Abstract

The worldwide increase in antibiotic resistance is a concern for public health. The fact that the choice of dose and treatment duration can affect the selection of antibiotic-resistant mutants is becoming more evident, and an increased number of studies have used pharmacodynamic models to describe the drug exposure and pharmacodynamic breakpoints needed to minimize and predict the development of resistance. However, there remains a lack of sufficient data, and future work is needed to fully characterize these target drug concentrations. More knowledge is also needed of drug pharmacodynamics versus bacteria with different resistance mutations and susceptibility levels. The dosing regimens should exhibit high efficacy not only against susceptible wild-type bacteria but, preferably, also against mutated bacteria that may exist in low numbers in “susceptible” populations. Thus, to prolong the life span of existing and new antibiotics, it is important that dosing regimens be carefully selected on the basis of pharmacokinetic and pharmacodynamic properties that prevent emergence of preexisting and newly formed mutants.

Antibiotic Resistance

Antibiotic resistance has become a serious global problem and affects almost every bacterial species for which treatment with antibiotics is available. Resistance to multiple antibiotics has developed among many common pathogens, such as staphylococci, pneumococci, Pseudomonas organisms, and extended-spectrum β-lactamase (ESBL)-producing strains of Enterobacteriaceae, and the resistance problem is steadily increasing worldwide. Consequently, the therapeutic options for the treatment of some infections are limited, especially in developing countries, where second- and third-line antibiotics are unavailable or unaffordable. Infectious diseases cause >11 million deaths annually [1], and treatment failure caused by antibiotic-resistant bacteria is a contributing factor in these deaths, although the quantitative burden of antibiotic resistance is not certain.

The development and spread of antibiotic-resistant bacteria are affected by several factors. Some factors are bacteria specific, such as the mutation rate, the transmission rate, the biological fitness costs, and the ability to compensate for such costs. Other factors of major importance in the emergence of resistance are the volume and quality of antibiotic use, including prescription when there is no clinical indication, over-the-counter sales or sales by drug vendors, inappropriate drug choice, and suboptimal dosing [2, 3]. Dissemination of antibiotic resistance is also influenced by environmental factors in the community and hospitals. Direct or indirect person-to-person transmission is affected by population density and hospital structure and significantly increases in association with poor hygiene.

Mutation Rate

In a bacterial cell, spontaneous mutations normally occur at a rate of 10-9–10-10 mutations/bp/generation during DNA replication [4, 5]. In contrast to the mutation rate, the mutation frequency measures all of the mutants present in a given population, irrespective of when the mutations occurred. Because one antibiotic-resistance phenotype usually can be caused by several mutations, both in different base pairs and in different genes, the mutation frequency is generally higher than the general mutation rate. Bacteria with highly increased mutation rates are called “mutators” and can have mutation rates that are 10-fold to nearly 1000-fold higher than those of wild-type strains [6]. The mutator phenotype is often associated with inactivation of the methyl-directed mismatch repair, a repair system that recognizes mismatches on newly replicated DNA and restores the sequence with the correct base [7].

Because resistance development is very much dependent on mutations, the mutation frequency may play a significant role in the development of resistance. Studies have shown that mutators accelerate resistance development in vitro [8] and in animals [9] and that strains that are clinically fluoroquinolone resistant are often associated with a mutator phenotype [10]. In addition, Gustafsson et al. [11] presented evidence that antibiotic treatment selected for commensal bacteria with increased ciprofloxacin resistance and that there was a correlation between increased mutation frequency and resistance development.

Biological Cost of Antibiotic Resistance

Antibiotic resistance often confers a biological fitness cost, because the resistance mutations typically occur in genes of target molecules with essential functions in the cell. For example, mutations in the rpoB gene that encodes the β subunit of RNA polymerase cause rifampicin resistance in Escherichia coli and may also affect the rate of transcription [12]. Fusidic acid resistance caused by mutations in the ribosomal elongation factor G of Staphylococcus aureus [13] and Salmonella enterica serotype Typhimurium [14, 15] is often associated with a decreased rate of protein synthesis. These cost-associated mutations confer a reduction in bacterial fitness that can be measured as relative growth rates within and outside the host, transmission rates between hosts, and clearance from the hosts [16]. In normal settings, consequently, resistant bacteria usually are inferior to normal bacteria and can easily be outcompeted when no antibiotic pressure is present. For example, fosfomycin resistance, which can be conferred by mutations in several genes and by any loss-of-function mutations in these genes, is easily selected for E. coli under experimental conditions. However, fosfomycin resistance is rarely found in clinical isolates, in spite of the relatively high mutation frequency in vitro. This can be explained by the fact that fosfomycin resistance is often associated with a reduced bacterial growth rate. A mathematical model showed that such fitness costs rapidly reduced the probability of development of fosfomycin resistance, because they prevented the establishment of resistant bacteria in the bladder [17]. In a human competition model, it has also been shown that biological fitness is a significant determinant of bacterial survival [18].

Compensatory Evolution

Bacteria may reduce the biological cost associated with resistance through compensatory evolution. Such compensation can involve true reversion of the resistance mutation or acquisition of second-site compensatory mutations (intra- or extragenic suppressor mutations). Amelioration of the costs of resistance by compensatory mutations has been noted for several antibiotics and bacterial species in vitro, in animal experiments, and in clinical settings [19]. For example, a number of compensatory intragenic mutations have partly or fully restored the biological fitness in fusidic acid-resistant mutants of S. aureus and S. Typhimurium [13, 15], rifampin-resistant mutants of E. coli [12], and clinical isolates of isoniazid-resistant Mycobacterium tuberculosis [20]. A study of the epidemiology of M. tuberculosis has shown that, among clinical isolates with streptomycin resistance, strains with a low biological fitness cost are more frequent [21]. It has been shown that resistant bacteria can persist in humans for a long time without any antibiotic pressure [22, 23]. In one study, Staphylococcus epidermidis isolates with high-level resistance to clarithromycin were recovered from patients up to 4 years after antibiotic treatment [23]. Antibiotic resistance can, thus, be stabilized with fitness-restoring compensatory mutations and may allow fully resistant strains to compete successfully with susceptible strains in an antibiotic-free environment [24]. Consequently, restrictions in antibacterial use might not lead to reversibility of antibiotic resistance in the community.

Dosage-Related Selection

The fact that resistance development can be affected by antibiotic dosing regimens is evident from the results of a number of in vitro and animal experiments. A few selected examples will be given here. In a study by Blaser et al. [25], Klebsiella pneumoniae was challenged with the quinolone enoxacin in different dosing regimens in an in vitro kinetic model. When enoxacin was administered in a twice-daily dosage, bacteria with increased MICs were selected. When, instead, the total daily dose was given as 1 dose daily, the bacteria were eradicated. The need for high peak fluoroquinolone levels for the suppression of resistant bacteria was also noted by Marchbanks et al. [26], who exposed Pseudomonas aeruginosa to ciprofloxacin in an in vitro model. A single dose prevented growth of the ciprofloxacin-resistant bacteria that were selected during exposure to smaller fractionated doses [26]. The impact of dose fractionation on survival was also investigated in a neutropenic rat model of P. aeruginosa sepsis. Once-daily administration of the fluoroquinolone lomefloxacin gave a higher survival rate for the rats, compared with the use of smaller doses administered after shorter intervals [27]. In an in vivo abscess model, dose-dependent selection of mutants of Enterobacter cloacae was demonstrated after treatment with ceftizoxime [28]. When healthy pigs were infected with Salmonella organisms and were treated orally with the recommended dose of the fluoroquinolone enrofloxacin (2.5 mg/kg of body weight), resistant Salmonella organisms were rapidly selected. By instead using the intramuscular route of administration and by escalating the dose to 3 or 6 times the recommended dose, resistance selection was reduced [29]. The association between β-lactam use and penicillin-resistant Streptococcus pneumoniae was analyzed in an observational study of schoolchildren. Low daily doses and a long duration of treatment with a β-lactam antibiotic were associated with an increased risk of pharyngeal carriage of penicillin-resistant S. pneumoniae [30].

Clinical and experimental studies thus indicate that the choice of dose and treatment duration can influence the selection of antibiotic-resistant mutants. For example, the initially recommended dose of levofloxacin has been questioned on the basis of clinical experience indicating that selection of resistance can take place during treatment of infection with S. pneumoniae [31]. Still, there is poor knowledge of the optimal dosing strategies to treat bacterial infections while simultaneously preventing the selection and emergence of resistance.

Selective Concentrations

During antibiotic therapy, gradients of antibiotic concentrations are formed in the human body. These gradients will depend on the rates of drug diffusion into various tissues and the variation in the rate of elimination from different compartments (figure 1). In the body, diverse environmental antibiotic selective pressures will, thus, be created, and the selection process can be expected to differ accordingly.

Figure 1

Multicompartment pharmacokinetics. Antibiotic concentrations in the blood will be transported to the interstitial fluid of different body sites, such as those in the figure. The differences in pharmacokinetics within the different sites will depend on the rate of diffusion, potential diffusion barriers, and the volume of compartments.

A subpopulation of resistant bacteria often exists and will be selected by certain drug concentrations, leading to a regrowth during treatment [32] (figure 2). Low antibiotic concentrations can select for low-level resistance, which could have a major effect on the emergence of high-level antibiotic resistance [33, 34]. During antibiotic treatment, selection of resistance may take place at several sites in the human body. In the evaluation of optimal drug concentrations, it is important to focus not only on the infective pathogens and the infectious sites of the body but, also, on the commensal flora (e.g., in the intestinal tract or the skin), in which much higher numbers of bacteria exist and which perhaps is even more important in the selection of resistance. When the fluoroquinolone clinafloxacin was administered to healthy humans, ecological disturbances in the intestinal microflora were created. The treatment effectively eradicated aerobic bacteria in most subjects, whereas the effect on anaerobic bacteria was somewhat less effective and resistance was noted to develop in Bacteroides species [35].

Figure 2

Emergence of resistance. The susceptible (wild-type) population is readily killed after drug exposure, whereas the resistant (mutant) population may be amplified. The total population is explained by the changes in the 2 different populations. Adapted from Drusano [32], with permission from the Nature Publishing Group.

However, because of these large fluctuations in drug concentrations and because of the difficulties associated with accurately measuring the concentrations at some sites, the actual drug concentrations are often uncertain. To simplify this complex system, in vitro models in which serum concentrations can be simulated are increasingly being used. It has been shown that the free (nonprotein-bound) concentrations in interstitial tissue fluid, where most bacterial infections take place, are similar to those in serum [36–38].

Pharmacokinetics (Pk) and Pharmacodynamics (Pd)

During the past decades, the knowledge of the interaction between the PK and PD of antibiotics has significantly increased, and the International Society of Anti-Infective Pharmacology has proposed standardization of PK/PD terminology [39, 40]. However, studies of antibiotic dosing regimens that consider the development of resistance have not been of high priority, and most PK/PD indices thus focus on clinical or microbiological efficacy. Not until recently has some research included resistance development as an end point, and several studies, especially studies of fluoroquinolones, have used PD models in the search for PK/PD principles and breakpoints that predict the emergence of resistance. Another important issue of concern is that most PK/PD studies include susceptible, wild-type bacteria as target organisms. More knowledge is needed of drug pharmacodynamics versus bacteria with different resistance mutations and susceptibility levels.

Resistance Prevention Studies

End points. To investigate the optimal dosing regimens and to determine the breakpoints needed for prevention of resistance, different end points can be used. A simple method to measure resistance and detect changes in susceptibility is MIC determination. The disadvantage of MIC determination is that often only 1 or 2 colonies are tested, and minor resistant subpopulations may, thus, not be detected. A more sensitive method, cultivation of bacterial samples on agar plates containing antibiotics, makes it possible to measure the changes in the number of resistant bacteria during experiments [25, 26, 41–43]. Another method of studying resistance selection is through the use of competition assays that use strains with different drug susceptibilities. The different populations can be measured using a discriminative marker; the presence of a mutation in the arabinose gene results in a different colony color on a certain agar and allows for the simple identification of each strain in a mixed culture [34, 44]. Measurement of resistance development in the clinical setting is, as for efficacy studies, limited by discontinuous end points. Ideally, successive samples for cultures should be obtained during treatment and the results related to the individual drug exposure, but this is most often not possible. Therefore, studies of resistance development during treatment of patients with antibiotics have used single measurements determined before treatment and either at the end of treatment or at clinical failure.

Resistance prevention predictors. An increased number of studies have attempted to describe the drug exposure needed to minimize resistance development. Thomas et al. [45] retrospectively evaluated 107 patients treated in 4 clinical trials of nosocomial lower respiratory tract infection, and they found that the selection of antibiotic resistance among the pathogens was significantly decreased when the area under the concentration-time curve (AUC)/MIC was ⩾100. These results have been criticized because of the great time differential between the studies and because of combination in the analysis of drugs that have different pharmacodynamic indices for efficacy [46]. Using mixed cultures of susceptible, intermediately susceptible, and resistant pneumococci in an in vitro kinetic model, Odenholt et al. [47] studied resistance selection after exposure to different concentrations of benzylpenicillin. Although the study design did not allow evaluation of the exact time above the MIC needed to suppress mutant growth, no regrowth was noted when the time above the MIC was 48%, 38%, and 46% for the penicillin-susceptible, -intermediately susceptible, and -resistant subpopulations, respectively.

The hypothesis that the selection of resistant bacteria is driven by particular selective antibiotic concentrations was first suggested by Baquero et al. [33, 34, 48]. They predicted that there would be a range of concentrations for which selection of strains with increased levels of resistance to a drug would be the most intense, and they called this range a “selective window” [34]. Using the same bacterial strains, we have confirmed, in an in vitro kinetic model, that the duration of antibiotic concentrations within the selective window affected the selection of resistant bacteria [44].

The mutant prevention concentration (MPC) is a relatively new pharmacodynamic parameter that has been introduced as a measure of a drug's resistance prevention potency. MPC is defined as the lowest antibiotic concentration that prevents growth of the least susceptible single-step mutants. Resistant subpopulations are proposed to be selectively enriched in the mutant selection window (MSW)—that is, the drug concentration range between the MIC and the MPC [49, 50], as described in figure 3. The methodology of MPC determination has not been standardized; however, in most experimental designs, a bacterial population of 1010 cells is applied to agar plates containing antibiotics, and the MPC is the minimum concentration that allows no colony recovery. For bacteria to grow in the presence of antibiotic concentrations above the MPC, ⩾2 resistance mutations have to arise. If the mutation frequency is ⩽10-7 mutations, then a bacterial population of >1014 cells is needed for 2 mutations to arise, a population size that is unlikely to be achieved in most clinical infections [51].

Figure 3

Illustration of the drug concentration over time and the concept of the mutant selection window (MSW). The hypothesis is that selection of resistant mutants occurs when drug concentrations are within the MSW (shaded area)—that is, between the MIC and the mutant prevention concentration (MPC). T > MPC, time above the MPC; TMSW, time within the MSW.

The MPC corresponds to the MIC of the least drug-susceptible single-step mutant in a population. It has been questioned whether the MPC can be predicted from the MIC. In a study by Marcusson et al. [52], the MICs and MPCs of ciprofloxacin for 22 clinical urinary tract infection E. coli isolates were measured. Comparing the MICs and MPCs showed only weak correlation (e.g., there were several isolates with low MICs and high MPCs) (figure 4). This poor correlation between the MIC and the MPC has been reported for a variety of fluoroquinolones and different bacteria [53]. Mutations that can lead to resistance may arise in many different combinations, and the correlation (or lack of correlation) between the MIC and the MPC is probably dependent on where the mutation is likely to arise. In S. pneumoniae, it has been shown that the presence of a parC mutation increases the MPC for fluoroquinolones [54]. A consequence of the variation in the MPC relative to the MIC is that the MPC cannot be accurately predicted from the MIC on an individual basis but, rather, has to be measured separately. This implies that, at present, MPC determination might be more applicable during the process of antibiotic development than in clinical practice. To be used in the latter context, MPC methodology needs to be further developed and simplified.

Figure 4

MICs and mutant prevention concentrations (MPCs) of ciprofloxacin for 22 clinical urinary tract infection Escherichia coli isolates [52].

The concept of the MSW is not equally applicable to all antibiotic-organism combinations. For MPC determination to be useful in the approach to restricting the selection of antibacterial resistance, the resistance mechanisms observed in the in vitro MPC studies must be the same as the resistance mechanisms observed in vivo. The MPC has mostly been studied for antibiotics for which resistance primarily develops by stepwise chromosomal point mutations, especially the fluoroquinolones. The application of MPC determination to other drugs has raised questions regarding the relevance of mutational events for such drugs as the β-lactams and the aminoglycosides and whether MPC measurements can be performed for drugs with other resistance mechanisms (e.g., efflux and β-lactamases) [55–57]. The significance of these concerns is based on whether the MSW hypothesis addresses the first step in resistance development (i.e., spontaneous mutations or horizontal DNA acquisition) or the second step (i.e., the selective enrichment of mutant subpopulations) [56]. If the second step is the most important, then the individual resistance mechanisms are of less relevance for the MSW concept.

In theory, drug dosing that yields concentrations above the MPC during the whole dosing interval would prevent resistance selection. This could possibly be achieved by using higher doses and/or shorter dosing intervals than would generally be needed to cure an infection. Furthermore, the time within the MSW can be narrowed using compounds with small differences between the MIC and the MPC or compounds that quickly pass through the MSW. However, for some currently available drugs, concentrations above the MPC cannot be reached in patients because of reasons related to PK or toxicity limits. Like the MIC, the MPC is measured with static concentrations. When evaluating dosing regimens from a resistance prevention perspective, it is important to consider the PK properties of a drug. Thus, different PK/PD indices, such as the AUC/MPC, the maximum drug concentration/MPC, and the time within the MSW, have been investigated for their propensity to correlate with antibiotic resistance in vitro and in vivo [41, 42, 58–64]. Zinner et al. [41] tested the theory of the MSW by use of moxifloxacin and S. pneumoniae, and they showed that an AUC/MIC <10 h and >100 h, which represented a time within the MSW of <20%, prevented resistance. Also, when S. aureus was exposed to 4 fluoroquinolones, no resistant bacteria were observed with an AUC/MIC >200 h and a time within the MSW of <20% [62]. Thus, the drug concentrations may not have to exceed the MPC for the entire dosing interval to prevent resistance development.

A study showed that the time above the MPC needed to prevent ciprofloxacin resistance in E. coli was shorter when the maximum drug concentration was increased, and the PK/PD index that was best associated with resistance development was the AUC/MPC [64]. The potential use of the AUC/MPC as a predictor of resistance development has also been suggested in a study exposing S. aureus to different fluoroquinolones [42]. of the MPC-related indices tested, the AUC/MPC was the only one found to correlate with the prevention of fluoroquinolone resistance. Thus, MPC determination and appropriate PK/PD indices could possibly be of use in making decisions about the dosing regimen with respect to a given dosing regimen's potential for the selection and enrichment of mutants. Among antibiotic-resistant mutants isolated from patients, there is a great variation in both the MIC and the MPC [10, 52], and the target breakpoint could vary significantly depending on these values. However, because of the lack of consistency in the ratio of the MIC to the MPC for individual bacterial strains, the use of the MPC as a denominator in these target PD indices would probably lead to less variability.

Implications For Clinical Practice and Drug Development

Antibiotic resistance is of concern for public health, and the rapid emergence of resistant bacteria has led to an increasing awareness of the problem. Still, strict regulations concerning antibiotic use are lacking, and the development of new antibiotics is becoming increasingly expensive and difficult. It is well known that antibiotic resistance can be selected during antibiotic treatment [31, 45, 65]; selection takes place both at the site of infection and in the commensal flora. However, the association between drug dosage and resistance development has been a neglected research area. To prolong the life span of existing and new antibiotics, it is of utmost importance that the dosing regimens are carefully selected on the basis of the PK/PD properties that prevent emergence of preexisting or newly formed mutants. In future drug development, other strategies to prevent resistance development should also be included. One such strategy should be to identify targets for which the resistance mechanism has the most negative effect on fitness. An ideal antibiotic would be a drug for which resistance mutations are rare, the fitness cost of resistance is high, and the rate of fitness-restoring compensatory mutations is low.

To prevent and/or minimize resistance, dosing regimens should exhibit high efficacy to susceptible wild-type bacteria but, preferably, also to mutated bacteria with different degrees of resistance that may exist in low numbers. To achieve useful results, the resistance mechanisms studied in vitro should be the same as the resistance that is expressed in vivo. When an optimal resistance-preventing dosage regimen is limited by the PK and toxicity of the drug, this may be circumvented using combination therapy. The concurrent administration of 2 antibiotics of different classes would require 2 concurrent mutations for bacterial growth to occur. For M. tuberculosis, concentrations above the MPC cannot be attained for most of the first-line agents [66]. In the case of tuberculosis, treatment duration is long, and, to minimize the risk of resistance, 2–4 drugs usually are prescribed.

Although the number of studies that have demonstrated a PD target to prevent the emergence of resistance have been increasing during the past few years, the knowledge gained is far from complete (e.g., only a few studies have addressed the fact that an antibiotic dosage that may prevent the emergence of resistance in the target pathogen may have a collateral effect in the commensal flora, where other pathogens may be selected). However, it is becoming obvious that dosage regimens, on the basis of studies of the drug exposure needed for eradication of the main (wild-type) bacterial population, seem to be clearly associated with a risk of selecting preexisting or newly formed mutants. Thus, higher dosages may be needed to slow down resistance development. Standardization of methods and future studies are needed to define such drug exposure targets for different classes of antibiotics versus different pathogens. These issues should be addressed by academic researchers, by the pharmaceutical industry during drug development, and by regulatory authorities.

Acknowledgments

Supplement sponsorship. This article was published as part of a supplement entitled “Annual Conference on Antimicrobial Resistance,” sponsored by the National Foundation for Infectious Diseases.

Potential conflicts of interest. S.K.O. and O.C.: no conflicts.

References

1
World Health Organization
Shaping the future, World Health report 2003. 2003
 
2
Ball
P
Baquero
F
Cars
O
, et al. 
Antibiotic therapy of community respiratory tract infections: strategies for optimal outcomes and minimized resistance emergence
J Antimicrob Chemother
2002
, vol. 
49
 (pg. 
31
-
40
)
3
DeRyke
CA
Lee
SY
Kuti
JL
Nicolau
DP
Optimising dosing strategies of antibacterials utilising pharmacodynamic principles: impact on the development of resistance
Drugs
2006
, vol. 
66
 (pg. 
1
-
14
)
4
Andersson
DI
Hughes
D
Muller's ratchet decreases fitness of a DNA-based microbe
Proc Natl Acad Sci U S A
1996
, vol. 
93
 (pg. 
906
-
7
)
5
Drake
JW
Charlesworth
B
Charlesworth
D
Crow
JF
Rates of spontaneous mutation
Genetics
1998
, vol. 
148
 (pg. 
1667
-
86
)
6
Hall
LM
Henderson-Begg
SK
Hypermutable bacteria isolated from humans—a critical analysis
Microbiology
2006
, vol. 
152
 (pg. 
2505
-
14
)
7
Miller
JH
Spontaneous mutators in bacteria: insights into pathways of mutagenesis and repair
Annu Rev Microbiol
1996
, vol. 
50
 (pg. 
625
-
43
)
8
Schaaff
F
Reipert
A
Bierbaum
G
An elevated mutation frequency favors development of vancomycin resistance in Staphylococcus aureus
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
3540
-
8
)
9
Giraud
A
Matic
I
Radman
M
Fons
M
Taddei
F
Mutator bacteria as a risk factor in treatment of infectious diseases
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
863
-
5
)
10
Komp Lindgren
P
Karlsson
A
Hughes
D
Mutation rate and evolution of fluoroquinolone resistance in Escherichia coli isolates from patients with urinary tract infections
Antimicrob Agents Chemother
2003
, vol. 
47
 (pg. 
3222
-
32
)
11
Gustafsson
I
Sjölund
M
Torell
E
, et al. 
Bacteria with increased mutation frequency and antibiotic resistance are enriched in the commensal flora of patients with high antibiotic usage
J Antimicrob Chemother
2003
, vol. 
52
 (pg. 
645
-
50
)
12
Reynolds
MG
Compensatory evolution in rifampin-resistant Escherichia coli
Genetics
2000
, vol. 
156
 (pg. 
1471
-
81
)
13
Nagaev
I
Björkman
J
Andersson
DI
Hughes
D
Biological cost and compensatory evolution in fusidic acid-resistant Staphylococcus aureus
Mol Microbiol
2001
, vol. 
40
 (pg. 
433
-
9
)
14
Johanson
U
Aevarsson
A
Liljas
A
Hughes
D
The dynamic structure of EF-G studied by fusidic acid resistance and internal revertants
J Mol Biol
1996
, vol. 
258
 (pg. 
420
-
32
)
15
Björkman
J
Nagaev
I
Berg
OG
Hughes
D
Andersson
DI
Effects of environment on compensatory mutations to ameliorate costs of antibiotic resistance
Science
2000
, vol. 
287
 (pg. 
1479
-
82
)
16
Andersson
DI
Levin
BR
The biological cost of antibiotic resistance
Curr Opin Microbiol
1999
, vol. 
2
 (pg. 
489
-
93
)
17
Nilsson
AI
Berg
OG
Aspevall
O
Kahlmeter
G
Andersson
DI
Biological costs and mechanisms of fosfomycin resistance in Escherichia coli
Antimicrob Agents Chemother
2003
, vol. 
47
 (pg. 
2850
-
8
)
18
Gustafsson
I
Cars
O
Andersson
DI
Fitness of antibiotic resistant Staphylococcus epidermidis assessed by competition on the skin of human volunteers
J Antimicrob Chemother
2003
, vol. 
52
 (pg. 
258
-
63
)
19
Björkman
J
Andersson
DI
The cost of antibiotic resistance from a bacterial perspective
Drug Resist Updat
2000
, vol. 
3
 (pg. 
237
-
45
)
20
Sherman
DR
Mdluli
K
Hickey
MJ
, et al. 
Compensatory ahpC gene expression in isoniazid-resistant Mycobacterium tuberculosis
Science
1996
, vol. 
272
 (pg. 
1641
-
3
)
21
Sander
P
Springer
B
Prammananan
T
, et al. 
Fitness cost of chromosomal drug resistance-conferring mutations
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
1204
-
11
)
22
Sjölund
M
Wreiber
K
Andersson
DI
Blaser
MJ
Engstrand
L
Long-term persistence of resistant Enterococcus species after antibiotics to eradicate Helicobacter pylori
Ann Intern Med
2003
, vol. 
139
 (pg. 
483
-
7
)
23
Sjölund
M
Tano
E
Blaser
MJ
Andersson
DI
Engstrand
L
Persistence of resistant Staphylococcus epidermidis after single course of clarithromycin
Emerg Infect Dis
2005
, vol. 
11
 (pg. 
1389
-
93
)
24
Björkman
J
Hughes
D
Andersson
DI
Virulence of antibiotic-resistant Salmonella typhimurium
Proc Natl Acad Sci USA
1998
, vol. 
95
 (pg. 
3949
-
53
)
25
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 bactericidal activity and emergence of resistance
Antimicrob Agents Chemother
1987
, vol. 
31
 (pg. 
1054
-
60
)
26
Marchbanks
CR
McKiel
JR
Gilbert
DH
, et al. 
Dose ranging and fractionation of intravenous ciprofloxacin against Pseudomonas aeruginosa and Staphylococcus aureus in an in vitro model of infection
Antimicrob Agents Chemother
1993
, vol. 
37
 (pg. 
1756
-
63
)
27
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
, vol. 
37
 (pg. 
483
-
90
)
28
Stearne
LE
van Boxtel
D
Lemmens
N
Goessens
WH
Mouton
JW
Gyssens
IC
Comparative study of the effects of ceftizoxime, piperacillin, and piperacillin-tazobactam concentrations on antibacterial activity and selection of antibiotic-resistant mutants of Enterobacter cloacae and Bacteroides fragilis in vitro and in vivo in mixed-infection abscesses
Antimicrob Agents Chemother
2004
, vol. 
48
 (pg. 
1688
-
98
)
29
Wiuff
C
Lykkesfeldt
J
Svendsen
O
Aarestrup
FM
The effects of oral and intramuscular administration and dose escalation of enrofloxacin on the selection of quinolone resistance among Salmonella and coliforms in pigs
Res Vet Sci
2003
, vol. 
75
 (pg. 
185
-
93
)
30
Guillemot
D
Carbon
C
Balkau
B
, et al. 
Low dosage and long treatment duration of β-lactam: risk factors for carriage of penicillin-resistant Streptococcus pneumoniae
JAMA
1998
, vol. 
279
 (pg. 
365
-
70
)
31
Davidson
R
Cavalcanti
R
Brunton
JL
, et al. 
Resistance to levofloxacin and failure of treatment of pneumococcal pneumonia
N Engl J Med
2002
, vol. 
346
 (pg. 
747
-
50
)
32
Drusano
GL
Antimicrobial pharmacodynamics: critical interactions of “bug and drug”
Nat Rev Microbiol
2004
, vol. 
2
 (pg. 
289
-
300
)
33
Baquero
F
Negri
MC
Morosini
MI
Blazquez
J
The antibiotic selective process: concentration-specific amplification of low-level resistant populations
Ciba Found Symp
1997
, vol. 
207
 (pg. 
93
-
105
discussion 105-11
34
Negri
MC
Lipsitch
M
Blazquez
J
Levin
BR
Baquero
F
Concentration-dependent selection of small phenotypic differences in TEM β-lactamase-mediated antibiotic resistance
Antimicrob Agents Chemother
2000
, vol. 
44
 (pg. 
2485
-
91
)
35
Oh
H
Nord
CE
Barkholt
L
Hedberg
M
Edlund
C
Ecological disturbances in intestinal microflora caused by clinafloxacin, an extended-spectrum quinolone
Infection
2000
, vol. 
28
 (pg. 
272
-
7
)
36
Brunner
M
Derendorf
H
Muller
M
Microdialysis for in vivo pharmacokinetic/pharmacodynamic characterization of anti-infective drugs
Curr Opin Pharmacol
2005
, vol. 
5
 (pg. 
495
-
9
)
37
Cars
O
Pharmacokinetics of antibiotics in tissues and tissue fluids: a review
Scand J Infect Dis Suppl
1990
, vol. 
74
 (pg. 
23
-
33
)
38
Cars
O
Ogren
S
Antibiotic tissue concentrations: methodological aspects and interpretation of results
Scand J Infect Dis Suppl
1985
, vol. 
44
 (pg. 
7
-
15
)
39
Mouton
JW
Dudley
MN
Cars
O
Derendorf
H
Drusano
GL
Standardization of pharmacokinetic/pharmacodynamic (PK/PD) terminology for anti-infective drugs
Int J Antimicrob Agents
2002
, vol. 
19
 (pg. 
355
-
8
)
40
Mouton
JW
Dudley
MN
Cars
O
Derendorf
H
Drusano
GL
Standardization of pharmacokinetic/pharmacodynamic (PK/PD) terminology for anti-infective drugs: an update
J Antimicrob Chemother
2005
, vol. 
55
 (pg. 
601
-
17
)
41
Zinner
SH
Lubenko
IY
Gilbert
D
, et al. 
Emergence of resistant Streptococcus pneumoniae in an in vitro dynamic model that simulates moxifloxacin concentrations inside and outside the mutant selection window: related changes in susceptibility, resistance frequency and bacterial killing
J Antimicrob Chemother
2003
, vol. 
52
 (pg. 
616
-
22
)
42
Allen
GP
Kaatz
GW
Rybak
MJ
In vitro activities of mutant prevention concentration-targeted concentrations of fluoroquinolones against Staphylococcus aureus in a pharmacodynamic model
Int J Antimicrob Agents
2004
, vol. 
24
 (pg. 
150
-
60
)
43
MacGowan
AP
Rogers
CA
Holt
HA
Bowker
KE
Activities of moxifloxacin against, and emergence of resistance in, Streptococcus pneumoniae and Pseudomonas aeruginosa in an in vitro pharmacokinetic model
Antimicrob Agents Chemother
2003
, vol. 
47
 (pg. 
1088
-
95
)
44
Olofsson
SK
Geli
P
Andersson
DI
Cars
O
Pharmacodynamic model to describe the concentration-dependent selection of cefotaxime-resistant Escherichia coli
Antimicrob Agents Chemother
2005
, vol. 
49
 (pg. 
5081
-
91
)
45
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
, vol. 
42
 (pg. 
521
-
7
)
46
Drusano
GL
Prevention of resistance: a goal for dose selection for antimicrobial agents
Clin Infect Dis
2003
, vol. 
36
 
Suppl 1
(pg. 
S42
-
50
)
47
Odenholt
I
Gustafsson
I
Löwdin
E
Cars
O
Suboptimal antibiotic dosage as a risk factor for selection of penicillin-resistant Streptococcus pneumoniae: in vitro kinetic model
Antimicrob Agents Chemother
2003
, vol. 
47
 (pg. 
518
-
23
)
48
Negri
MC
Morosini
MI
Loza
E
Baquero
F
In vitro selective antibiotic concentrations of β-lactams for penicillin-resistant Streptococcus pneumoniae populations
Antimicrob Agents Chemother
1994
, vol. 
38
 (pg. 
122
-
5
)
49
Zhao
X
Drlica
K
Restricting the selection of antibiotic-resistant mutants: a general strategy derived from fluoroquinolone studies
Clin Infect Dis
2001
, vol. 
33
 
Suppl 3
(pg. 
S147
-
56
)
50
Drlica
K
Zhao
X
Mutant selection window hypothesis updated
Clin Infect Dis
2007
, vol. 
44
 (pg. 
681
-
8
)
51
Low
DE
Antimicrobial drug use and resistance among respiratory pathogens in the community
Clin Infect Dis
2001
, vol. 
33
 
Suppl 3
(pg. 
S206
-
13
)
52
Marcusson
LL
Olofsson
SK
Komp Lindgren
P
Cars
O
Hughes
D
Mutant prevention concentrations of ciprofloxacin for urinary tract infection isolates of Escherichia coli
J Antimicrob Chemother
2005
, vol. 
55
 (pg. 
938
-
43
)
53
Drlica
K
Zhao
X
Blondeau
JM
Hesje
C
Low correlation between MIC and mutant prevention concentration
Antimicrob Agents Chemother
2006
, vol. 
50
 (pg. 
403
-
4
)
54
Smith
HJ
Walters
M
Hisanaga
T
Zhanel
GG
Hoban
DJ
Mutant prevention concentrations for single-step fluoroquinolone-resistant mutants of wild-type, efflux-positive, or ParC or GyrA mutation-containing Streptococcus pneumoniae isolates
Antimicrob Agents Chemother
2004
, vol. 
48
 (pg. 
3954
-
8
)
55
Smith
HJ
Nichol
KA
Hoban
DJ
Zhanel
GG
Stretching the mutant prevention concentration (MPC) beyond its limits
J Antimicrob Chemother
2003
, vol. 
51
 (pg. 
1323
-
5
)
56
Zhao
X
Clarification of MPC and the mutant selection window concept
J Antimicrob Chemother
2003
, vol. 
52
 pg. 
731
  
author reply 732-3
57
Livermore
DM
Overstretching the mutant prevention concentration
J Antimicrob Chemother
2003
, vol. 
52
 pg. 
732
  
author reply 732-3
58
Dong
Y
Zhao
X
Domagala
J
Drlica
K
Effect of fluoroquinolone concentration on selection of resistant mutants of Mycobacterium bovis BCG and Staphylococcus aureus
Antimicrob Agents Chemother
1999
, vol. 
43
 (pg. 
1756
-
8
)
59
Drlica
K
The mutant selection window and antimicrobial resistance
J Antimicrob Chemother
2003
, vol. 
52
 (pg. 
11
-
7
)
60
Blondeau
JM
Zhao
X
Hansen
G
Drlica
K
Mutant prevention concentrations of fluoroquinolones for clinical isolates of Streptococcus pneumoniae
Antimicrob Agents Chemother
2001
, vol. 
45
 (pg. 
433
-
8
)
61
Allen
GP
Kaatz
GW
Rybak
MJ
Activities of mutant prevention concentration-targeted moxifloxacin and levofloxacin against Streptococcus pneumoniae in an in vitro pharmacodynamic model
Antimicrob Agents Chemother
2003
, vol. 
47
 (pg. 
2606
-
14
)
62
Firsov
AA
Vostrov
SN
Lubenko
IY
Drlica
K
Portnoy
YA
Zinner
SH
In vitro pharmacodynamic evaluation of the mutant selection window hypothesis using four fluoroquinolones against Staphylococcus aureus
Antimicrob Agents Chemother
2003
, vol. 
47
 (pg. 
1604
-
13
)
63
Cui
J
Liu
Y
Wang
R
Tong
W
Drlica
K
Zhao
X
The mutant selection window in rabbits infected with Staphylococcus aureus
J Infect Dis
2006
, vol. 
194
 (pg. 
1601
-
8
)
64
Olofsson
SK
Marcusson
LL
Komp Lindgren
P
Hughes
D
Cars
O
Selection of ciprofloxacin resistance in Escherichia coli in an in vitro kinetic model: relation between drug exposure and mutant prevention concentration
J Antimicrob Chemother
2006
, vol. 
57
 (pg. 
1116
-
21
)
65
Fish
DN
Piscitelli
SC
Danziger
LH
Development of resistance during antimicrobial therapy: a review of antibiotic classes and patient characteristics in 173 studies
Pharmacotherapy
1995
, vol. 
15
 (pg. 
279
-
91
)
66
Dong
Y
Zhao
X
Kreiswirth
BN
Drlica
K
Mutant prevention concentration as a measure of antibiotic potency: studies with clinical isolates of Mycobacterium tuberculosis
Antimicrob Agents Chemother
2000
, vol. 
44
 (pg. 
2581
-
4
)

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.