch Dermatol20041401537815611441
  • NovielloSIannielloFLeoneSComparative activity of garenoxacin and other agents by susceptibility and time-kill testing against Staphylococcus aureus, Streptococcus pyogenes and respiratory pathogensJ Antimicrob Chemother2003528697114519672
  • OdenholtILowdinEGustafssonIPharmacodynamics of moxifloxacin against Streptococcus pyogenes in an in vitro kinetic modelAntimicrob Agents Chemother2002462046812019138
  • ParishLCRouthHBMiskinBMoxifloxacin versus cephalexin in the treatment of uncomplicated skin infectionsInt J Clin Prac200054497503
  • Park-WyllieLYJuurlinkDNKoppAOutpatient gatifloxacin therapy and dysglycemia in older adultsN Engl J Med200635413526116510739
  • PatelMVDe SouzaNJGupteSVAntistaphylococcal activity of WCK 771, a tricyclic fluoroquinolone, in animal infection modelsAntimicrob Agents Chemother20044847546115561853
  • PeaFPavanFLugattiEPharmacokinetic and pharmacodynamic aspects of oral moxifloxacin 400 mg/day in elderly patients with acute exacerbation of chronic bronchitisClin Pharmacokinet2006452879516509760
  • Perez-GiraldoCGonzalez-VelascoCSanchez-SilosRMMoxifloxacin and biofilm production by coagulase-negative staphylococciChemotherapy200450101415211086
  • PericMJacobsMRAppelbaumPCAntianaerobic activity of a novel fluoroquinolone, WCK 771, compared with those of nine other agentsAntimicrob Agents Chemother20044831889215273148
  • RaghavanMLindenPKNewer treatment options for skin and soft tissue infectionsDrugs20046416214215257625
  • Rodriguez DiazJCLopezMRuizMIn vitro activity of new fluoroquinolones and linezolid against non-tuberculous mycobacteriaInt J Antimicrob Agents200321585812791475
  • RubinsteinEDiamantsteinLYosephGThe effect of albumin, globulin, pus and dead bacteria in aerobic and anaerobic conditions on the antibacterial activity of moxifloxacin, trovafloxacin and ciprofloxacin against Streptococcus pneumoniae, Staphylococcus aureus and Escherichia coliClin Microbiol Infect200066788111284929
  • RuizJMechanism of resistance to quinolones: target alterations, decreased accumulation and DNA gyrase protectionJ Antimicrob Chemother20035111091712697644
  • SableDMurakawaGJQuinolones in dermatologyClin Dermatol200321566312609589
  • SchaumannRAckermannGPlessBIn vitro activities of fourteen antimicrobial agents against obligately anaerobic bacteriaInt J Antimicrob Agents2000162253211091040
  • SchaumannRGoldsteinEJCForbergJActivity of moxifloxacin against Bacteroides fragilis and Escherichia coli in an in vitro pharmacokinetic/pharmacodynamic model employing pure and mixed culturesJ Med Microbiol2005547495316014428
  • SeralCVan BambekeFTulkensPMQuantitative analysis of gentamicin, azithromycin, telithromycin, ciprofloxacin, moxifloxacin, and oritavancin (LY333328) activities against intracellular Staphylococcus aureus in mouse J774 macrophagesAntimicrob Agents Chemother20034722839212821480
  • SillerstromEWahlundENordCEIn vitro activity of ABT-492 against anaerobic bacteriaJ Chemother200416227915330316
  • SnydmanDRJacobusNVMcDermottLAIn vitro activities of newer quinolones against Bacteroides group organismsAntimicrob Agents Chemother2002463276912234859
  • SpecialeAMusumeciRBlandinoGMinimal inhibitory concentrations and time-kill determination of moxifloxacin against aerobic and anaerobic isolatesInt J Antimicrob Agents2002191111811850163
  • StassHDalhoffAKubitzaDPharmacokinetics, safety, and tolerability of ascending single doses of moxifloxacin, a new 8-methoxy quinolone, administered to healthy subjectsAntimicrob Agents Chemother199842206059687407
  • StassHKubitzaDPharmacokinetics and elimination of moxifloxacin after oral and intravenous administration in manJ Antimicrob Chemother199943Suppl B839010382880
  • StassHKubitzaDSchuhlyUPharmacokinetics, safety and tolerability of moxifloxacin, a novel 8-methoxyfluoroquinolone, after repeated oral administrationClin Pharmacokinet200140Suppl 11911352436
  • StassHKubitzaDHalabiAPharmacokinetics of moxifloxacin, a novel 8-methoxy-quinolone, in patients with renal dysfunctionBr J Clin Pharmacol200253232711874385
  • StassHNagelschmitzJMoellerJGPharmacokinetics of moxifloxacin are not influenced by a 7-day pretreatment with 200mg oral itraconazole given once a day in healthy subjectsInt J Clin Pharmacol Ther20044223914756383
  • StefanopoulosPKTarantzopoulouADFacial bite wounds: management updateInt J Oral Maxillofac Surg2005344647216053863
  • StevensDLBisnoALChambersHFPractice guidelines for the diagnosis and management of skin and soft-tissue infectionsClin Infect Dis200541137340616231249
  • StratchounskiLSDekhnichAVKretchikovVAAntimicrobial resistance of nosocomial strains of Staphylococcus aureus in Russia: results of a prospective studyJ Chemother200517546015828444
  • SullivanJTWoodruffMLettieriJPharmacokinetics of a once-daily oral dose of moxifloxacin (Bay 12-8039), a new enantiomerically pure 8-methoxy quinoloneAntimicrob Agents Chemother1999432793710543767
  • SullivanJTLettieriJTLiuPThe influence of age and gender on the pharmacokinetics of moxifloxacinClin Pharmacokinet200140Suppl 1111811352437
  • TarasiACassoneMMonacoMActivity of moxifloxacin in combination with vancomycin or teicoplanin against Staphylococcus aureus isolated from device-associated infections unresponsive to glycopeptide therapyJ Chemother2003152394312868549
  • TzikourisJPPeetersMJCoxCDEffects of three fluoroquinolones on QT analysis after standard treatment coursesAnn Noninvasive Electrocardiol20061152616472283
  • von EiffCPetersGComparative in-vitro activities of moxifloxacin, trovafloxacin, quinupristin/dalfopristin and linezolid against staphylococciJ Antimicrob Chemother1999435697310350389
  • WenzlerSSchmidt-EisenlohrEDaschnerFComparative in vitro activities of three new quinolones and azithromycin against aerobic pathogens causing respiratory tract and abdominal wound infectionsChemotherapy20045040215084805
  • WiseRAndrewsJMMarshallGPharmacokinetics and inflammatory-fluid penetration of moxifloxacin following oral or intravenous administrationAntimicrob Agents Chemother19994315081010348784
  • WoodcockJMAndrewsJMBoswellFJIn vitro activity of Bay 12-8039, a new fluoroquinoloneAntimicrob Agents Chemother19974110168980763
  • WrightDHGundersonBWHovdeLBComparative pharmacodynamics of three newer fluoroquinolones versus six strains of staphylococci in an in vitro model under aerobic and anaerobic conditionsAntimicrob Agents Chemother2002461561311959599
  • YangSCHsuehPRLaiHCHigh prevalence of antimicrobial resistance in rapidly growing mycobacteria in TaiwanAntimicrob Agents Chemother20034719586212760874