http://www.medscape.com/viewarticle/409533 Advertisement

Medscape   Home   Site Map   Marketplace   My Medscape   CME Center   Feedback   Help Desk

NOTE: This page has been formatted for easy printing. To view the article with Web enhancements, go to:
http://www.medscape.com/ACCP/Pharmacotherapy/2000/v20.n04/pharm2004.09.pick/pharm2004.09.pick-01.html.


Comparison of the Fluoroquinolones Based on Pharmacokinetic and Pharmacodynamic Parameters

Katherine E. Pickerill, Pharm.D., Joseph A. Paladino, Pharm.D., FCCP, and Jerome J. Schentag, Pharm.D., FCCP., The Clinical Pharmacokinetics Laboratory, Millard Fillmore Hospitals, Buffalo, New York.

[Pharmacotherapy 20(4):417-428, 2000. © 2000 Pharmacotherapy Publications, Inc.]

Abstract

Assessment of pharmacodynamic activity from standard in vitro minimum inhibitory concentrations (MICs) alone is insufficient to predict in vivo potency. Achievable serum and tissue concentrations as well as pharmacokinetic characteristics must be considered. When pharmacokinetic and pharmacodynamic values are combined, the area under the inhibitory curve (AUIC) and peak concentration:MIC ratio predict clinical cure for fluoroquinolones. Clinical data and animal models indicate that a peak:MIC of 10:1 and above and an AUIC of 125 and above are predictive of a clinical cure for this class of antimicrobials against gram-negative organisms. The values may be used to compare and contrast fluoroquinolones to determine which would be best for treating a specific microorganism. Pharmacodynamic data also can be used to design regimens that minimize the risk of suboptimal drug levels. Ensuring the optimal fluoroquinolone dosage based on pharmacodynamic principles would diminish the emergence of resistant organisms and prevent treatment failures.

Introduction

Several fluoroquinolones are marketed in the United States and numerous others are in development. It is important to compare and contrast the relative strengths, weaknesses, and distinguishing characteristics of the members of this expanding class of antimicrobials. Unfortunately, few clinical trials directly compare clinical efficacy and side effects of these agents. Although the minimum inhibitory concentration (MIC) is a useful predictor of the activity of an antimicrobial against a microorganism, it does not necessarily indicate relative in vivo potency. For example, two antibiotics with an MIC of 1.0 µg/ml should be evaluated quite differently if one has a peak concentration of 2 µg/ml and the other a peak of 20 µg/ml. Thus, pharmacokinetic parameters must be considered for proper assessment of a drug and dosage.

Pharmacodynamics study the relationship between drug concentration and pharmacologic effect. For antimicrobials, pharmacodynamic activity can be described as concentration dependent or time dependent.[1] As fluoroquinolones have concentration-dependent killing, the peak:MIC and/or area under the curve in 24 hours (AUC24):MIC are pharmacodynamic values that best correlate with efficacy.[2-4] Understanding these parameters can facilitate selection of effective antibiotics and optimal regimens to hasten response, prevent treatment failures, and minimize the development of resistance.[5]

Spectrum of Activity

Median MIC90 values were used due to the broad range of results in various in vitro studies. Each fluoroquinolone has excellent in vitro activity against most Enterobacteriaceae (Table 1).[6-22 ]Clinafloxacin, ciprofloxacin, moxifloxacin, and gatifloxacin have the lowest MICs against various gram-negative pathogens such as Escherichia coli and Klebsiella, Enterobacter, Citrobacter, Proteus, Salmonella, and Shigella sp.[6-18] Although gatifloxacin, clinafloxacin, and moxifloxacin show excellent in vitro gram-negative activity[7,11,12,15-18] comparable with or superior to that of ciprofloxacin, it will be interesting to see if in vitro results correlate with clinical results. Clinafloxacin and ciprofloxacin produce the lowest MICs for Pseudomonas aeruginosa (range 0.5-4 µg/ml, median 2 µg/ml).[7,9-13,16,18-20] The MICs of other fluoroquinolones against P. aeruginosa are at least 2 times higher than those of ciprofloxacin and clinafloxacin. Trovafloxacin, grepafloxacin, moxi-floxacin, and clinafloxacin have the best in vitro activity against Stenotrophomonas maltophilia with an MIC of 2 µg/ml, whereas activity is limited for all of the other fluoroquinolones.[7,8,13,20] In general, these drugs have excellent in vitro activity against Haemophilus influenzae and Moraxella catarrhalis.[9,21-23]

Data suggest that fluoroquinolones preferentially target DNA gyrase in gram-negative bacteria, whereas topoisomerase IV is the primary target in gram-positive bacteria.[24-26] The preferential topoisomerase target within the cell also may be determined by fluoroquinolone structure.[26] This implies that it is possible for some fluoroquinolones to have equal activity against both DNA gyrase and topoisomerase IV.[26] Therefore, they would show activity against a broad range of gram-positive and -negative bacteria. A deficiency of ciprofloxacin, ofloxacin, levofloxacin, and other earlier fluoroquinolones is limited potency against gram-positive bacteria. The newer fluoroquinolones and others under development have improved activity against these organisms while retaining activity against gram-negative pathogens. Sparfloxacin, grepafloxacin, trovafloxacin, gatifloxacin, clinafloxacin, and moxifloxacin have greater in vitro activity than ciprofloxacin and ofloxacin against Staphylococcus aureus and Staphylococcus epidermidis (Table 2).[7-13,23,27] However, staphylococcal strains that are methicillin resistant usually are resistant to fluoroquinolones.[7-13,23,27] Gatifloxacin, clinafloxacin, and moxifloxacin have in vitro activity against methicillin-resistant strains of S. epidermidis, but clinical activity will depend on whether resistance develops during therapy.[11,15,16] Most studies report MIC90s of ciprofloxacin between 2 and 4 µg/ml for Enterococcus faecalis and greater than 4 µg/ml for Enterococcus faecium.[7,8,10,11,13-15,22,27] Neither ofloxacin nor levofloxacin offers any advantage over ciprofloxacin for these bacterial strains.[9] Grepafloxacin, sparfloxacin, trovafloxacin, clinafloxacin, and moxifloxacin inhibit E. faecalis at concentrations of 0.5-1 µg/ml, but E. faecium tends to be more resistant.[7,8,10,11,13-15,22,27] For S. pneumoniae, ciprofloxacin and ofloxacin MICs are 2 µg/ml or greater compared with the other fluoroquinolones, 0.06-1 µg/ml.[16,21,28] Strains resistant to, or with decreased susceptibility to, penicillin remain susceptible to the agents.[16,21,28 ] Sparfloxacin, grepafloxacin, trovafloxacin, gatifloxacin, clinafloxacin, and moxifloxacin show excellent in vitro activity against Streptococcus pyogenes and Streptococcus agalactiae.[7,11,15,16]

All drugs in this class are highly active against Legionella sp, but other atypical microorganisms exhibit variable susceptibility patterns (Table 3).[11,23,29-39] Several newer agents appear to be substantially more active than ciprofloxacin or ofloxacin against Mycoplasma pneumoniae and Chlamydia pneumoniae. The in vitro suscepti-bilities of 21 isolates of C. pneumoniae to grepafloxacin were 0.06-0.12 µg/ml, with an MIC90 of 0.12 µg/ml.[31] Gatifloxacin and moxifloxacin have the greatest in vitro activity against C. pneumoniae, followed by grepafloxacin and sparfloxacin.[11,31,34,35] Gatifloxacin, moxifloxacin, trovafloxacin, and sparfloxacin show the best activity against M. pneumoniae. [36-38]

Ciprofloxacin and ofloxacin have relatively weak in vitro activity against most anaerobes. Whereas levofloxacin, grepafloxacin, and sparfloxacin have somewhat lower MIC90s than ciprofloxacin to some anaerobic bacteria, they should not be considered clinically active against all anaerobes (Table 4).[7,9,10,13,27,40-42] Of currently available fluoroquinolones, trovafloxacin has considerably better in vitro activity against these pathogens. Trovafloxacin inhibits 90% of Bacteroides fragilis and Clostridium sp at concentrations 0.5-2 µg/ml, and 90% of Peptostreptococcus sp at concentrations 0.5-1 µg/ml.[7,10,11,40] Gatifloxacin and clinafloxacin have lower MIC levels than trovafloxacin against Peptostreptococcus, Clostridium perfringens, and other clostridia sp[7,11,41,42] Moxifloxacin appears to have some anaerobic activity but less than trovafloxacin, clinafloxacin, or gatifloxacin.

Pharmacokinetics

Although a microorganism's MIC90 is an important indicator of antibiotic activity, drugs must be compared based on achievable concentrations in vivo to determine clinical potency. Thus, pharmacokinetics must be considered when comparing and evaluating fluoroquinolones. These drugs vary in their pharmacokinetic properties (Table 5).[43-74]

Absorption and Distribution

Fluoroquinolones typically have excellent bioavailability, large volumes of distribution, extensive tissue penetration, and low plasma protein binding. Bioavailability of oral ofloxacin, levofloxacin, sparfloxacin, trovafloxacin, gatifloxacin, clinafloxacin, and moxifloxacin is in excess of 85%.[47,49,53,58,68-70,73,74] Bioavailability for oral ciprofloxacin and grepafloxacin is 70-80%.[47,55,56] Peak serum concentrations usually are achieved 1-2 hours after dosing in healthy individuals, except for sparfloxacin, which reaches peak serum concentrations 4-5 hours after the dose.[47,53,55,58,62-65,68,72] This most likely is due to the drug's low solubility in aqueous solutions.[58] The maximum concentration (Cmax) varies significantly among new and investigational fluoroquinolones. In dose-ranging studies, Cmaxand AUC increased in a linear, dose-proportional fashion.[43,58,69,74] As indicated by their large volumes of distribution, most fluoroquinolones penetrate rapidly and efficiently through the body, achieving tissue and fluid concentrations that are generally higher than those in plasma.[47,53,55,60,66] Most fluoroquinolones have low protein binding, ranging from 2-40%.[47,52,53,55,58,63,65] Thus, any compromise of antimicrobial activity by changes in the extent of plasma protein binding should be minimal. The protein binding of trovafloxacin is 70%, which is high for a member of this class[61]; values for grepafloxacin and moxifloxacin are 50% and 55%, respectively.[56,74] The implications of the higher protein binding of these three agents are unclear as it is difficult to determine in vivo concentration changes.

Metabolism and Elimination

Longer beta-half-lives of trovafloxacin, grepafloxacin, sparfloxacin, and moxifloxacin allow for once-daily dosing. The elimination half-lives of trovafloxacin, grepafloxacin, sparfloxacin, and moxifloxacin exceed 10 hours, whereas those of ciprofloxacin, ofloxacin, levofloxacin, gatifloxacin, and clinafloxacin range between 5 and 8 hours.[47,54,56,58,63-65,67] Although levofloxacin's elimination half-life compares with those of ciprofloxacin and ofloxacin, this drug is marketed for once-daily dosing. Based on elimination rates, AUC values, and evolving clinical data, it appears that moxifloxacin and gatifloxacin can be dosed once/day.[63-65,68,69,72] Clinafloxacin would provide adequate MIC90 coverage with twice-daily 200- and 400-mg oral or intravenous regimens.[70]

Fluoroquinolones are removed from the body by both renal and nonrenal mechanisms. Ofloxacin, levofloxacin, gatifloxacin, and clinafloxacin undergo limited metabolism in humans, with more than 60% of unchanged drug found in urine after administration.[53,63-66] A dosage adjustment should be made for levofloxacin and ofloxacin when the patient's creatinine clearance is less than 50 ml/minute.[52,53] It appears that the daily clinafloxacin dose should be halved in patients with creatinine clearances less than 40 ml/minute.[66,70] Trovafloxacin, moxifloxacin, and grepafloxacin do not require a decrease in dosage in patients with impaired renal function. Trovafloxacin undergoes extensive hepatic metabolism to an inactive metabolite, and less than 10% of unchanged drug is renally eliminated[73]; it is recommended that the dosage be decreased in patients with mild or moderate cirrhosis.[72 ] Moxifloxacin undergoes conjugation to two metabolites that appear to have no major antimicrobial activity, and approximately 20% of the administered dose is excreted in urine.[74] Grepafloxacin is eliminated primarily through hepatic metabolism and biliary excretion, and should be avoided in patients with hepatic failure as less than 10% is eliminated in urine as unchanged drug.[57] Sparfloxacin and ciprofloxacin have two routes of elimination; nonetheless, a decrease in dosage is recommended for patients with creatinine clearances less than 50 and less than 30 ml/minute, respectively.[45,71]

Pharmacodynamics

Pharmacodynamics integrate antimicrobial activity and pharmacokinetics of an antibiotic by focusing on inhibiting growth and killing bacteria. The value most often used to characterize antimicrobial activity is the MIC, which reflects the net drug effect after incubation of a standard inoculum of an organism with a fixed and constant concentration of drug for 18-24 hours.[1] Since it does not account for the time course of antimicrobial activity, MIC cannot adequately characterize an antibiotic's pharmacodynamic properties.[5] Investigators employed a variety of methods to study the pharmacodynamic parameter that best correlates with efficacy. The most common methodologies are direct comparisons of different dosing schedules in vitro and in treatment of infections in animals and humans. As fluoroquinolones have concentration-dependent killing, the peak:MIC or the AUC24:MIC (AUIC) would be most predictive of outcome.[2-4,75-77]

Studies in dynamic in vitro models of infection examined the relationship between dosage and bacterial killing. In these systems, bacteria are exposed to fluctuating concentrations of drug adjusted to simulate peak and trough serum concentrations observed in humans. The models suggest that a peak:MIC ratio of 10 or above and an AUIC ratio of 125 or above optimize rapid bacterial killing and prevent regrowth of resistant gram-negative bacterial subpopulations.[75,76]

Animal studies confirmed these findings. In two murine models, total exposure or AUC was the variable most closely linked to outcome.[77] An AUIC of 100 or greater, peak:MIC above 8, and serum levels above MIC 100% of the time predicted efficacy of fluoroquinolones in treating animal models of endocarditis; however, the AUIC showed the best linear correlation.[78] In a study that examined how altering the dosage or treating a more susceptible organism influenced survival in a neutropenic rat model of P. aeruginosa sepsis, either the peak:MIC ratio or AUIC ratio of lomefloxacin correlated with efficacy.[4] However, when the AUC was kept constant and the dosage was changed to increase the peak:MIC ratio (>= 20:1), outcomes improved significantly. The investigators hypothesized that the peak:MIC was an important parameter because, in vitro, ratios of 10 or more prevented emergence of resistant organisms.[75] However, dose-related toxic side effects associated with fluoroquinolones prevent the attainment of these high peak concentrations in the clinical setting.

Relatively few studies were conducted in humans to confirm the hypothesis generated by in vitro and animal models. Treatment of respiratory, urinary, or skin and soft tissue infections with levofloxacin resulted in a high frequency of clinical and microbiologic cure when the peak:MIC ratio was greater than 12.2.3 It should be noted that the peak:MIC ratio and AUIC ratio were highly correlated, with a mean AUIC ratio below 65 in the group with unsuccessful outcome but greater than 100 in the group with successful outcome.

The AUIC was the most significant variable for the probability of a clinical cure for seriously ill patients treated with ciprofloxacin.[2] The probability of a clinical cure was 69% at an AUIC of 125 (log10 = 2.1) and approximately 80% at an AUIC of 250 (log10 = 2.4; Figure 1). In this trial, daily cultures were obtained to derive time to bacterial eradication. A statistically significant relationship was seen between time to bacterial eradication and the individual AUIC achieved in each patient (Figure 2). An AUIC of 125-250 resulted in effective bacterial killing, with eradication requiring approximately 7 days. When AUIC values were 250, bacterial killing was extremely rapid, with eradication averaging 2 days. However, increasing the AUIC beyond 250 did not increase the rate of eradication. The authors concluded that an AUIC of 125 or above is an effective threshold of antimicrobial activity for seriously ill hospitalized patients. Similar to an earlier study,[3] the AUIC and peak:MIC ratio were highly correlated in these patients.

art

Figure 1. Percentage probability of a clinical cure versus AUIC fit to a modified Hill equation. The curve is the fitted relationship; each point represents three or four patients. A log10AUIC of 2.1 is equivalent to an AUIC of 125, and log10AUIC of 2.4 is equivalent to 250.[2]
art

Figure 2. Time (days of therapy) to bacterial eradication versus AUIC illustrated by a time-to-event plot. Shown is the day of therapy versus patients remaining culture-positive on that day.[2]
The question of whether a relationship exists between AUIC and development of bacterial resistance was examined in a population of patients, virtually all of whom were treated with ciprofloxacin for lower respiratory tract infections.[79] An inverse relationship was found between the probability of developing bacterial resistance and AUIC. When the AUIC ratio was below 100, 82% of patients developed an infection with a resistant organism, most likely by selection of a subpopulation. However, when the AUIC ratio was above 100, only 9% of the patients developed a resistant infection. This suggests that resistance can be avoided with attention to dosing, since regimens that provide an AUIC of at least 100 appear to reduce the rate of resistance (Figure 3).[79] Therefore, an estimated AUIC of 125 or peak:MIC of 8-10:1 may result in optimal bacterial killing, minimize the potential for resistance, and improve clinical outcome against gram-negative pathogens.

art

Figure 3. Relationship between the probability of developing resistance and treatment duration (days). When the AUIC ratio was 100 or greater, only 8% of organisms developed resistance, whereas only 7% remained susceptible when the AUIC ratio was less than 100. (Modified from reference 79.)
In an outpatient study,[80] treatment of acute exacerbations of chronic bronchitis with grepafloxacin resulted in bacteriologic and clinical cures when the AUIC was 75 or above. The AUIC breakpoint in these patients was lower than the 125 or greater that was recommended. This difference probably was due to patients' immunocompetence. This effect also may be related to disease severity and virulence of the organism. Of interest, the analysis revealed that an AUIC above 175 was optimal as it achieved rapid eradication of the organism.[80]

The killing rate of ciprofloxacin was examined against strains of S. pneumoniae, S. aureus, and P. aeruginosa for which MICs were similar.[81] At clinically achievable concentrations (peak 3-6 times MIC), ciprofloxacin killed P. aeruginosa more rapidly than it did either S. aureus or S. pneumoniae. At higher concentrations of 15-18 times MIC, killing rates were similar for the three organisms. This implies some differences in interorganism susceptibility that cannot be discerned solely by measuring MIC.

A study of the impact of the dosing interval on the AUIC for six fluoroquinolones against various gram-negative and -positive organisms in thighs and lungs of neutropenic mice produced similar results.[82,83] A 24-hour AUC:MIC ratio of 25 for gram-negative bacteria and 53 for gram-positive bacteria was required to achieve a bacteriostatic effect, suggesting that gram-positive bacteria are more difficult to eradicate than gram-negative. An AUIC breakpoint lower than 125 may be adequate for treating bacterial infections in immunocompetent patients, as suggested by the grepafloxacin outpatient study.[80] However, an AUIC of 25-50 would be ineffective for seriously ill patients.[2,3] Thus, when directly comparing fluoroquinolones, an AUIC break-point of 125 should be used as it predicts clinical efficacy in humans with serious infections and may prevent treatment failures.

Integrating Pharmacokinetics and Pharmacodynamics

Choosing a drug with the appropriate spectrum of activity is an important component of empirically treating bacterial infections. However, understanding the pharmacokinetic and pharmacodynamic characteristics of an antibiotic can provide insight into the optimal dosage for a given drug. For these agents, regimens of large doses given at infrequent intervals are the most efficacious in terms of bacterial killing, eradication time, and reducing the selection of resistant bacteria. It is difficult to distinguish which pharmacodynamic value -- peak:MIC or AUIC -- is most predictive of a clinical cure as a strong interrelationship exists between them.[2-4,75-78]

Data required to determine these pharmaco-dynamic parameters are MICs, peak serum concentrations, and AUCs for typical dosages extracted from pharmacokinetic studies conducted at steady state in healthy volunteers (Table 5). Pharmacodynamic data in Table 6 indicate AUICs achieved at recommended dosages in patients with estimated creatinine clearances above 90 ml/minute. Creatinine clearance was chosen because it was the cut-off value frequently used in studies in healthy volunteers. The AUIC values of Legionella, M. pneumoniae, and C. pneumoniae were not included in this table as the pharmacodynamics of fluoroquinolones against intracellular-growing bacteria have yet to be examined.[82]

When available, intravenous dosages were used to calculate AUICs. Fluoroquinolones do not achieve an effective AUIC against several bacterial species; however, in some instances they may be clinically effective even though pharmacodynamic data predict otherwise. One potential explanation is that AUCs in infected patients are larger than those in healthy volunteers. Second, AUCs of fluoroquinolones that are excreted renally may be increased in patients with mild renal insufficiency. Another explanation is that actual MICs of these organisms are less than median MIC90s. To explore the implications of an MIC lower than the MIC90, AUIC values can be recalculated using MIC50 values; resulting AUIC values approach 125 for many fluoroquinolones against more organisms. However, this may not be clinically acceptable since only half of the pathogens are inhibited. The MIC90 should be used as it is more conservative, but this may result in an overdose from an empiric standpoint if the goal is obtaining an AUIC of 125 for an organism with a lower than predicted MIC. Once sensitivity data are reported by the microbiology laboratory, the actual MIC should be used to determine dosage.

To achieve an AUIC of 125, MICs of sparfloxacin, trovafloxacin, gatifloxacin, and clinafloxacin should be 0.25 µg/ml or less, and MICs of ciprofloxacin and grepafloxacin should be 0.20 µg/ml or less. The MICs of levofloxacin and moxifloxacin should be 0.43 µg/ml or below and 0.38 µg/ml or below, respectively. These MIC breakpoints that result in an AUIC of 125 differ from National Committee for Clinical Laboratory Standards (NCCLS) breakpoints of susceptibility (Table 7).[84] Using these breakpoints to calculate AUIC will result in values of 20-40, similar to the AUIC ratio necessary to produce a net bacteriostatic effect in animal models of infection, and may be adequate to treat outpatients.[80, 82]

Pharmacodynamic data suggest that cipro-floxacin, ofloxacin, levofloxacin, moxifloxacin, gatifloxacin, and clinafloxacin would be effective in treating infections caused by gram-negative enteric pathogens as the AUIC threshold of 125 is achieved with recommended dosages. Grepafloxacin, sparfloxacin, and trovafloxacin have adequate pharmacodynamic activity against most Enterobacteriaceae but have decreased pharmacodynamic activity against several genera in that family.

The only fluoroquinolones that produce adequate peak:MIC ratios for treating susceptible strains of P. aeruginosa are ciprofloxacin and clinafloxacin, but dosages of 400 mg twice/day do not achieve an AUIC of 125 or greater in patients with normal renal function. Increasing the ciprofloxacin schedule from twice/day to 3 times/day has little benefit because the AUIC continues to be suboptimal. With normal renal function, ciprofloxacin 400 mg administered intravenously every 8 hours may not be sufficient to achieve a breakpoint of 125 in infections caused by bacteria for which MICs are 0.5 µg/ml or greater. Further studies are necessary to determine if higher dosages or combination therapy is the best strategy. The other fluoro-quinolones do not appear to be acceptable options for treatment of susceptible strains of P. aeruginosa due to defects in either pharmaco-kinetics (low AUC) or potency (high MIC).

Recommended dosages for ofloxacin, grepafloxacin, sparfloxacin, and trovafloxacin produce peak:MIC and AUIC ratios against methicillin-sensitive S. aureus that suggest efficacy of these agents against this micro-organism. Levofloxacin 500 mg/day does not result in an effective AUIC; however, this may change in patients with impaired renal function, as 85% of the drug is eliminated renally. For example, a patient with an estimated creatinine clearance of 50 ml/minute would have an AUIC of 192 secondary to an increase in the AUC.[85] Dosage reductions of levofloxacin are recommended for patients with creatinine clearances less than 50 ml/minute, which may not produce an adequate AUIC. In patients with normal renal function, ciprofloxacin does not appear to be an option for treating staphylococcal infections unless the MIC is 0.20 µg/ml or below. Preliminary data imply that gatifloxacin, clinafloxacin, and moxifloxacin would be effective in treating methicillin-susceptible S. aureus.

Although MIC data for grepafloxacin, sparfloxacin, gatifloxacin, and clinafloxacin show good activity against groups A and B streptococci, pharmacodynamic data suggest otherwise. However, an AUIC of 125 or above for trovafloxacin and moxifloxacin indicates that these agents would be effective in eradicating the organisms. The favorable pharmacokinetic profiles, particularly high AUC, of ciprofloxacin, ofloxacin, and levofloxacin, are not sufficient to overcome the high MICs; thus they should not be given for empiric treatment of groups A and B streptococcal infections. Fluoroquinolones are not reliable choices for empiric treatment of suspected Enterococcus infection, as neither peak:MICs nor AUICs are at values that would predict acceptable clinical efficacy for this genus.

Because of weak activity against S. pneumoniae, ofloxacin and ciprofloxacin usually are not considered first-line treatment for respiratory tract infections, but this is not true of newer fluoroquinolones. Based on the AUIC target of 125, drugs with the best activity against S. pneumoniae are clinafloxacin, moxifloxacin, sparfloxacin, and trovafloxacin. Grepafloxacin will be close to an AUIC of 125, but the AUIC of levofloxacin and gatifloxacin may be too low if the goal is covering the MIC90 and preventing microbial resistance. Although pharmacodynamic parameters imply that grepafloxacin, levofloxacin, and gatifloxacin are not optimally active against S. pneumoniae, clinical data demonstrate efficacy of these agents.[86-89] An explanation for this contradiction may be that an outpatient population may not require an AUIC threshold of 125 to achieve clinical response. They may have to achieve an AUIC of only 75, as suggested by one outpatient grepafloxacin study.[80] These newer agents have close to an ideal spectrum of activity against respiratory pathogens, showing effective pharmacodynamic activity against S. pneumoniae, H. influenzae, M. catarrhalis, C. pneumoniae, M. pneumoniae, and Legionella sp. Levofloxacin, sparfloxacin, clinafloxacin, gatifloxacin, and moxifloxacin appear to be dependable for empiric treatment of community-acquired pneumonia. Trovafloxacin should not be given to treat outpatient community-acquired pneumonia since concerns of hepatic toxicity limit its application to hospitalized patients. Grepafloxacin is no longer a consideration as it has been withdrawn from the market.

Trovafloxacin is the first of the fluoroquinolones to be approved as monotherapy for intraabdominal infections caused by mixed aerobic and anaerobic flora, due in part to its in vitro activity against anaerobic organisms.[60] It was effective in treating these infections in animal models and clinical trials.[90-92] However, when MIC and pharmacokinetic data are both incorporated, trovafloxacin does not achieve an AUIC of 125 or above against anaerobic organisms. Unfortunately, none of the clinical trials evaluating pharmacodynamics of fluoroquinolones included infections caused by anaerobic bacteria, so it is unknown if a lower threshold of AUIC or peak:MIC would be sufficient.

Summary

The peak:MIC and AUIC predict efficacy of the fluoroquinolones. As they demonstrate concentration-dependent activity, dosing once or twice/day to attain high peak:MIC is the obvious schedule, whereas AUIC guides the daily amount of drug necessary for optimal outcomes. Adequate peak:MIC ratio and AUIC appear to be necessary to prevent selection of resistant organisms. The data presented here show that clinical efficacy of the fluoroquinolones in hospitalized patients correlates with an AUIC of 125 and above and a peak:MIC of 10:1 and above, although only ciprofloxacin and levofloxacin have been evaluated in this patient population. It appears that a lower AUIC (>= 75) produces an adequate response in outpatients treated with grepafloxacin, whereas a value above 175 resulted in more rapid killing of the organism. Further studies must be conducted to determine if these indexes relate to the remaining fluoroquinolones and to infections other than pneumonia and acute exacerbations of chronic bronchitis. It is important to remember that pharmacokinetics reflective of healthy volunteers were used in these pharmacodynamic calculations and that clinical results with any of the fluoroquinolones will depend on the actual MIC for the organism and the AUC achieved in each individual patient. One could compensate for a defect in either value by increasing the dosage, thereby making these drugs more active. This occurs naturally in patients with decreased renal function who receive maximum dosages of drugs that are eliminated renally.

Selection of the optimal fluoroquinolone and regimen requires careful consideration of both organism- and patient-specific factors, including spectrum of activity and pharmacokinetics. Using the above concepts, clinicians can design dosing regimens to optimize fluoroquinolone therapy and prevent emergence of resistant organisms and treatment failures.

Table 1. Median MIC90s (µg/ml) of Gram-Negative Organisms to Fluoroquinolones[6-22]

Organism Cipro Oflox Levo Grepa Spar Trova Gati Clina Moxi
E. coli
0.03
0.12
0.06
0.12
0.10
0.25
0.016
0.008
0.008
Klebsiella sp
0.06
0.50
0.25
0.25
0.25
0.25
0.13
0.03
0.13
Enterobacter sp
0.06
0.25
0.06
0.25
0.12
0.12
0.06
0.03
0.06
Citrobacter sp
0.06
0.25
0.12
0.50
0.25
0.25
0.25
0.13
0.25
Proteus sp
0.06
0.50
0.25
0.50
0.50
0.50
0.25
0.03
0.25
M. morganii
0.06
0.25
0.06
0.50
0.50
0.50
0.25
0.06
0.25
Salmonella sp
0.03
0.12
0.06
0.10
0.06
0.06
0.06
0.008
0.13
Shigella sp
0.03
0.06
0.016
0.10
0.03
0.03
0.016
0.004
0.03
P. aeruginosa
2.0
6.0
4.0
8.0
4.0
4.0
6.0
2.0
8.0
S. maltophilia
8.0
8.0
8.0
2.0
8.0
2.0
3.13
2.0
2.0
H. influenzae
0.015
0.06
0.03
0.03
0.015
0.015
0.016
0.008
0.06
M. cattarhalis
0.06
0.12
0.06
0.03
0.015
0.03
0.03
0.008
0.06
Cipro = ciprofloxacin; Oflox = ofloxacin; Levo = levofloxacin; Grepa = grepafloxacin; Spar = sparfloxacin; Trova = trovafloxacin; Gati = gatifloxacin; Clina = clinafloxacin; Moxi = moxifloxacin.

Table 2. Median MIC90s (µg/ml) of Gram-Positive Organisms to Fluoroquinolones[7-13,15,16,20,21,27]

Organism Cipro Oflox Levo Grepa Spar Trova Gati Clina Moxi
S. aureus, MS
1.0
0.5
0.5
0.12
0.13
0.06
0.13
0.06
0.06
S. aureus, MR
>16.0
>16.0
16.0
16.0
>16.0
4.0
6.25
8.0
4.0
S. epidermidis, MS
1.0
>4.0
0.5
0.12
0.25
0.13
0.25
0.25
0.13
S. epidermidis, MR
>16.0
>16.0
16.0
8.0
4.0
4.0
0.25
0.25
0.13
S. pyogenes
1.0
2.0
1.0
0.5
0.5
0.13
0.5
0.5
0.25
S. agalactiae
1.0
2.0
1.0
0.5
0.5
0.25
0.5
0.25
0.25
E. faecalis
2.0
4.0
2.0
0.5
0.5
1.0
2.0
0.5
0.5
E. faecium
4.0
>16.0
>16.0
8.0
1.0
2.0
4.0
2.0
2.0
S. pneumoniae
     PCN susceptiblea
2.0
2.0
1.0
0.5
0.25
0.25
1.0
0.06
0.25
     PCN intermediateb
2.0
2.0
1.0
0.25
0.25
0.25
0.5
0.06
0.25
     PCN resistantc
2.0
2.0
1.0
0.25
0.25
0.25
0.5
0.06
0.12
MS = methicillin sensitive; MR = methicillin resistant; PCN = penicillin.
aPenicillin susceptible is defined by an MIC of 0.06 µg/ml or less.
bPenicillin intermediate is defined by an MIC of 0.12-1 µg/ml.
cPenicillin resistant is defined by an MIC of 2 µg/ml or greater.

Table 3. Median MIC90s (µg/ml) of Atypical Bacteria to Fluoroquinolones[11,29-39]

Organism Cipro Oflox Levo Grepa Spar Trova Gati Clina Moxi
L. pneumophila
0.03
0.03
0.03
0.015
0.03
0.015
0.016
0.03
0.06
M. pneumoniae
2.0
2.0
0.5
0.25
0.12
0.12
0.06
NA
0.12
C. pneumoniae
2.0
1.0
0.5
0.25
0.25
1.0
0.12
NA
0.06
NA = not available.

Table 4. Median MIC90s (µg/ml) of Anaerobic Bacteria to Fluoroquinolones[7,9-11,13,21,40-42]

Organism Cipro Oflox Levo Grepa Spar Trova Gati Clina Moxi
B. fragilis
16.0
16.0
2.0
8.0
4.0
0.5
1.0
1.0
1.0
Bacteroides sp
32.0
NA
4.0
8.0
4.0
2.0
2.0
2.0
4.0
Peptostreptococcus sp
4.0
8.0
8.0
2.0
1.0
1.0
0.39
0.5
1.0
C. perfringens
0.5
0.5
NA
1.0
0.5
0.5
0.39
0.13
0.5
C. difficile
16.0
8.0
8.0
NA
NA
2.0
2.0
1.0
2.0
Clostridium sp
4.0
8.0
1.0
16.0
16.0
1.0
0.5
0.5
0.12
NA = not available.

Table 5. Pharmacokinetics of Fluoroquinolones in Healthy Volunteers (creatinine clearances > 90 ml/min)[43-74]

Drug Dosage Cmax
(µg/ml)
Tmax
(hrs)
Half-life
(hrs)
AUC24
(µg/ml hr)
Vd
(L/kg)
F (%) Protein
Binding (%)
Excretion
Ciprofloxacin
500 mg p.o. q12h
3.0
1.1
5-6
27.6-28.2
2.1-5
70-80
20-40
66% renal
 
400 mg i.v. q12h
4.4
 
 
25.4
 
 
 
33% hepatic
 
750 mg p.o. q12h
4.4
 
 
39.2-42.2
 
 
 
 
Ofloxacin
400 mg p.o. q12h
4.0
1.4
4.8
82.4-96.2
1.5
98
32
70% renal
 
400 mg i.v. q12h
6.5
 
6.5
87.0
 
 
 
 
Levofloxacin
500 mg p.o. q24h
5.7
1.1
6-8
47.5
1.1-1.3
99
24-38
85% renal
 
500 mg i.v. q24h
6.4
 
 
54.6
 
 
 
 
Grepafloxacin
400 mg p.o. q24h
1.4
2
12
14.4
5
70
50
10% renal
 
600 mg p.o. q24h
1.9
 
 
24.6
 
 
 
90% hepatic
Sparfloxacin
200 mg p.o. q 24h
0.7-1.3
4-5
15-20
17.7-18.8
4.3-5.5
92
40
15% renal
 
400 mg p.o. q24h
1.2-1.5
 
 
32.3-35.7
 
 
 
55% hepatic
Trovafloxacin
200 mg p.o. q24h
2.3
1.2
12
31.2
1.3
88
70
23% renal
 
300 mg i.v. q24h
4.3
 
 
40.0
 
 
 
63% hepatic
Gatifloxacin
400 mg p.o. q24h
4.3
1.0
7-8
34.4
1.7-2.0
93
20
80% renal
 
400 mg i.v. q24h
4.6
 
 
35.2
 
 
 
 
Clinafloxacin
200 mg i.v. q12h
2.6
1.5
5-7.6
22.0
1.9-2.5
90
2-7
50-70% renal
 
400 mg i.v. q 12h
5.0
 
 
47.0
 
 
 
 
Moxifloxacin
400 mg p.o. q24h
4.5
1.2
12
48.0
1.8
95
55
20% renal
 
400 mg i.v.a
4.6
 
 
36.9
 
 
 
 
F = bioavailability.
aDerived from single-dose studies.

Table 6. AUICs of Fluoroquinolones for Selected Pathogens (numbers in italics indicate AUIC values of 75-125, numbers in bold indicate AUIC values < 75)

Organism Cipro Oflox Levo Grepa Spar Trova Gati Clina Moxi
E. coli
847
725
910
205
340
160
2213
5875
6000
Klebsiella sp
423
174
218
98
136
160
272
1567
369
Enterobacter sp
423
348
910
98
283
333
590
1567
800
Citrobacter sp
423
348
455
49
136
160
142
362
192
Proteus sp
423
174
218
49
68
80
142
1567
192
M. morganii
423
348
910
49
68
80
142
783
192
Salmonella sp
847
725
910
246
567
667
590
5875
369
Shigella sp
847
1450
3640
246
1133
1333
2213
11750
1600
P. aeruginosa
13
15
14
3
9
10
6
24
6
S. maltophilia
3
11
7
12
4
20
11
24
24
H. influenzae
1693
1450
1820
820
2267
2667
2213
5875
800
M. cattarhalis
423
725
910
820
2267
1333
1180
5875
800
MSSA
25
174
109
205
262
667
272
783
800
MRSA
<2
5
3
2
<2
10
6
6
12
MSSE
25
22
109
205
136
308
142
188
369
MRSE
<2
5
3
3
9
10
142
188
369
S. pyogenes
25
44
55
49
68
308
71
94
192
S. agalactiae
25
44
55
49
68
160
71
188
192
E. faecalis
13
22
27
49
68
40
18
94
96
E. faecium
6
5
<3
3
34
20
9
24
24
S. pneumoniae
6
44
55
98
136
160
71
783
192
B. fragilis
2
5
27
3
9
80
35
47
48
Bacteroides sp
<1
--
14
3
9
20
18
24
12
Peptostreptococcus sp
6
11
7
12
34
40
90
94
48
C. perfringens
51
174
--
25
68
80
90
361
96
C. difficile
2
11
7
--
--
20
18
47
24
Clostridium sp
6
11
55
2
2
40
71
94
400
MSSA = methicillin-sensitive S. aureus; MRSA = methicillin resistant S. aureus; MSSE = methicillin-sensitive S. epidermidis; MRSE = methicillin-resistant S. epidermidis.

Table 7. Pharmacodynamics at NCCLS Breakpoints for Susceptibility[84]

NCCLS
MIC
Drug Dosage
(mg/day)
Breakpoint
(µg/ml)
Cmax:MIC AUIC24
Ciprofloxacin
800
1
4:1
25
Ofloxacin
800
2
3:1
44
Levofloxacin
500
2
3:1
28
Grepafloxacin
600
1
2:1
25
Sparfloxacin
400
1
2:1
34
Trovafloxacin
300
2
2:1
20
Moxifloxacin
400
2
2:1
24
NCCLS = National Committee for Clinical Laboratory Standards.

References

  1. Vogelman B, Craig WA. Kinetics of antimicrobial activity. J Pediatr 1986;108:835-40.
  2. Forrest A, Nix DE, Ballow CH, Goss TF, Birmingham MC, Schentag JJ. Pharmacodynamics of intravenous ciprofloxacin in seriously ill patients. Antimicrob Agents Chemother 1993;37:1073-81.
  3. Preston SL, Drusano GL, Berman AL, et al. Pharmaco-dynamics of levofloxacin: a new paradigm for early clinical trials. JAMA 1998;279:125-9.
  4. Drusano GL, Johnson DE, Rosen M, Standiford HC. Pharmacodynamics of a fluoroquinolone antimicrobial agent in a neutropenic rat model of Pseudomonas sepsis. Antimicrob Agents Chemother 1993;37:483-90.
  5. Ebert SC, Craig WA. Pharmacodynamic properties of antibiotics: application to drug monitoring and dosage regimen design. Infect Control Hosp Epidemiol 1990;11:319-26.
  6. Rolstore KVI, Ho DH, LeBlanc B, Streeter H, Dvorak T. In vitro activity of trovafloxacin against clinical bacterial isolates from patients with cancer. J Antimicrob Chemother 1997;39:S15-22.
  7. Bauernfeind A. Comparison of the antimicrobial activities of the quinolones Bay 12-8039, gatifloxacin (AM-1155), trovafloxacin, clinafloxacin, levofloxacin, and ciprofloxacin. J Antimicrob Chemother 1997;40:639-51.
  8. Canton E, Peman J, Jimenez MT, Ramon MS, Gobernado M. In vitro activity of sparfloxacin compared with those of five other quinolones. Antimicrob Agents Chemother 1992;36:558-65.
  9. Fu KP, Lafredo SC, Foleno B, et al. In vitro and in vivo antibacterial activities of levofloxacin, an optically active ofloxacin. Antimicrob Agents Chemother 1992;36:860-6.
  10. Neu HC, Chin NX. In vitro activity of the new fluoroquinolone CP 99-219. Antimicrob Agents Chemother 1994;38:2615-22.
  11. Woodcock JM, Andrews JM, Boswell FJ, Brenwald NP, Wise R. In vitro activity of Bay 12-8039, a new fluoroquinolone. Antimicrob Agents Chemother 1997;41:101-6.
  12. Fuchs PC, Barry AL, Pfaller MA, Allen SD, Gerlach EH. Multicenter evaluation of the in vitro activities of 3 new quinolones, sparfloxacin, CI-960, and PD 131-628, compared with the activity of ciprofloxacin against 5,252 clinical bacterial isolates. Antimicrob Agents Chemother 1991;35:764-6.
  13. Neu HC, Fang W, Gu JW, Chin NX. In vitro activity of OPC-17116. Antimicrob Agents Chemother 1992;36:1310-15.
  14. Imada T, Miyazaki S, Nishida M, Yamaguchi K, Goto S. In vitro and in vivo antibacterial activities of a new quinolone, OPC-17116. Antimicrob Agents Chemother 1992;36:573-9.
  15. Fuchs PC, Barry AL, Brown SD. In vitro activities of clinafloxacin against contemporary clinical bacterial isolates from 10 North American centers. Antimicrob Agents Chemother 1998;42:1274-7.
  16. Wakabayashi E, Mitsuhashi S. In vitro antibacterial activity of AM-1155, a novel 6-fluoro-8-methoxy quinolone. Antimicrob Agents Chemother 1994;38:594-601.
  17. Hosaka M, Yasue T, Fukuda H, Tomizawa H, Aoyama H, Hirai K. In vitro and in vivo antibacterial activities of AM-1155, a new 6-fluoro-8-methoxy quinolone. Antimicrob Agents Chemother 1992;36:2108-17.
  18. Ednie LM, Jacobs MR, Appelbaum PC. Comparative activities of ciprofloxacin against gram-positive and -negative bacteria. Antimicrob Agents Chemother 1998;42:1269-73.
  19. Felmingham D, Robbins MJ, Ingley K, et al. In vitro activity of trovafloxacin, a new fluoroquinolone, against recent clinical isolates. J Antimicrob Chemother 1997;39(suppl B):43-9.
  20. Cunha BA, Hussain Qadri SM, Ueno Y, Walters EA, Domenico P. Antibacterial activity of trovafloxacin against nosocomial gram-positive and gram-negative isolates. J Antimicrob Chemother 1997;39(suppl B):29-34.
  21. Brueggemann AB, Kugler KC, Doern GV. In vitro activity of Bay 12-8039, a novel 8-methoxyquinolone, compared to activities of six fluoroquinolones against S. pneumoniae, H. influenzae, and Moraxella catarrhalis. Antimicrob Agents Chemother 1997;41:1594-7.
  22. Barry AL, Fuchs PC. Antibacterial activities of grepafloxacin, ciprofloxacin, ofloxacin, and fleroxacin. J Chemother 1997;9:9-16.
  23. Hoogkamp-Korstanje JAA. In vitro activities of ciprofloxacin, levofloxacin, lomefloxacin, ofloxacin, pefloxacin, sparfloxacin, and trovafloxacin against gram-positive and gram-negative pathogens from respiratory tract infections. J Antimicrob Chemother 1997;40:427-31.
  24. Hoshino K, Kitamura A, Morrissey I, Sato K, Kato JI, Ikeda H. Comparison of inhibition of Escherichia coli topoisomerase IV by quinolones with DNA gyrase inhibition. Antimicrob Agents Chemother 1994;38:2623-7.
  25. Tanaka M, Onodera Y, Uchida Y, Sato K, Hayakawa I. Inhibitory activities of quinolones against DNA gyrase and topoisomerase IV purified from Staphylococcus aureus. Antimicrob Agents Chemother 1997;41:2362-6.
  26. Hooper DC. Bacterial topoisomerases, anti-topoisomerases, and anti-topoisomerase resistance. Clin Infect Dis 1998;27(suppl 1):S54-63.
  27. Wakebe H, Mitsuhashi S. Comparative in vitro activities of a new quinolone, OPC-17116, possessing potent activity against gram-positive bacteria. Antimicrob Agents Chemother 1992;36:2185-91.
  28. Wakebe H, Imada T, Yoneda H, et al. Evaluation of OPC-17116 against important pathogens that cause respiratory tract infections. Antimicrob Agents Chemother 1994;38:2340-5.
  29. Gooding BB, Erwin ME, Barrett MS, Johnson DM, Jones RN. Antimicrobial activities of two investigational fluoroquinolones (CI-960 and E-4695) against over 100 Legionella sp isolates. Antimicrob Agents Chemother 1992;36:2049-50.
  30. Jones RN. Activity of sparfloxacin (AT-4140), PD127391 and PD13168 against Legionella spp. J Antimicrob Chemother 1991;27:389-90.
  31. Ridgway GL, Salman H, Robbins MJ, Dencer C, Felmingham D. The in-vitro activity of grepafloxacin against Chlamydia spp., Mycoplasma spp., Ureaplasma urealyticum, and Legionella spp. J Antimicrob Chemother 1997;40(suppl A):31-4.
  32. Dubois J, St. Pierre C. An in vitro susceptibility study of gatifloxacin against Legionella species [abstr]. Presented at the 38th Interscience Conference on Antimicrobial Agents and Chemotherapy, San Diego, CA, September 24-27, 1998.
  33. Schulin T, Wennersten CB, Ferraro MJ, Moellering RC Jr, Eliopoulos GM. Susceptibilities of Legionella spp. to newer antimicrobials. Antimicrob Agents Chemother 1998;42:1520-3.
  34. Miyashita N, Niki Y, Kishimoto T, Nakajima M, Matsushima T. In vitro and in vivo activities of AM-1155, a new fluoroquinolone, against Chlamydia spp. Antimicrob Agents Chemother 1997;41:1331-4.
  35. Roblin PM, Kutlin A, Hammerschlag MR. In vitro activity of trovafloxacin against Chlamydia pneumoniae. Antimicrob Agents Chemother 1997;41:2033-4.
  36. Ishida K, Kaku M, Irifune K, et al. In-vitro and in-vivo activity of a new quinolone AM-1155 against Mycoplasma pneumoniae. J Antimicrob Chemother 1994;34:875-83.
  37. Bebear CM, Renaudin H, Boudjadja A, Bebear C. In vitro activity of Bay 12-8039, a new fluoroquinolone against mycoplasmas. Antimicrob Agents Chemother 1998;42:703-4.
  38. Kenny GE, Cartwright FD. Susceptibilities of Mycoplasma pneumoniae, Mycoplasma hominis, and Ureaplasma urealyticum to a new quinolone, trovafloxacin (CP-99,219). Antimicrob Agents Chemother 1996;40:1048-9.
  39. Kaku M, Ishida K, Irifune K, et al. In vitro and in vivo activities of sparfloxacin against Mycoplasma pneumoniae. Antimicrob Agents Chemother 1994;38:738-41.
  40. Spangler SK, Jacobs MR, Appelbaum PC. Susceptibility of anaerobic bacteria to trovafloxacin: comparison with other quinolones and non-quinolone antibiotics. Infect Dis Clin Pract 1996;5(suppl 3):S101-9.
  41. Goldstein EJC, Citron DM. Comparative activity of ciprofloxacin, ofloxacin, sparfloxacin, temafloxacin, CI-960, CI-990, and WIN-57273 against anaerobic bacteria. Antimicrob Agents Chemother 1992;36:1158-62.
  42. Kato N, Kato H, Tanaka-Bandoh K, Watanabe K, Ueno K. Comparative in-vitro and in-vivo activity of AM-1155 against anaerobic bacteria. J Antimicrob Chemother 1997;40:631-7.
  43. Nix DE, Spivey JM, Norman A, Schentag JJ. Dose-ranging pharmacokinetic study of ciprofloxacin after 200-, 300-, and 400-mg intravenous doses. Ann Pharmacother 1992;26:8-10.
  44. Shah A, Lettieri J, Kaiser L, Echols R, Heller AH. Comparative pharmacokinetics and safety of ciprofloxacin 400 mg iv thrice daily versus 750 mg p.o. twice daily. J Antimicrob Chemother 1994;33:795-801.
  45. Shah A, Lettieri J, Blum R, Millikin S, Sica D, Heller AH. Pharmacokinetics of intravenous ciprofloxacin in normal and renally impaired subjects. J Antimicrob Chemother 1996;38:103-16.
  46. Shah A, Lettieri J, Nix D, Wilton J, Heller AH. Pharmacokinetics of high-dose intravenous ciprofloxacin in young and elderly and in male and female subjects. Antimicrob Agents Chemother 1995;39:1003-6.
  47. Wolfson JS, Hooper DC. Comparative pharmacokinetics of ofloxacin and ciprofloxacin. Am J Med 1989;87(suppl 6C):31-6.
  48. Israel D, Gillum JG, Turik M, et al. Pharmacokinetics and serum bactericidal titers of ciprofloxacin and ofloxacin following multiple oral doses in healthy volunteers. Antimicrob Agents Chemother 1993;37:2193-9.
  49. Lode H, Hoffken G, Olschewski P, et al. Pharmacokinetics of ofloxacin after parenteral and oral administration. Antimicrob Agents Chemother 1987;31:1338-42.
  50. Kalager T, Digranes A, Bergan T, Rolstad T. Ofloxacin: serum and skin blister fluid pharmacokinetics in the fasting and non-fasting state. J Antimicrob Chemother 1986;17:795-800.
  51. Farinotti R, Trouvin JH, Bocquet V, Vermerie N, Carbon C. Pharmacokinetics of ofloxacin after single and multiple intravenous infusions in healthy subjects. Antimicrob Agents Chemother 1988;32:1590-2.
  52. Flor S. Pharmacokinetics of ofloxacin. Am J Med 1989; 87(suppl 6C):24-30.
  53. Fish DN, Chow AT. The clinical pharmacokinetics of levofloxacin. Clin Pharmacokinet 1997;32:101-19.
  54. Chien SC, Rogge MR, Gisclon LG, et al. Pharmacokinetic profile of levofloxacin following once-daily 500-mg oral or intravenous doses. Antimicrob Agents Chemother 1997;41: 2256-60.
  55. Wagstaff AJ, Balfour JA. Grepafloxacin. Drugs 1997;53: 817-24.
  56. Efthymiopoulos C, Bramer SL, Maroli A. Pharmacokinetics of grepafloxacin after oral administration of single and repeat doses in healthy young males. Clin Pharmacokinet 1997;33(suppl 1):1-8.
  57. Efthymiopoulos C, Bramer SL, Maroli A. Effect of age and gender on the pharmacokinetics of grepafloxacin. Clin Pharmacokinet 1997;33(suppl 1):9-17.
  58. Montay G, Bruno R, Verginol JC, et al. Pharmacokinetics of sparfloxacin in humans after single oral administration at doses of 200, 400, 600, and 800 mg. J Clin Pharmacol 1994;34: 1071-6.
  59. Montay G, Bruno R, Verginol JC, et al. Pharmacokinetics of sparfloxacin after repeated administration in healthy elderly volunteers [abstr]. Presented at the 31st Interscience Conference on Antimicrobial Agents and Chemotherapy, Chicago, September 29-October 2, 1991.
  60. Haria M, Lamb HM. Trovafloxacin. Drugs 1997;54:435-45.
  61. Teng R, Liston TE, Harris SC. Multiple-dose pharmacokinetics and safety of trovafloxacin in healthy volunteers. J Antimicrob Chemother 1996;37:955-63.
  62. Vincent J, Venitz J, Teng R, et al. Pharmacokinetics and safety of trovafloxacin in healthy male volunteers following administration of single intravenous doses of the prodrug, alatrovafloxacin. J Antimicrob Chemother 1997;39(suppl B):75-80.
  63. Nakashima M, Uematsu T, Kosuge K, et al. Single- and multiple-dose pharmacokinetics of Am-1155, a new 6-fluoro-8-methoxy quinolone, in humans. Antimicrob Agents Chemother 1995;39:2635-40.
  64. Bristol-Myers Squibb Co. Tequin (gatifloxacin) package insert. Princeton, NJ;December 1999.
  65. Bron NJ, Dorr MB, Mant TG, Webb CL, Vassos AB. The tolerance and pharmacokinetics of clinafloxacin (CI-960) in healthy subjects. J Antimicrob Chemother 1996;38:1023-9.
  66. Wise R, Jones S, Das I, Andrews JM. Pharmacokinetics and inflammatory fluid penetration of clinafloxacin. Antimicrob Agents Chemother 1998;42:428-30.
  67. Stass HH, Dietrich H, Sachse E. Influence of a four-times dosing of 500 mg probenecid on kinetics of Bay 12-8039 after administration of a single 400 mg dose in healthy male volunteers [abstr]. Presented at the 37th Interscience Conference on Antimicrobial Agents and Chemotherapy, Toronto, Canada, September 28-October 1, 1997.
  68. Kubitza D, Stass HH, Wingender W, Kuhlmann J. Bay 12-8039, a new 9-methoxy-quinolone: safety, tolerability, and steady state pharmacokinetics in healthy male volunteers [abstr]. Presented at the 36th Interscience Conference on Antimicrobial Agents and Chemotherapy, New Orleans, September 15-18, 1996.
  69. Stass HH, Kubitza D, Schuhly U. Bay 12-8039, a new 8-methoxyquinolone: pharmacokinetics, safety, and tolerability of single ascending intravenous doses in healthy male volunteers [abstr]. Presented at the 37th Interscience Conference on Antimicrobial Agents and Chemotherapy, Toronto, Canada, September 28-October 1, 1997.
  70. Randinitis EJ, Brodfuehrer J, Vassos AB. Pharmacokinetics of clinafloxacin following oral and intravenous single and multiple dosing in volunteers [abstr]. Presented at the 38th Interscience Conference on Antimicrobial Agents and Chemotherapy, San Diego, CA, September 24-27, 1998.
  71. Goa KL, Bryson HM, Markham A. Sparfloxacin: a review of its antibacterial activity, pharmacokinetic properties, clinical efficacy and tolerability in lower respiratory tract infections. Drugs 1997;53:700-25.
  72. Sullivan JT, Woodruff M, Lettieri J, Agarwal V, Krol G, Heller AH. Pharmacokinetics and tolerability of the new methoxyquinolone BAY 12-8039: 10 days' treatment at 400 mg daily [abstr]. Presented at the 7th European Congress of Clinical Microbiology and Infectious Diseases, Lausanne, Switzerland, May 25-28, 1997.
  73. Teng R, Harris SC, Nix DE, et al. Pharmacokinetics and safety of trovafloxacin (CP-99,219), a new quinolone antibiotic, following administration of single oral doses to healthy male volunteers. J Antimicrob Chemother 1995;36:385-94.
  74. Stass H, Dalhoff A, Kubitza D, Shuhly U. Pharmacokinetics, safety, and tolerability of ascending single doses of moxi-floxacin, a new 8-methoxy quinolone, administered to healthy subjects. Antimicrob Agents Chemother 1998;42:2060-5.
  75. Blaser J, Stone BB, Groner MC, Zinner SH. Comparative study with enoxacin and netilmicin in a pharmacodynamic model to determine importance of ratio of antibiotic peak concentration to MIC for bacterial activity and emergence of resistance. Antimicrob Agents Chemother 1987;31:1054-60.
  76. Madaras-Kelly KJ, Ostergaard BE, Hovde LB, Rotschafer JC. Twenty-four-hour area under the concentration-time curve/MIC ratio as a generic predictor of fluoroquinolone antimicrobial effect by using three strains of Pseudomonas aeruginosa and an in vitro pharmacodynamic model. Antimicrob Agents Chemother 1996;40:627-32.
  77. Leggett JE, Ebert S, Fantin B, Craig WA. Comparative dose-effect relations at several dosing intervals for beta-lactam, aminoglycoside and quinolone antibiotics against gram-negative bacilli in murine thigh infection and pneumonitis models. Scand J Infect Dis Suppl 1991;74:179-84.
  78. Andes DR, Craig WA. Pharmacodynamics of fluoroquinolones in experimental models of endocarditis. Clin Infect Dis 1998;27:47-50.
  79. Thomas JK, Forrest A, Bhavnani SM, et al. Pharmacodynamic evaluation of factors associated with the development of bacterial resistance in acutely ill patients during therapy. Antimicrob Agents Chemother 1998;42:521-7.
  80. Forrest A, Chodosh S, Amantea MA, Collins DA, Schentag JJ. Pharmacokinetics and pharmacodynamics of oral grepafloxacin in patients with acute bacterial exacerbations of chronic bronchitis. J Antimicrob Chemother 1997;40(suppl A):45-57.
  81. Hyatt JM, Nix DE, Schentag JJ. Pharmacokinetic and pharmacodynamic activities of ciprofloxacin against strains of S. pneumoniae, S. aureus, and P. aeruginosa for which MICs are similar. Antimicrob Agents Chemother 1994;38:2730-7.
  82. Craig W, Dalhoff A. Pharmacodynamics of fluoroquinolones in experimental animals. In: Kuhlman J, Dalhoff A, Zeiler HJ, eds. Handbook of experimental pharmacology. New York: Springer, 1998, 127:207-32.
  83. Watanabee Y, Ebert S, Craig W. AUC/MIC ratio is unifying parameter for comparison of in-vivo activity among fluoroquinolones [abstr]. Presented at the 32nd Interscience Conference on Antimicrobial Agents and Chemotherapy, Anaheim, CA, October 11-14, 1992.
  84. National Committee for Clinical Laboratory Standards. Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically, 4th ed, vol 17, no. 1. Villanova, PA: NCCLS, January 1997.
  85. Gisdon LG, Curtin CR, Chien SC, Williams R, Corrado M, Reichl V. The pharmacokinetics of levofloxacin in subjects with renal impairment, and in subjects receiving hemodialysis or CAPD [abstr]. Presented at the 36th Interscience Conference on Antimicrobial Agents and Chemotherapy, New Orleans, September 15-18, 1996.
  86. Lode H, Garau J, Grassi C, et al. Treatment of community-acquired pneumonia: a randomized comparison of sparfloxacin, amoxycillin-clavulanic acid and erythromycin. Eur Respir J 1995;8:1999-2007.
  87. Tremolieres F, de Kock F, Pluck N, Daniel R. Trovafloxacin versus high-dose amoxicillin (1 g three times daily) in the treatment of community-acquired pneumonia. Eur J Clin Microbiol Infect Dis 1998;17:447-53.
  88. File TM Jr, Segreti J, Dunbar L, et al. A multicenter, randomized study comparing the efficacy and safety of intravenous and/or oral levofloxacin versus ceftriaxone and/or cefuroxime axetil in treatment of adults with community-acquired pneumonia. Antimicrob Agents Chemother 1997;41:1965-72.
  89. O'Doherty B, Dutchman DA, Pettit R, Maroli A. Randomized, double-blind, comparative study of grepafloxacin and amoxycillin in the treatment of patients with community-acquired pneumonia. J Antimicrob Chemother 1997;40(suppl A):73-81.
  90. Onderdonk AB. Efficacy of trovafloxacin, a new fluoro-quinolone, in an animal model of intraabdominal sepsis. Infect Dis Clin Pract 1996;5(suppl 3):S117-19.
  91. Thadepalli H, Reddy U, Chuah SK, Thadepalli F, Malilay C, Gollapudi S. Effect of new quinolone, CP-99,219-27, in curing intraabdominal abscesses in rats. Infect Dis Clin Pract 1996;5(suppl 3):S120-1.
  92. Donahue PE, Smith DL, Yellin AE, Mintz SJ, Bur F, Luke DR. Trovafloxacin in the treatment of intra-abdominal infections: results of a double-blind, multicenter comparison with imipenem/cilastatin. Am J Surg 1998;176(suppl 6A):53S-61.


Advertisement

  Home   Site Map   Marketplace   My Medscape   CME Center   Feedback   Help Desk

Medscape Search Options
Select a database to search, enter a search term, then click “go.”    Advanced Search Forms

All material on this website is protected by copyright. Copyright © 1994-2000 by Medscape Inc. All rights reserved. This website also contains material copyrighted by 3rd parties. Medscape requires 3.x browsers or better from Netscape or Microsoft.