Sunteți pe pagina 1din 70

Expert Opinion on Drug Metabolism & Toxicology

ISSN: 1742-5255 (Print) 1744-7607 (Online) Journal homepage: http://www.tandfonline.com/loi/iemt20

Pharmacokinetics and Pharmacodynamics in


Antibiotic Dose Optimization

Sherwin K. B. Sy, Luning Zhuang & Hartmut Derendorf

To cite this article: Sherwin K. B. Sy, Luning Zhuang & Hartmut Derendorf (2015):
Pharmacokinetics and Pharmacodynamics in Antibiotic Dose Optimization, Expert Opinion on
Drug Metabolism & Toxicology, DOI: 10.1517/17425255.2016.1123250

To link to this article: http://dx.doi.org/10.1517/17425255.2016.1123250

Accepted author version posted online: 10


Dec 2015.

Submit your article to this journal

Article views: 4

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iemt20

Download by: [University of Nebraska, Lincoln] Date: 13 December 2015, At: 21:43
Publisher: Taylor & Francis

Journal: Expert Opinion on Drug Metabolism & Toxicology

DOI: 10.1517/17425255.2016.1123250
Pharmacokinetics and Pharmacodynamics in Antibiotic Dose

Optimization

Sherwin K. B. Sy1,2, Luning Zhuang1,3 and Hartmut Derendorf1‡


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

1
Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville,
Florida, USA
2
Post-Graduate Program in Biostatistics, Universidade Estadual de Maringá, Maringá,
PR, Brazil
3
Division of Pharmacometrics, Office of Clinical Pharmacology, Center for Drug
Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA


Author of correspondence:

Hartmut Derendorf, PhD


Chair, Distinguished Professor of Pharmaceutics, V. Ravi Chandran Professor in
Pharmaceutical Sciences
Department of Pharmaceutics
1345 Center Drive, Room P3-20
PO Box 100494
Gainesville, FL 32610, USA
Tel: +1.352.273.7856
Fax: +1.352.273.7854
E-mail: hartmut@ufl.edu

1
Abstract

Introduction: Identifying the optimized dosing regimen and algorithm is critical in the

development of antibiotics. Suboptimal regimens and inappropriate choice of drug give

rise to drug-resistant bacteria which have limited the therapeutic utility of many

commercially available antimicrobial agents. Strategies to optimize therapy of

antimicrobial candidates to speed up the development process are urgently needed.


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

Areas covered: We examined pharmacokinetics and pharmacodynamics of

antimicrobial agents with modeling and simulation approaches. The approach that is

based on minimum inhibitory concentration to evaluate antimicrobial dosing strategy is

widely utilized in drug development. The modeling approach utilizing information from

time-kill kinetic studies is a tool that can provide more information on the time-course of

bacterial response to a particular dosing regimen. Animal studies of dosing regimens

that mimic human pharmacokinetics are another option to evaluate antimicrobial

efficacy. Empirical, semi-mechanistic and mechanistic models of bacterial dynamics and

development of drug resistance in response to drug therapy are discussed.

Expert opinion: Both theories and applications of these approaches provide an overall

understanding of how the tools can streamline drug development process and help

make crucial decisions. Many opportunities and potentials are presented to incorporate

more rigorous integration of PK-PD modeling approaches even at preclinical stage to

extrapolate to clinical settings, thus enabling successful trials and optimizing dosing

strategies in relevant populations where the drug is mostly used.

Keywords: PK-PD modeling, antibiotics, in vitro models, translational research

2
Article Highlights

 We identified and summarized PK-PD approaches that are routinely practiced in

drug development of antimicrobial agents

 The MIC-based approach is the most extensively used approach to identify PD

index of antibiotic that is predictive of microbiological efficacy and outcome

 The state-of-the-art modeling and simulation strategies are now available and
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

can be more rigorously applied in the evaluation of drug candidates

 The population PK-PD modeling and simulation approach can elucidate more

information related to the time-course of antibacterial activities as well as

development of drug resistance in bacteria

 PK-PD has been successfully applied in optimizing antimicrobial dosing regimens

in special population such as patients with renal disease and critically ill patients

with obesity

3
1. Introduction

The utility of pharmacokinetic-pharmacodynamic (PK-PD) approach in

antimicrobial drug development is very well established and now acknowledged by

regulatory authorities. Thus, we will discuss how PK-PD information can be used for

rational drug design, dose finding and optimization of dosing regimen from the context

of antibiotics. There are several articles that discussed optimizing treatment with
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

existing commercially available antimicrobial agents to combat global rise in drug-

resistant bacteria.1-3 In fact, the same PK-PD concepts can also be used for candidate

selection and dose finding. A survey from 2002 showed that for drugs in therapeutic

areas other antimicrobial drugs, the “defined daily dose” were reduced whereas the

dosage regimens for antimicrobial drugs were increased after the drugs were approved

during the period between 1988 and 2000.4 For antibiotics, the dosage increase was

due to change in susceptibility towards higher drug resistance in the target

microorganism.4 Therefore, it is important that drug developers determine the optimal

dosage regimen in the first place before the drug receives regulatory approval and

labeling. The dosage regimen refers to a dose with a dosage interval and duration of

treatment. The PK-PD approaches are useful in determination of the first two; the

duration of treatment is more of a clinical issue. In this chapter, tools to help drug

developers get the correct dose and dosing interval will be discussed in detail. These

concepts are broadly applicable to drug development in other therapeutic areas.

4
2. Concepts in Pharmacokinetics and Pharmacodynamics

Once an antimicrobial agent is administered to a patient, there are processes

that govern the drug presence as well as its removal in the systemic circulation and in

the tissues. These processes are collectively known as ADME (Absorption, Distribution,

Metabolism and Excretion). Pharmacokinetics, also referred to as “what the body does

to the drug”, studies these kinetic behavior or processes and the concentration-time
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

profile of the drug, by determining PK parameters such as total body clearance, volume

of distribution, bioavailability and protein binding. These parameters allow one to

characterize and predict the time-course of drug kinetics under either physiological and

pathological conditions.5

When the drug reaches its site of action with the appropriate concentration and

stays at this site for a sufficient duration of time, its interaction with the target ultimately

results in some pharmacological action and the study of which is commonly referred to

as pharmacodynamics, also known as “what the drug does to the body”. In the case of

antimicrobial therapy, the site of action for the drug is the bacterial pathogen. The PK-

PD approach is a concept that links the PK and PD of the drug, describing the time-

course of the pharmacological effect, which in turn is dependent on the time-course of

the PK behavior and the dosing regimen.

In antimicrobial therapy, the time-course of the pharmacological effect, which is

characterized by the change in bacterial density over time in the host, is very difficult to

obtain in the clinical setting. Instead, a “snapshot” view using minimum inhibitory

concentration (MIC) can provide valuable information on the susceptibility of the

5
pathogen toward a specific or combination of antimicrobial agent(s).6 The MIC, which is

the lowest antimicrobial concentration that inhibits the growth of the microorganism

usually over a 16-20 h incubation window, is simple and relatively easy to determine.7

However, this simplistic approach also has its shortcomings and does not represent the

complex interactions between the host, the pathogen and the drug itself. There are

complex interactions between these three players namely, host, pathogen and the drug
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

that have to be considered in the drug development process: role of the host immune

response against the infection; development of multi-drug resistant bacteria in response

to suboptimal drug dosing regimens; adverse effects of the drug to humans; and the

drug’s PK and PD properties.

2.1 MIC-Based Approach

The PD surrogate indices in antimicrobial treatment was first identified in the 40s

and 50s by Eagle, who noticed the time-dependent properties of penicillin bactericidal

activity in rodents and the concentration-dependent killing of streptomycin and

bacitracin and the mixed pattern in tetracyclines.8, 9 Knowing the implication of these

characteristic PD properties for clinical therapies, Eagle realized that penicillins are best

administered as continuous infusion while concentration-dependent agents are better

given as intravenous bolus to achieve high maximum concentrations.10, 11 In the 90s,

Craig expanded on the concept of PD indices through a number of rodent studies

correlating PK parameters with bacterial kill over 24 hours.12 This PK-PD information is

very useful in the selection of the dose and dosing regimen for antimicrobial candidates

entering the clinical stage of drug development, as well as for clinicians in deciding

6
whether a specific antibiotic is suitable for the patient’s infection and how the antibiotic

should be administered.

2.1.1 Pharmacodynamic Indices

The quantitative relationship between PK parameter and microbiological

outcome, the change in log10 colony forming unit (cfu) in post-dose 24-h sampling from

the starting bacterial density often obtained from dose fractionation in the rodent or the
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

time-kill kinetic studies, is referred to as a PD surrogate index. The characterization of

PD surrogate index is primarily based on the relationship between drug exposure and

MIC. The three common PD indices used to predict antimicrobial efficacy are time at

which the drug concentration is above MIC over the 24-h interval (T>MIC), the ratio of

peak drug concentration and MIC (Cmax/MIC), and the 24-h area under the

concentration-time curve over MIC (AUC/MIC) at steady state conditions. These indices

are often prefixed by an italic f, representing the free and unbound drug concentration,

as it is often assumed that only the unbound drug can exert its pharmacological effect.

In the literature, one often finds the percentage of time above MIC over the 24-h period

rather than the absolute time above MIC being reported.13 These patterns of

antimicrobial killing effect are summarily characterized as either time-dependent or

concentration-dependent killing effects. Some investigators included a third

classification as concentration-independent killing with prolonged post-antibiotic

effects.1 The post-antibiotic effect is the duration from the time after the microorganism

is exposed to the drug until the survivors start to multiply rapidly.14

7
Time-dependent killing with short or no post-antibiotic effects. Drugs exhibiting

this killing pattern is best described by the PD index fT>MIC. The dosing strategy is to

maintain the free drug concentration above the MIC value for an extended period of

time. The class of β-lactam antibiotics is commonly associated with the term

“concentration-independent” or “time-dependent” kill. That is because the efficacy of β-

lactams is enhanced with longer exposure times of the free drug concentration

maintained above MIC. β-lactams with the exception of carbapenems have limited
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

postantibiotic effect against gram-negative bacteria.14, 15 A classic illustration of β-lactam

exhibiting time-dependent killing effect is shown in Figure 1. In this example of

ceftazidime against Klebsiella pneumoniae ATCC 43816 in the lungs of neutropenic

mice, the time above MIC is best correlated with the 24-h cfu.16 The vertical line in

Figure 1 indicates that ceftazidime concentration above the MIC in at least 60% of the

dosing interval achieves the greatest bacterial kill.

Concentration-dependent killing with extended post-antibiotic effects. Drugs of

this killing trend tend to be best characterized by the fCmax/MIC ratio. The killing pattern

of aminoglycosides is best correlated with this ratio. Whether the concentrations of

these antimicrobial agents are maintained above the MIC for an extended period or not

seem to have only limited impact on their efficacy. This is due to their post-antibiotic

effect. Even when the active free drug concentration falls below the MIC, the extended

effects provide protection against the regrowth of bacteria. The magnitude of the peak

concentration is often associated with its bacterial killing efficiency. The initial kill rate of

aminoglycosides in the time-kill kinetic studies is proportional to the drug concentration

measured by the number of fold of MIC, as shown in Figure 2 for tobramycin.17 In

8
contrast, the initial kill rates do not change significantly with increasing concentration of

ticarcillin, a β-lactam which exhibits a time-dependent killing pattern. The shapes of the

initial kill from the static time-kill curves are not necessarily indicative that the drug is a

concentration-dependent agent. The presence and the duration of the post-antibiotic

effect are just as important.14

Concentration-independent killing with extended post-antibiotic effects. Drug


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

showing this type of killing pattern is often characterized by fAUC/MIC. The quinolones,

tetracyclines, oxazolidinones, vancomycin, collistin, as well as the macrolide antibiotics,

exhibit this type of killing pattern. The prolonged effect inhibits bacteria regrowth even

when the drug concentration is below MIC but their drug effect is not dependent on the

peak drug concentration. The example of linezolid in Figure 3 shows that the 24-h

AUC/MIC has the tightest correlation with the change in 24-h log10 cfu with a coefficient

of determination, R2 value, of 0.82.18 The criteria for deciding as to which of the PD

indices best characterizes the drug killing effect is based on fitting a sigmoidal Emax

model on the microbiological outcome such as the bacterial 24-h log cfu against the

three PD indices. The parameter that had the tightest correlation with the

microbiological outcome is selected as the PD index for the drug.

As a well-established approach to predict the success or failure of therapy, the

three empirical pharmacodynamic indices show co-linearity in some scenarios (R2

values are not significantly distinctive), resulting in difficulty in decision making for dose

selection. The predictive capacity of empirical pharmacodynamic indices has been

evaluated by Nielsen using semi-mechanism PK/PD model.19 The results suggest that

the efficacy of moxifloxacin can be described by both fCmax/MIC (R2=0.93) and

9
fAUC/MIC (R2=0.99) and the efficacy of erythromycin can be characterized by both

fAUC/MIC (R2=0.93) and fT>MIC (R2=0.90). The dose fractionation study conducted by

Andes demonstrated that the correlations of AUC/MIC, T>MIC with bacterial count were

82% and 57%, respectively, for linezolid against Streptococcus pneumoniae and 75%

and 74%, respectively, for linezolid against Staphylococcus aureus.18 Although the

author claimed that the narrow insufficient difference between effective and ineffective
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

dosing regimens may lead to indistinguishable pharmacodynamic parameters against S.

aureus, the result still left us questioning whether the empirical pharmacodynamic

parameters derived from animal infection model may be bacteria dependent and

whether the information provides complementary information of drug efficacy from

different perspectives. A new pharmacodynamic index, weighted AUC/MIC, was

proposed by Corvaisier et al. in order to take into account both the concentration-

dependent part and the concentration-independent part of the antibiotic efficacy.20 As

the selection of the appropriate PD index as a surrogate marker of efficacy is important,

the magnitude of these indices that maximizes efficacy is equally important if not more

important. It was previously suggested that the PD index determined in rodent studies

can be extrapolated to clinical efficacy.14 Many of the current dosing regimens in the

clinic were based on PD indices that were determined from mice. Table 1 lists the target

PD indices for several antimicrobial agents. As in the example from Figure 1, the fT>MIC

of at least 60% of the dosing interval for ceftazidime is associated with the lowest log10

cfu at 24 hour post-dose. Vancomycin dosing regimen was optimized to achieve the

target AUC/MIC ratio of 400 h in treating ventilator-associated S. aureus pneumonia.21,


22
The vancomycin nomogram of dosing regimen based on body weight and renal

10
function was designed to achieve a target trough concentration of 15-20 mg/L to avoid

vancomycin-related nephrotoxicity and at the same time targeting an AUC/MIC ratio of

400 h.23, 24 Interestingly, AUC/MIC is a PD surrogate index actually possessing a time

dimension (h). Thus, the ratio of 400 is computed over a 24 h period at steady state.

Toutain et al. suggested a more universal metric by dividing the AUC/MIC by the time

interval of interest to permit the assessment of a truly dimensionless ratio and offer
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

direct clinical interpretation, based on which the target concentration of vancomycin at

steady state would be 16.6 fold MIC.25 There are some indications that these PD indices

are not affected by bacterial strain. In the study of gatifloxacin against Salmonella

enterica serotype Typhi, where two isolates consisting of both susceptible and resistant

strains, the PD index was the same against both strains, regardless of the MIC value.26

Studies of colistin PD index, on the other hand, seems to indicate that the magnitude of

the index value is bacteria- and strain-dependent. Dudhani et al. reported fAUC/MIC

value between 23 and 46 for colistin against Pseudomonas aeruginosa 27, 28whereas

Guyonnet et al. reported a much lower index value against Escherichia coli for its

bactericidal effect characterized by a reduction of at least 3 log unit in cfu.29 Bergen et

al. reported an index value between 6.81 and 35.7 for several strains of P. aeruginosa.30

2.2 Pharmacodynamic Indices in Antimicrobial Combinations

2.2.1 β-lactam – β-lactamase combination

As multidrug-resistant infections have contributed to a rise in mortality in the

hospital setting, many antimicrobial agents are combined with another agent that

counterattacks the bacteria’s drug resistance mechanism. β-lactamases, which are

11
responsible for the bacteria’s resistance to β-lactams, threatened the utility of the class

of β-lactams. Consequently, β-lactamase inhibitors were developed and introduced to

clinical practice. These inhibitors when combined with a partnering β-lactam would

confer susceptibility of the bacteria to β-lactams and restore the antibacterial activity of

a partnering β-lactam. The clinically available combinations include amoxicillin-

clavulanate, ticarcillin-clavulanate, ampicillin-sulbactam, piperacillin-tazobactam,

ceftolozane-tazobactam and ceftazidime-avibactam, listed in the format, β-lactam – β-


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

lactamase inhibitor. With the exception of ceftazidime-avibactam combination which

recently received marketing approval from the US Food and Drug Administration, drug

resistance to β-lactam – β-lactamase inhibitor combinations has been documented in

the literature.31-34 The drug resistance was primarily due to β-lactamases that were not

covered by these inhibitors. For example, TEM-30 and SHV-10 β-lactamases are

resistant to clavulanic acid, sulbactam and tazobactam.35 Table 2 shows the molecular

classification of β-lactamases, the corresponding β-lactam substrates and the β-

lactamase inhibitors that inhibit the specific class of enzymes. The molecular

classification is based on the conserved and distinct amino acid motifs. The β-

lactamases in classes A, C and D hydrolyzes their substrate by forming an acyl enzyme

through their serine active site whereas those in class B are metalloenzymes that use

the zinc active site for hydrolysis of β-lactams. There are no commercially available β-

lactamase inhibitors for the class B metalloenzymes, whereas the available inhibitors

have only limited coverage of the class D serine-based enzymes. The competitive

landscape indicates a commercial opportunity for broader spectrum inhibitors or

antibiotics that can evade β-lactam resistance through a different mechanism of action.

12
For the past several decades, β-lactamase inhibitors have been used clinically

but very limited information were found in the literature on optimizing dosing regimens of

these inhibitors using PD indices. The PD index for β-lactam–β-lactamase inhibitor

combination is based on the established T>MIC for the β-lactam and T>threshold concentration

(or T>TC) for the β-lactamase inhibitor. The threshold concentration is the lowest

concentration of the β-lactamase inhibitor at which a significant difference between MIC

of the partnering β-lactam as monotherapy and the MIC of the combination can be
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

observed in the resistant strain bacteria. The threshold concentration for tazobactam

varied in several articles. An early study reported a threshold concentration of 4 mg/L

tazobactam in an in vitro model of infection that was carried out in piperacillin-

susceptible E. coli J53 and P. aeruginosa ATCC 27853 and piperacillin-resistant

Staphylococcus aureus 7176 and isogenic TEM-3 construct in E. coli J53.36 Their

approach is based on the proposal that the activities of various β-lactam – β-lactamase

combinations be determined by assessment of the concentration of inhibitor needed to

bring the susceptibilities of individual organisms to below accepted susceptibility

breakpoints for the β-lactam alone or even to the susceptibilities for non-β-lactamase

wildtype strains.37 The resulting T>4 mg/L tazobactam was 50 and 38% for 3g/0.375g q6h and

4g/0.5g q8h piperacillin/tazobactam, respectively; both dosing regimens of the

combination were highly active against all strains resulting in at least 4 log kill.36 In two

later studies, the process of determination of threshold proposed for the ceftolozane-

tazobactam combination utilized a different algorithm wherein the threshold candidate

concentrations of 0.01 to 1 and 0.25 to 4 mg/L were evaluated in time-kill kinetic

studies.38, 39 In one study, their analysis returned a much lower concentration values of

13
0.05 and 0.25 mg/L tazobactam concentration for the low to moderate and high β-

lactamase expression E. coli strain consisting of blaCTX-M-15 constructs, respectively.39 In

another study, the same authors determined that the threshold concentration for

tazobactam ranged from 0.5 to 4 mg/L in four E. coli and three Klebsiela pneumoniae

clinical isolates.38

The relationships between three tazobactam exposure measures, AUC, Cmax and
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

T>TC, and the change in log10 cfu after 24 h therapy in a PK-PD in vitro infection model

were examined.39 The dose fractionation schedules were q6h, q8h, q12h and q24h. The

relationship between T>TC and microbiological outcome resulted in an R2 value of 0.938.

The authors concluded that the magnitudes of T >TC for tazobactam associated with net

stasis (no change in log10 cfu), and a 1- and 2-log10 cfu reduction based on the model-

predicted data at 24 h were 35, 50 and 70%, respectively, regardless of the level of

transcription efficiency of the β-lactamase enzyme.39

The optimal PD index for avibactam determined from dose fractionation

experiments was also T>TC.40 The T>TC for avibactam was approximately 60% to 80%,

corresponding to 14.8 h and at most 19.2 h in which the partnering ceftaroline at 600

mg q8h was used and the threshold avibactam concentration of ≥4 mg/L was

assumed.40 In the ceftazidime-avibactam combination, a threshold avibactam

concentration of 0.5 mg/L was required to suppress regrowth of Enterobacteriaceae

expressing AmpC β-lactamase.41

In the dose-finding trials, the threshold avibactam concentration was 1 mg/L.42

The information presented by the developer of ceftazidime-avibactam combination at

14
the EMA dose finding symposium indicated that the same duration (as percentage of

the dosing interval) for avibactam as that for ceftazidime to be maintained above the

MIC was required to maintain continued suppression of β-lactamase activity, which in

this case was 50% fT>ceftazidime-avibactam MIC and 50% fT>avibactam TC.

2.2.2 β-lactam – aminoglycoside/fluoroquinolone combination.

Combination therapy, comprised of a β-lactam and an aminoglycoside or a


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

fluoroquinolone, is highly recommended to treat patients with serious infections caused

by P. aeruginosa according to a guideline from the Infectious Diseases Society of

America (IDSA).43 Combination chemotherapy has the potential to provide the highest

probability of attaining optimal organism kill by suppressing resistance development and

the lowest probability of side effect by decreasing the levels of both components. The

study published by Louie evaluated combination chemotherapy of meropenem and

tobramycin for P. aeruginosa in a murine pneumonia model.44 The results show the

near-maximal effect was reached at 60 mg/kg/day and 50 mg/kg/day of meropenem

and tobramycin, which produced a T>MIC of 35.25% and an AUC/MIC ratio of 80.1 h in

epithelial lining fluid (ELF). The similar effect requires an AUC/MIC ratio in the epithelial

lining fluid of 240.3 h when tobramycin was administered as a single agent, implying the

advantage of combination therapy over monotherapy. However, Joly-Guillou compared

the in vivo activity of levofloxacin alone and in combination with imipenem in a mouse

model of Acinetobacter baumannii pneumoniae and observed no efficacy enhancement

with combination therapy.45 The benefit of combination therapy of a β-lactam and an

aminoglycoside was also questioned by several papers based on meta-analyses in

some special populations, demonstrating that all-cause mortality rates are comparable,

15
whereas combination treatment carries a significant risk of nephrotoxicity compared to

monotherapy of β-lactam.46, 47 Clinically, combination therapy has been widely practiced

in treatment of infections caused by multidrug-resistant organism (MDRO) with the

expectation of introducing synergism to reduce the selection of mutant bacterial

resistance to each antimicrobial component. The PD indices could serve as a good

efficacy surrogate to assess the drug-drug interactive effect for antimicrobial


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

combination and help dose selection in the future clinical study design.

2.3 Issues with MIC-based approach

The MIC is a measure of the potency of an antimicrobial drug. However, MIC is

usually determined in vitro by aerobic incubation in a protein-free liquid medium at pH

7.2. The conditions are highly distinct from those at the site of infection, where the

environment is anaerobic, acidic and surrounded with protein.48 Thus, the MIC values

derived from artificial fluids do not necessarily indicate accurate drug efficacy in vivo in

biological fluids. A MIC comparison of oxytetracycline was conducted by Brentnall in

Cation Adjusted Mueller Hinton Broth (CA-MHB) and calf serum.49 The results showed

that the MIC value was 19-fold higher in serum than in broth, indicating a significant loss

of potency of oxytetracycline in calf serum. Protein binding may have contributed to the

higher MIC obtained in serum as protein binding of 18.6% to 72% has been reported for

cattles,50, 51 which would only account for partial of the ratio. The difference of calcium

and magnesium concentrations in two matrices may also affect the MIC value. A further

assessment is required to provide grounded explanations. On the other hand, Buyck

found that the susceptibility of P. aeruginosa to macrolides and ketolides in eukaryotic

cell culture media and biological fluid would be dramatically enhanced compared to CA-

16
MHB due to decreased expression of oprM and increased outer-membrane

permeability. The work opens new perspectives that macrolides may exert in vivo

antibacterial activity against P. aeruginosa that would not be predicted on the basis of

conventional susceptibility testing system.52 Pruul also examined the interaction of

azithromycin and other macrolides with normal human serum in relation to MIC.53 The

result elucidated that the MIC of azithromycin was decreased by twenty-six-fold for
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

serum resistant E. coli and fifteen-fold for S. aureus in presence of 40% serum with low

protein binding characteristics (8.5% at a concentration of 0.01 nM in 95% serum).

Erythromycin had a similar susceptibility pattern, while roxithromycin was less active

against S. aureus in the presence of serum.53 In general, free unbound drug

concentrations at the site of action are responsible for the pharmacodynamic effect of

anti-infective agents. Although many other factors may have certain impact on efficacy

of antimicrobial drugs, MIC-based approach is still a well-established method offering a

profound understanding of why a particular dosing regimen achieves clinical success.

In addition, one of the widely-accepted antimicrobial susceptibility testing

methods is macrobroth or tube dilution method. This procedure involved preparing two-

fold dilutions of antibiotics in a liquid growth medium dispensed in test tubes.54 The MIC

determined by 2-fold dilution is considered as a relatively crude index since the “true”

MIC is a point between the lowest test concentration that inhibits the growth of the

bacteria and the next lower test concentration. To improve accuracy of MIC, Aliabadi

and his colleagues used five overlapping sets of doubling dilution and their result

illustrated that the MICs of danofloxacin were 0.03 μg/mL in broth, sheep serum and

exudate and 0.035 μg/mL in transudate.55 Disk diffusion and gradient diffusion are also

17
reliable manual methods that provide reproducible result and convenience of

preparation. Currently, four automated instrument instruments have been approved by

the FDA for use in the United State to produce susceptibility test results in a shorter

period than manual reading; these instruments have significantly reduced the manual

labor involved in running susceptibility testings.7

2.4 In vitro Time-Course-Based Approach


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

The in vitro time-course of bacterial response to both static and dynamic drug

concentration characterized by time-kill assays has been used in pharmaceutical drug

development of antimicrobial agents. The time-course approach provides greater detail

of how the colony population of bacteria reacts to an antimicrobial challenge over time.

Depending on the objective of the study, the drug concentration in the in vitro time-kill

experiment can be constant in the static setting or dynamically changing to mimic the in

vivo half-life of the drug in humans. The constant or static exposure is achieved by not

replacing the medium, while the dynamic assay is accomplished by changing the

medium with fresh ones that do not contain the antibiotic. A loss of bacteria due to the

experimental setting, which is observed in some models, may have a substantial

influence on the results. By using semi-permeable membranes, loss of bacteria can be

avoided during replacement of medium that mimics the loss of drug or decreasing drug

concentrations.56

The information from static time-kill experiments are often used to develop a

mathematical model that links the free drug concentrations to the bacterial response

whereas the dynamic time-kill data can be used to validate the model or to evaluate the

18
outcome in the clinic using humanized dosing regimens.57 The dynamic kill-curve also

provides an alternative for evaluating PK-PD relationships, as it simulates the time

course of the free drug concentrations in the plasma based on the characteristic half-life

of the drug.2 The effects of different dosing regimens, drug half-lives and even starting

inoculum sizes can be simulated to study their effects on the bacterial population

dynamics over a specific time period, such as 24, 48 or 72 h.58-60


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

The common type of models that are often developed for the purpose of

establishing PK-PD relationship are the semi-mechanistic models. As opposed to

mechanistic models, the semi-mechanistic models are empirical models that capture the

broad general characteristics of bacterial growth and drug effects, as well as

development of bacterial resistance. These models are often developed from in vitro

information that comes from both static and dynamic time-kill experimental data.

The most common type of semi-mechanistic models in the literature to describe

the PK-PD relationship of antimicrobial agents and bacterial population dynamics is the

logistic growth model, which was introduced by Pierre-François Verhulst in 1838 to

study human population:61

1
( )

where N is the population number in unit of cfu/mL, kgrowth is the growth rate constant

with units often in h-1, and Nmax is the maximum population size that is supported by the

environment, also known as the carrying capacity and in unit of cfu/mL. The important

characteristic of this differential equation is that there is a limit as time approaches

19
( )
infinity for its analytical solution, which is ( ) , where N0 is the
[ ( ) ]

initial population size. This limit is Nmax (i.e. ( ) ). In the in vitro bacterial

system, the bacterial cfu is restricted from growing indefinitely and reaches a plateau,

where there is no net growth. For this reason, the logistic growth model is suitable for

describing this behavior.

The effect of antimicrobial intervention on the capacity limited growth model in


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

equation 1 is incorporated to the model with a function that describes the drug effect:

2
( ) ( )

where the ( ) is a function of drug concentration and is often

represented by an Emax or sigmoidal Emax model,

3
( )

wherein Emax is also termed as kkill,max with unit of h-1 and EC50 has the same unit as the

drug concentration.

We note that the MIC, EC50 and the stationary concentration are not parameters

but rather variables as their values are dependent on the inoculum size. 62 The

stationary concentration or the concentration at which the growth rate equals the kill rate

where no net change in the number of bacteria is occurring can be derived by assuming

a linear growth at the initial growth phase where and equation 2 becomes

( ) .63 The analytical solution for this differential equation is

20
( ) [( ) ]. The stationary concentration occurs when

( ) ( )
( ) or . By taking the natural log of , this value is 0. Thus,

. Solving for C, one obtain the following equation:

4
( )
[ ]
( )
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

( )

( )
When there is no net change in the number of bacteria, the term approaches 0

and the static concentration (SC) is:

5
[ ]

It is not to be confused between SC and MIC. Because at SC, we assumed that

( ) , the initial inoculum size is approximately 5  105 cfu/mL, which is not visible

to the human eye. The visible bacterial density is approximately 10 7 to 108 cfu/mL. Thus

a correction factor to the SC equation is required to obtain the MIC:63

6
[ ]
( )

The value of 0.29 came from the assumption that ( ) reached 108 cfu/mL at 18 h with

an initial inoculum of 5  105 cfu/mL. This correction factor is dependent on the specific

system that the drug is being tested.

21
Further modifications to the logistic growth factor included incorporating an

adaptation factor to the EC50 parameter and “delay functions” to both the growth and

drug action part of the model. Both of these modifications allow the model to adapt to

the bacterial regrowth that is often observed in time-kill kinetic studies. The adaptation

factor to the EC50 parameter has the following form:64

7
( )
( )
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

wherein α is defined as:

( ) 8

where is the exponent of the adaptation factor and is the maximum value for the

adaptation, since the range of values for the function is between 0 and 1. A

baseline EC50 value at time 0 would either increase or decrease over time to a maximal

value of , depending if is positive or negative, respectively.

The delay function has the form, , which behaves like a cumulative

density function starting from 0 at time 0 to a maximum value of 1 as time approaches

infinity. The equation 2 incorporating two delay functions is shown in equation 9:

9
[ ( )( )

( )( )]

22
This model was applied to model bacterial dynamics of S. pneumoniae, Haemophilus

influenza and Moraxella catarrhalis in response to azithromycin.65

The logistic growth model was recently adapted for combination therapy utilizing

Loewe additivity to evaluate whether a combination of an aminoglycoside and a β-

lactam would result in synergistic activities,66 since both agents have antimicrobial

activities with different mechanism of actions. Greco proposed a generalized sigmoidal


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

Emax equation utilizing Loewe additivity model:67

10
⁄ ⁄
( ) ( )

( ⁄ ⁄ )
( )

The first two terms describe the additive effect of the two agents while the third term is

an interaction term, wherein the parameter indicates synergism-antagonism

interaction. The interaction is considered additive if the 95% confidence interval of the

estimate overlaps the zero value. If or , the interaction is either synergistic or

antagonistic, respectively. An additional interaction evaluating the initial killing rate (k max)

was included:

11

The final Emax model with the adaptation factor to evaluate the combination of these two

agents was:

23
12
( )

( )

where refers to the adaptation factor mentioned in equation 8. This empirical

approach described well the combined effects of an aminoglycoside and a β-lactam

against P. aeruginosa.66
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

In the examples above, these models were fitted to the data. It is important to

consider whether the parameters estimated are identifiable and do not have co-linearity

with other parameters. In cases of increasing model complexity where parameters

outnumber the available data points to estimate the parameters precisely, non-

identifiability of parameters can arise. Strategies to detect non-identifiability in

parameters include slight perturbation to a parameter and evaluation of which

parameters are affected. Co-linearity can be detected by correlation or by plotting the

perturbed parameter values against other parameters estimated by the model. 68

Algorithms to detect non-linear manifolds have been developed69 but are not very

sensitive. When co-linearity arises, one can consider fixing specific parameters to

literature values.

To overcome parameter identifiability problems with increasing model complexity,

for example in mechanistic models, simulation approach rather than fitting model to data

is often undertaken. The simulation approach requires more efforts in collecting

literature information on the parameter values. The complex modeling of networks

24
involved in resistance development is one such example of highly complex models. 70, 71

In later section, models for resistance development in bacteria are discussed.

2.5 Measurement of free unbound drug concentrations for accurate

characterization of antimicrobial efficacy

The free unbound drug concentration in the interstitial fluid at the target site has

been shown to be better correlated with the drug’s antimicrobial efficacy. In contrast,
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

total plasma drug concentration is often a poor descriptor of the drug’s activities at the

site of action, particularly for antibiotics, as most infections do not occur in the plasma

but rather in the tissue sites. Given that tissues are not homogeneous compartments,

the use of protein binding as proportion of total drug concentration does not extend to

the free drug concentrations in the tissue. To measure free drug concentration in the

interstitial spaces, in vivo microdialysis is used to characterize drug distribution in the

tissues.72 This technique involves implanting a dialysis probe into the tissue or organ of

interest and the probe is constantly flushed with a compatible fluid for example saline

solution at a controlled flow rate. The protein-free drug in the interstitial fluid enters the

probe’s semipermeable membrane by passive diffusion. The equilibrium between

concentration gradient inside and outside the probe is established relatively rapidly. The

free drug concentration in the tissue is then computed based on the relative recovery

determined depending on the particular method such as retrodialysis and no net flux.

The important assumption for the method is that the recovery is constant over time.

Problems could arise implementing this procedure as the recovery can be influenced by

factors such as the membrane area of the probe, molecular weight cutoff for substance

entering the semipermeable membrane, flow rate of the fluid used to flush the probe,

25
perfusate and temperature.73 Thus, a pilot study evaluating the feasibility and

compatibility of the drug with the microdialysis procedure is often performed in vitro prior

to implementing microdialysis technique in human test subjects.

Many applications of microdialysis have been in antimicrobial therapies. The

prophylactic use of 1.5 g short-term infusion of cefuroxime in morbidly obese patients

during abdominal surgery was investigated by evaluating whether subcutaneous and


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

muscle drug concentration provides sufficient coverage against methicillin-suspectible

S. aureus and E. coli.74 Their study showed that this dosing regimen achieved free drug

concentrations in the respective tissues for over 40% of the dosing interval; cefuroxime

is a β-lactam whose pharmacodynamics index is characterized by fT>MIC. The

pharmacokinetics of ertapenem was monitored in the interstitium of healthy uninfected

tissue and infected tissue from patients suffering from diabetic foot infections to assess

the sufficiency of antibiotic concentrations at the target site. The results suggested that

the free interstitial ertapenem concentrations were higher than in inflamed tissue of

diabetic feet than in healthy adipose tissue, indicating good tissue penetration in

diabetic foot infections. However, 1 g of ertapenem daily given intravenously was

unable to reach fT>MIC higher than 40% in more than half of the patients in the

interstitium of infected tissue. Higher daily dose might be considered in patients with

diabetic foot infection to optimize bactericidal effect.75 The levofloxacin penetration into

the lung tissue after cardiac surgery was measured using microdialysis technique in six

patients receiving a dose of 500 mg via intravenous administration in addition to the

standard antibiotic prophylaxis regimen. The median fAUC/MIC in lung tissue was 2.4

(ranging from 1.3 to 4.2) for P. aeruginosa, demonstrating that the dose was borderline

26
sufficient for the treatment of nosocomial pneumonia caused by K. pneumoniae and

insufficient for the treatment of pneumonia caused by P. aeruginosa. An fAUCtissue/MIC

ratio of 30 to 40 is associated with high rates of bacterial killing and thus maximizes the

efficacies of fluoroquinolones.76

3. Modeling Approach

The model-based development approach is particularly useful for development of


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

antimicrobial agents, because the classification of antimicrobial classes based on the

PD indices are well established and the translational value of in vitro time-kill kinetics to

the clinical practice has been proven to work. This section will discuss modeling

strategies that can be incorporated into the development of antimicrobial agents.

3.1 PK-PD models based on static MIC and time-kill curves

In the MIC-based approach, the PK exposure parameters come from the serum

antimicrobial concentration and the PD parameter is the MIC. The goal is to find a dose

where the index is attained or exceeded. For example, if the antimicrobial agent is

concentration-dependent, one would first establish the ratio of fCmax/MIC that achieves

the optimized kill from either animal studies or in vitro dynamic time-kill studies. Once

the PD index is established, the human PK information would then come from phase I

ascending dose studies to determine which dose would result in the required free drug

Cmax (i.e. protein-binding-corrected Cmax) to achieve or exceed the target fCmax/MIC

ratio. The human PK is modeled by a population PK model so that simulations of

different dosing regimens can be done for the purpose designing future trials. The

population PK model consists of both deterministic and stochastic components.5 The

27
deterministic model takes the form of the classical compartmental model. The stochastic

component describes the variability between individuals as well as determines the

sources of inter-subject variability through covariate models. The second level of

variability is within-subject variability which can be described by the residual error model

and inter-occasion variability.5

The covariate model quantifies the source of inter-subject variability contributing


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

to a PK parameter. For example, the covariate model may include body weight as a

covariate of the volume of distribution and drug clearance, sometimes utilizing allometric

scaling. Many antimicrobial agents such as vancomycin, amikacin are dosed by body

weight.21, 77 For many antibiotics, the renal function has an important impact on the

clearance of the drug. Creatinine clearance, in this case, would be an important

covariate of the population PK model. These covariate models are important and should

be considered during model development.

3.2 Monte Carlo PK-PD simulations

Once the population PK model is established for humans and the PD indices

determined from animal or in vitro studies, several dosing regimens can be simulated

with virtual individuals to determine which dose and dosing interval are most optimized

to achieve the target PD index. In the example of drug-drug interaction study of

efavirenz and para-aminosalicylic acid, efavirenz was shown to induce the clearance of

para-aminosalicylic acid, which is used as companion drug to prevent further

development of drug resistance in tuberculosis infection.78, 79 This drug-drug interaction

was evaluated because many tuberculosis patients in the African continent also have

28
HIV co-infection and often are on highly active antiretroviral regimen containing

efavirenz in addition to their multiple-drug therapy for tuberculosis. In that study,

several dosing regimens of para-aminosalicylic acid were simulated to determine which

doses and dosing intervals would not achieve the target PD index of maintaining the

free trough para-aminosalicylic acid concentration above the MIC of 1 mg/L with and

without concomitant efavirenz. The simulations were stratified by whether concomitant


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

efavirenz medication was present or not. The population PK model-based exposure

parameters generated from the simulations were 24-h steady-state AUC, peak, trough

and the free trough concentrations. The results indicate that at least 4g twice-daily

dosing regimen ensures free drug concentrations exceeding MIC over the entire dosing

interval, even in HIV-infected patients who received efavirenz, whereas once-daily

dosing even as high as 12 g would risk the free trough concentration falling below the

MIC in some patients.78

Once both the population PK model and the PK-PD model for the in vitro time-kill

kinetics are developed, the time course of bacterial cfu can be simulated to study which

dosing strategy would give a favorable microbiological outcome. In the study by Zhuang

et al (2015),66 the prediction of the antibacterial activity of an aminoglycoside in

development for three dosing regimens, namely, 3 mg/kg QD, 5 mg/kg QD and 2.5

mg/kg BID were simulated, as shown in Figure 4. The simulations were based on the

PK model using the population parameter mean values. The population parameter

mean values without including the intersubject variability were used for the simulations

because the in vitro semi-mechanistic PD model has its inherent error. If one were to

include intersubject variability in the PK model, the range of values in the antibacterial

29
activities would be amplified and the pattern of bacterial kill could become unclear due

to the exaggerated variability in the response. The same approach using PK-PD model

simulation can be utilized in drug development for dose optimization in the clinic. The

PK-PD model is often developed immediately before going into the phase II trial, since

the PK information from the phase I trial is used for the population PK model

development. The modeling and simulation approach can give better confidence to the
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

dose selection for the phase II and III trials.

3.3 Probability of target attainment and cumulative fraction of

response

The probability of target attainment is the percentage of the simulated individual’s

profiles that are over the threshold or target PD index. Very often, approximately 10,000

individual drug concentration-time profiles are simulated using a population PK model

developed from PK studies. The large number of simulations is required to characterize

accurately the probability values. The distribution of PD index becomes the basis for

determining the likelihood of achieving a specific target attainment. In the previous

example for para-aminosalicylic acid,78 we estimated the probability of target attainment

to evaluate the dosing regimens that would achieve free trough concentrations above

MIC. The PTA is computed in a statistical program by ranking the simulated trough

concentrations and estimating the percentiles that these values are greater than a range

of MIC values.

The alternative prediction of clinical outcome using Monte Carlo simulation is the

cumulative fraction of response (CFR), which is defined as the expected population PTA

30
given a population of microorganisms for a specific dosing regimen.1, 13 The equation to

compute CFR is as follows:

13
( ) ∑

wherein CFR is the sum of the PTA at a specific MIC multiplied by the frequency (F) of

bacterial isolates with that MIC over the range of MIC values. In this case, a population
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

distribution of MIC values rather than a single MIC value is used and the expected

probability of success of a dosing regimen is estimated for the population distribution of

MICs instead of estimating for one MIC value. This approach is said to be more useful

for community medicine than for drug development, as the situation may be such that

the pathogen susceptibility may not have been determined at the time of patient

evaluation. A key point is the selection of the adequate range of MIC values, as the

pathogen susceptibility may vary between countries, areas and hospitals, as well as

over time.1 Thus, CFR is often specific for an institution or hospital.80

4. Translational Approach to Bridge In vitro Information to the Clinic

Examples are presented in this section to show how the in vitro information is

utilized to shed light on dosing strategies in humans and how modeling was applied to

optimize dosing regimens in special population.

4.1 Modeling approach to determine pharmacodynamic indices

In the example of ceftazidime, the time-course of bacterial response to the drug

from the in vitro time-kill curves was used to infer why the PD index fT>MIC of at least

31
50% is required for bactericidal effect in vivo.81 The logistic-growth model was fitted to

the time-kill data. Using the in vitro PK-PD model, the hypothetical in vivo bacterial

response over time after 1 mg every two hours to 256 mg every 8 hour was simulated to

evaluate the dosing regimen that would be able to provide sufficient antibacterial

activities. The time-course of drug concentration profile was simulated from the PK

model for both humans and mice. The dosing regimens that achieved a predicted static
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

effect such that the cfu at 24 h is less than the cfu at 0 h were then evaluated. The

corresponding fT>MIC for that dosing regimen was subsequently determined. The

authors estimated that fT>MIC of at least 50% is required in order to achieve a 2 log10 kill

after 24 hours. This is one example of how modeling and simulation can be used to

derive a target PD index.

Another group used a semi-mechanistic PK-PD model to predict the PD indices

of several antibiotics.19 Even though the simulations using the in vitro information can be

used to predict target PD indices, the author cautioned that the suitable PD index for a

specific antimicrobial is sensitive to study conditions including dosing frequency and

uncertainty in the MIC determination.

4.2 PK-PD simulation to evaluate dosing regimens in patients with

end stage renal disease

Patients with end-stage renal disease have deteriorating chronic kidney disease

wherein their renal function are <10% of the normal capacity.82 Repeated hemodialysis

is required to prevent the accumulation of fluid, electrolytes and toxins. Infection is the

32
second leading cause of mortality in end-stage renal disease patients.83 Thus

antimicrobial agents are highly used in this patient population.

Dosing of antimicrobial agents in end-stage renal disease patients is still quite

controversial as these patients tend to have altered drug clearance and the dialysis

procedure tends to remove majority of the drug in the system.83 We note that drugs with

a rather small steady state volume of distribution are readily removed by dialysis. The
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

current guideline for dosing gentamicin is to administer half of the full dose immediately

after hemodialysis; this recommendation is being challenged by several studies that

argue for administration of full dose an hour before hemodialysis.84-87 The predialysis

dosing argument is based on PD index of fCmax/MIC that requires Cmax to be 8 to 10-fold

higher than MIC,86 without considering the area under the curve which is also

associated with the efficacy of gentamicin.

Zhuang and colleagues utilized a semi-mechanistic model developed from in

vitro time-kill curves and the PK model of gentamicin in end-stage renal disease

patients undergoing hemodialysis to compare the antimicrobial activities of

administering 120 mg gentamicin immediately after hemodialysis compared with

activities associated with 240 mg given 1 hour before hemodialysis.88 The results of the

simulation exercise show that the half dose of gentamicin at the end of hemodialysis

would result in similar microbiological outcome as that when a full dose of gentamicin is

administered 1 hour before hemodialysis. The end-stage renal disease patients present

a case wherein PK-PD indices may not give sufficient information about how

hemodialysis is affecting the time-course of action of the antibiotic that a modeling and

simulation exercise can provide a bigger picture of the time course.

33
4.3 Humanized dosing in animal models to evaluate efficacy

Translational strategy to verify the dosing regimen in the clinic was applied to the

recently approved β-lactamase inhibitor with a partnering β-lactam, ceftazidime-

avibactam. The study evaluated the efficacy of human-simulated plasma exposure of 2

g ceftazidime and 0.5 g avibactam every 8 h administered as a 2-h infusion in

neutropenic and immunocompetent mice using both thigh and lung infection model of
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

several P. aeruginosa clinical isolates.89, 90 The PK of the drug was based on that

reported in humans.91 Given that the half-lives of the drugs are different in the mice from

that in the humans, human-like drug clearance was achieved by administering 5-10

mg/kg uranyl nitrate 3 days prior to treatment to induce a stable and reversible renal

impairment sufficient enough to delay drug elimination. Drug infusion was adjusted to

mimic the exposure in humans including peak drug concentration and half-life. The

bacterial densities in the animal studies were evaluated after 24 hours and the change

in log10 cfu/mL was determined against the starting density. The efficacies were

compared to ceftazidime alone.

In the murine lung infection model, the time at which the serum free drug

concentrations remains above MIC (fT>MIC) in the epithelial lining fluid (ELF) was also

evaluated in the neutropenic mice.90 Again, the efficacy was evaluated based on the

change in bacterial density after 24 hours. The study found that for isolates with

ceftazidime MIC of 32 μg/mL, the corresponding ELF fT>MIC was ≥ 12% and bacterial

reductions were observed. No reduction in bacterial density was observed in isolates

with ceftazidime MIC of 64 μg/mL. This example shows how simulation of human drug

PK in the murine model can be used to evaluate and extrapolate efficacy.

34
4.4 Modeling strategies to counter development of bacterial

resistance

Though the approach using PD surrogate indices, such as AUC24/MIC, has been

used to guide dosing of antimicrobial agents, some investigators have suggested that

these indices may not be appropriate for prediction of bacterial resistance.92-94 The

surrogate indices approach assumes that the MIC is stationary, which is not the case,
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

as bacteria will acquire resistance and adaptation when faced with antibiotic therapeutic

pressure. Lee et al. showed that few highly resistant mutants improve the survival of the

population’s susceptible constituents by emitting signal molecules to its surrounding to

turn on protective mechanisms, including upregulation of drug efflux pumps, during an

antimicrobial stressed environment.95 When bacteria are exposed to insufficient drug

concentrations to eradicate the resistant mutants, the population will acquire resistance

that result in a shift towards higher MIC.96 For example, the AmpC β-lactamase is

induced when the bacteria are exposed to β-lactams.97 Thus, simplification of dosing

schemes based on the PD surrogate indices may foster emergence of resistant

bacterial populations.

Baquero suggested a dangerous zone of drug concentration that potentially

confers survival advantage to these resistant mutants;92, 93 this zone is called the mutant

selection window (MSW).98-100 The MSW is the range of drug concentrations between

the MIC of the susceptible bacteria and the mutant prevention concentration, which is

the concentration that inhibits the growth of resistant mutants when evaluated at high

inoculum (>109 cfu/mL) using agar dilution method. Firsov and colleagues have shown

that the time spent within the MSW (TMSW) is a better predictor of emergence of

35
resistance in S. aureus following exposure to fluroquinolones.101, 102 For TMSW >20% of

the dosing interval, susceptibility of S. aureus against gatifloxacin, levofloxacin,

moxifloxacin and ciprofloxacin declined regardless of the AUC24/MIC ratio whereas

there was no change in bacterial susceptibility at TMSW<20% of dosing interval.101

Khachman et al. utilized fTMSW ≤ 20% and fTMSW ≤ 30% to estimate the probability of

attainment for various ciprofloxacin dosing regimens against several Gram-negative

pathogens in the intensive care unit (ICU).94 Their mutant prevention concentrations
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

were assumed to be 4- to 6-fold of MIC. With a population pharmacokinetic model of

ciprofloxacin in ICU patients, their simulations have shown that the PTA for fTMSW ≤ 20%

were less than 40% for MIC between 0.25 and 1 mg/L against P. aeruginosa and

Acinetobacter baumannii and the authors recommend against using ciprofloxacin for

these two bacteria types.94

Models of time-kill kinetics depicting both susceptible (S) and resistant (R)

bacteria are accomplished by setting two compartments for the two susceptibility states.

The overall total number of bacteria would then be the sum of these two state

populations. The assumption is that resistant bacteria come from de novo spontaneous

mutations.103-105 The mutation frequency is low at a rate of 10-8 to 10-6 per cycle and the

R/S ratio is set to 10-6 as their initial value in the model.106 Nielsen presented a two-

compartment model to describe the in vitro effect of moxifloxacin, vancomycin,

benzylpenicillin, cefuroxime and erythromycin against Streptococcus pyogenes.107 In

their assumption, the antibiotics have no effect on the persistent population. In a follow-

up study, Mohamed et al. introduced two additional compartments to regulate

resistance development in E. coli against gentamicin.108

36
In characterizing antibiotic effect on rise of resistant bacteria in piglets, Nguyen et

al. utilized the logistic growth model to describe both the S and R bacteria.109 They

argued that the rise in ciprofloxacin-resistant bacteria more likely comes from the

environment (i.e. ingestion of resistant bacteria). The difference in fitness between the S

and R was taken into account by assuming that the rate of growth of S is greater than

that of the R. The altruistic efforts of resistant bacteria to confer resistance to its

susceptible constituents comes at a cost to its own fitness.95 Nguyen and colleagues
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

found that the best fit to their data was when the model assumed that ciprofloxacin had

no activity on the resistant bacteria; alternative assumptions were such that the ratio of

EC50 values of ciprofloxacin against resistant and against susceptible populations were

4, 16 and 100. Their study recommends reducing intestinal exposure of piglets to

antibiotic residues in order to minimize the excretion and dissemination of resistant

bacteria to the environment in the animal feces.109

Other investigators have developed technique to quantify resistant bacterial

subpopulation from an in vitro time-kill experiment by plating the bacteria in agar plates

containing drug concentration three times or greater than the MIC, which ensures that

susceptible bacteria would not survive in the drug-containing agar plate.110 Tam et al.

applied the PK-PD model to describe the dynamics of the response of garenoxacin-

sensitve and resistant subpopulations of P. aeruginosa and S. aureus to fluctuating

quinolone drug concentrations.110, 111 The results of their study indicated that exposure

below a specific breakpoint allowed resistant subpopulation to grow rapidly. Jumbe et

al. utilized simulation approach in estimating the proliferation of resistant population in

37
insufficient antimicrobial therapy and evaluated dosing regimens required to prevent in

vivo amplification of resistant subpopulation.112

Other mechanistic models utilized three-state susceptibility, consisting of

susceptible, intermediate and resistant states, plus a fourth compartment to account for

the difference between the initial total bacterial burden and the initial conditions of the

other three states; this model was used to investigate colistin effect on P. aeruginosa.113
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

Their model also considered the ability of colistin to displace Mg2+ and Ca2+

competitively from the binding sites in the outer membrane; this cation displacement is

the mechanism of colistin killing. The fractional occupancy of the aggregate cations was

also defined in their model to take into account the relative dissociation rates of the

cations and colistin. Common with increasing complexity of models is that very often

parameters are fixed and simulation rather than fitting is often the approach taken, as

we discussed in previous section.

5. Conclusion

The PK-PD of antimicrobial agents and the tools available to evaluate drug

activities are well-established and tested through various candidates that eventually

became commercially available drug products. As there are tremendous unmet medical

needs for drug discovery and development programs targeting suppression of

resistance selection and eradication of multidrug resistant infection, PK-PD modeling

and simulation is well positioned to tackle the challenges of getting the right dosing

strategy for various patient populations. As we have shown here, many of the current

38
dosing regimens for the newer antimicrobial agents benefitted tremendously from

understanding of the PK-PD relationship between the drug and the bacterial infection.

6. Expert Opinion

In this article, we established the fundamental principles in antimicrobial PK and

PD, including modeling and simulation strategies. Table 3 summarizes these

methodologies, objectives of the approach, advantages and disadvantages and their


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

application in dose optimization. Many of the development of antimicrobial agents are

still based on establishing the PD surrogate index while modeling and simulation

approach is still underutilized. Empirical, mechanistic and semi-mechanistic PK-PD

models presented in the literature have shown good predictive value and are aptly

capable of iterative optimization of trial designs. We show examples wherein modeling

and simulation approach can give more information than the MIC-based PD indices,

which is based on the assumption that the MIC is stationary over time. It has been

shown that MIC can shift resulting from bacteria acquiring resistance, especially when

bacteria are exposed to a low concentration of drug that is insufficient to eradicate

them.96 Relying on a “snapshot” view of MIC for defining the PK-PD relationship for the

entire treatment duration can be misleading and also foster emergence of resistance.

Complementing the MIC-based approach, drug developers should also consider

utilizing in vitro time-kill curve information through modeling and simulation to define and

refine the PK-PD relationship.

Another trend that we have seen is that many of the refinement and optimization

of dosing regimens in special population are done by clinicians and researchers, outside

39
of drug development and after the drug has already been approved for marketing. Some

of the antimicrobial agents where dosing regimens in special population were

determined long after the drug was approved include polymixin B and collistin.114-116

Polymixin and colistin were antibiotics available in the 1950s but became out of use

when other antibiotics with better safety profiles were approved. These two drugs are

making a comeback recently due to diminishing therapeutic options in face of multidrug-

resistant Gram-negative bacteria.115 Recent report on population PK of polymyxin B in


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

critically ill patients indicates that the drug is best scaled by total body weight and that

renal function has no impact on polymyxin PK.114 Another example of antibiotic

combination where modeling and simulation in special population have been applied as

post-approval analysis is the combination of piperacillin and tazobactam in patients with

renal disease117-119 and in obesity.120-122 The population PK models in these populations

started to appear in the literature as late as 2010, even though the combination therapy

was originally approved in 1993. Even with the recently approved ceftolozane-

tazobactam combination, the literature on PK-PD model that utilizes the in vitro time-kill

kinetic information is still not available.

Thus, we see many opportunities moving forward to utilize state-of-the-art tools

to further our understanding of dosage optimization during drug development. Both

theories and applications examined here provide an overall understanding of how these

tools can be used to streamline drug development process of antimicrobial agents and

help make crucial decisions including clinical trial design. The search for regimens and

drug combinations for effective antibacterial activities can benefit tremendously from

modeling and simulation approach. More progress is needed to maximize the benefits

40
to the patients by implementing evidence-based treatment programs that evaluate and

incorporate all available information from in vitro studies, animal models of infections

and clinical trials.

Declaration of Interest
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

SKB Sy is currently an employee of Nektar Therapeutics. L Zhang discloses that the

work contributed to this manuscript took place while at the University of Florida and prior

to joining the US Food and Drug Administration. The opinions presented here are those

of the authors and no official support or endorsement by the FDA is intended or should

be inferred. The authors have no other relevant affiliations or financial involvement with

any organization or entity with a financial interest in or financial conflict with the subject

matter or materials discussed in the manuscript apart from those disclosed.

41
Abbreviations

ADME Absorption, distribution, metabolism and elimination

AUC Area under the concentration-time curve

CFR Cumulative fraction of response

cfu Colony forming unit

Cmax Maximum drug concentration within a dosing interval


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

Cmin Minimum drug concentration within a dosing interval

f Free drug concentration

ICU Intensive care unit

MIC Minimum inhibitory concentration

MSW Mutant selection window

PK-PD Pharmacokinetic-Pharmacodynamic

PTA Probability of target attainment

R Resistant

S Susceptible

SC Static concentration

T>MIC Time above MIC as a percentage of the dosing interval

T>TC Time above threshold concentration

TMSW Time spent within the mutant selection window

42
Bibliography

Papers of special note have been highlighted as:


* of interest
** of considerable interest

1. Asin-Prieto E, Rodriguez-Gascon A, Isla A. Applications of the

pharmacokinetic/pharmacodynamic (PK/PD) analysis of antimicrobial agents.J Infect


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

Chemother 2015;45:504-11.

2. Sy SK, Derendorf H. Pharmacometrics in bacterial infections. In: Schmidt S,

Derendorf H, eds. Applied Pharmacometrics. First ed. New York: Springer 2014:229-58.

* This article provides a concise overview of pharmacokinetic modeling approach

in general.

3. Nielsen EI, Friberg LE. Pharmacokinetic-pharmacodynamic modeling of

antibacterial drugs. Pharmacol Rev 2013;65:1053-90.

** This article is a very thorough review of antimicrobial PK-PD research up until

2013.

4. Heerdink ER, Urquhart J, Leufkens HG. Changes in prescribed drug doses after

market introduction. Pharmacoepidemiol Drug Saf 2002;11:447-53.

5. Sy SK, Wang X, Derendorf H. Introduction to pharmacometrics and quantitative

pharmacology with an emphasis on physiologically based pharmacokinetics. In:

Derendorf H, Schmidt S, eds. Applied Pharmacometrics. New York: Springer 2014:1-64.

6. Wu B, Sy SK, Derendorf H. Principles of applied pharmacokinetic–

pharmacodynamic modeling. In: Vinks AA, Derendorf H, Mouton JW, eds.

43
Fundamentals of Antimicrobial Pharmacokinetics and Pharmacodynamics: Springer

2014:63-79.

7. Jorgensen JH, Ferraro MJ. Antimicrobial susceptibility testing: a review of

general principles and contemporary practices. Clin Infect Dis 2009;49:1749-55.

8. Eagle H, Fleischman R, Musselman AD. The effective concentrations of penicillin

in vitro and in vivo for streptococci, pneumococci, and Treponema pallidum. J Bacteriol
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

1950;59:625-43.

9. Eagle H, Fleischman R, Musselman AD. Effect of schedule of administration on

the therapeutic efficacy of penicillin; importance of the aggregate time penicillin remains

at effectively bactericidal levels. Am J Med 1950;9:280-99.

10. Eagle H, Fleischman R, Levy M. "Continuous" vs. "discontinuous" therapy with

penicillin; the effect of the interval between injections on therapeutic efficacy. N Engl J

Med 1953;248:481-8.

11. Eagle H, Fleischman R, Levy M. On the duration of penicillin action in relation to

its concentration in the serum. J Lab Clin Med 1953;41:122-32.

12. Craig WA. Pharmacokinetic/pharmacodynamic parameters: rationale for

antibacterial dosing of mice and men. Clin Infect Dis 1998;26:1-10; quiz 11-2.

13. Mouton JW, Dudley MN, Cars O, et al. Standardization of

pharmacokinetic/pharmacodynamic (PK/PD) terminology for anti-infective drugs: an

update. J Antimicrob Chemother 2005;55:601-7.

14. Ambrose PG, Bhavnani SM, Rubino CM, et al. Pharmacokinetics-

pharmacodynamics of antimicrobial therapy: it's not just for mice anymore. Clin Infect

Dis 2007;44:79-86.

44
15. Bustamante CI, Drusano GL, Tatem BA, et al. Postantibiotic effect of imipenem

on Pseudomonas aeruginosa. Antimicrob Agents Chemother 1984;26:678-82.

16. Andes D, Craig WA. Animal model pharmacokinetics and pharmacodynamics: a

critical review. Int J Antimicrob Agents 2002;19:261-8.

17. Craig WA, Ebert SC. Killing and regrowth of bacteria in vitro: a review. Scand J

Infect Dis Suppl 1990;74:63-70.


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

18. Andes D, van Ogtrop ML, Peng J, et al.. In vivo pharmacodynamics of a new

oxazolidinone (linezolid). Antimicrob Agents Chemother 2002;46:3484-9.

19. Nielsen EI, Cars O, Friberg LE. Pharmacokinetic/pharmacodynamic (PK/PD)

indices of antibiotics predicted by a semimechanistic PKPD model: a step toward

model-based dose optimization. Antimicrob Agents Chemother 2011;55:4619-30.

20. Corvaisier S, Maire PH, Bouvier d'Yvoire MY, et al. Comparisons between

antimicrobial pharmacodynamic indices and bacterial killing as described by using the

Zhi model. Antimicrob Agents Chemother 1998;42:1731-7.

21. Rybak MJ, Lomaestro BM, Rotschafer JC, et al. Vancomycin therapeutic

guidelines: a summary of consensus recommendations from the infectious diseases

Society of America, the American Society of Health-System Pharmacists, and the

Society of Infectious Diseases Pharmacists. Clin Infect Dis 2009;49:325-7.

22. Moise-Broder PA, Forrest A, Birmingham MC, et al. Pharmacodynamics of

vancomycin and other antimicrobials in patients with Staphylococcus aureus lower

respiratory tract infections. Clin Pharmacokinet 2004;43:925-42.

45
23. Kullar R, Leonard SN, Davis SL, et al. Validation of the effectiveness of a

vancomycin nomogram in achieving target trough concentrations of 15-20 mg/L

suggested by the vancomycin consensus guidelines. Pharmacotherapy 2011;31:441-8.

24. Wong-Beringer A, Joo J, Tse E, et al. Vancomycin-associated nephrotoxicity: a

critical appraisal of risk with high-dose therapy. Int J Antimicrob Agents 2011;37:95-101.

25. Toutain PL, Bousquet-Melou A, Martinez M. AUC/MIC: a PK/PD index for


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

antibiotics with a time dimension or simply a dimensionless scoring factor? J Antimicrob

Chemother 2007;60:1185-8.

26. Booker BM, Smith PF, Forrest A, et al. Application of an in vitro infection model

and simulation for reevaluation of fluoroquinolone breakpoints for Salmonella enterica

serotype typhi. Antimicrob Agents Chemother 2005;49:1775-81.

27. Dudhani RV, Turnidge JD, Coulthard K, et al. Elucidation of the

pharmacokinetic/pharmacodynamic determinant of colistin activity against

Pseudomonas aeruginosa in murine thigh and lung infection models. Antimicrob Agents

Chemother 2010;54:1117-24.

28. Dudhani RV, Turnidge JD, Nation RL, et al. fAUC/MIC is the most predictive

pharmacokinetic/pharmacodynamic index of colistin against Acinetobacter baumannii in

murine thigh and lung infection models. J Antimicrob Chemother 2010;65:1984-90.

29. Guyonnet J, Manco B, Baduel L, et al. Determination of a dosage regimen of

colistin by pharmacokinetic/pharmacodynamic integration and modeling for treatment of

G.I.T. disease in pigs. Res Vet Sci 2010;88:307-14.

46
30. Bergen PJ, Bulitta JB, Forrest A, et al. Pharmacokinetic/pharmacodynamic

investigation of colistin against Pseudomonas aeruginosa using an in vitro model.

Antimicrob Agents Chemother 2010;54:3783-9.

31. Helfand MS, Bonomo RA. Current challenges in antimicrobial chemotherapy: the

impact of extended-spectrum beta-lactamases and metallo-beta-lactamases on the

treatment of resistant Gram-negative pathogens. Curr Opin Pharmacol 2005;5:452-8.


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

32. Leflon-Guibout V, Bonacorsi S, Clermont O, et al. Pyelonephritis caused by

multiple clones of Escherichia coli, susceptible and resistant to co-amoxiclav, after a 45

day course of co-amoxiclav. J Antimicrob Chemother 2002;49:373-7.

33. Leflon-Guibout V, Ternat G, Heym B, et al. Exposure to co-amoxiclav as a risk

factor for co-amoxiclav-resistant Escherichia coli urinary tract infection. J Antimicrob

Chemother 2002;49:367-71.

34. Sanders CC, Iaconis JP, Bodey GP, et al. Resistance to ticarcillin-potassium

clavulanate among clinical isolates of the family Enterobacteriaceae: role of PSE-1

beta-lactamase and high levels of TEM-1 and SHV-1 and problems with false

susceptibility in disk diffusion tests. Antimicrob Agents Chemother 1988;32:1365-9.

35. Bush K, Jacoby GA. Updated functional classification of beta-lactamases.

Antimicrob Agents Chemother 2010;54:969-76.

36. Strayer AH, Gilbert DH, Pivarnik P, et al. Pharmacodynamics of piperacillin alone

and in combination with tazobactam against piperacillin-resistant and -susceptible

organisms in an in vitro model of infection. Antimicrob Agents Chemother

1994;38:2351-6.

47
37. Livermore DM. Determinants of the activity of beta-lactamase inhibitor

combinations. J Antimicrob Chemother 1993;31 Suppl A:9-21.

38. Vanscoy B, Mendes RE, McCauley J, et al. Pharmacological basis of beta-

lactamase inhibitor therapeutics: tazobactam in combination with Ceftolozane.

Antimicrob Agents Chemother 2013;57:5924-30.

39. VanScoy B, Mendes RE, Nicasio AM, et al. Pharmacokinetics-


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

pharmacodynamics of tazobactam in combination with ceftolozane in an in vitro

infection model. Antimicrob Agents Chemother 2013;57:2809-14.

40. Louie A, Castanheira M, Liu W, et al. Pharmacodynamics of beta-lactamase

inhibition by NXL104 in combination with ceftaroline: examining organisms with multiple

types of beta-lactamases. Antimicrob Agents Chemother 2012;56:258-70.

41. Coleman K, Levasseur P, Girard AM, et al. Activities of ceftazidime and

avibactam against beta-lactamase-producing Enterobacteriaceae in a hollow-fiber

pharmacodynamic model. Antimicrob Agents Chemother 2014;58:3366-72.

42. Dose finding for a β-lactam – β-lactamase inhibitor combination

http://www.ema.europa.eu/docs/en_GB/document_library/Presentation/2015/01/WC500

179802.pdf

43. Mandell LA, Wunderink RG, Anzueto A, et al. Infectious Diseases Society of

America/American Thoracic Society consensus guidelines on the management of

community-acquired pneumonia in adults. Clin Infect Dis 2007;44 Suppl 2:S27-72.

44. Louie A, Liu W, Fikes S, et al. Impact of meropenem in combination with

tobramycin in a murine model of Pseudomonas aeruginosa pneumonia. Antimicrob

Agents Chemother 2013;57:2788-92.

48
45. Joly-Guillou ML, Wolff M, Farinotti R, et al. In vivo activity of levofloxacin alone or

in combination with imipenem or amikacin in a mouse model of Acinetobacter

baumannii pneumonia. J Antimicrob Chemother 2000;46:827-30.

46. Paul M, Dickstein Y, Schlesinger A, et al. Beta-lactam versus beta-lactam-

aminoglycoside combination therapy in cancer patients with neutropenia. Cochrane

Database Syst Rev 2013;6:CD003038.


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

47. Paul M, Lador A, Grozinsky-Glasberg S, et al. Beta lactam antibiotic

monotherapy versus beta lactam-aminoglycoside antibiotic combination therapy for

sepsis. Cochrane Database Syst Rev 2014;1:CD003344.

48. Levison ME. Pharmacodynamics of antimicrobial drugs. Infect Dis Clin North Am

2004;18:451-65.

49. Brentnall C, Cheng Z, McKellar QA, et al. Pharmacodynamics of oxytetracycline

administered alone and in combination with carprofen in calves. Vet Rec 2012;171:273.

50. Ziv G, Sulman FG. Binding of antibiotics to bovine and ovine serum. Antimicrob

Agents Chemother 1972;2:206-13.

51. Nouws JF, Vree TB, Termond E, et al. Pharmacokinetics and renal clearance of

oxytetracycline after intravenous and intramuscular administration to dairy cows. Vet Q

1985;7:296-305.

52. Buyck JM, Plesiat P, Traore H, et al. Increased susceptibility of Pseudomonas

aeruginosa to macrolides and ketolides in eukaryotic cell culture media and biological

fluids due to decreased expression of oprM and increased outer-membrane

permeability. Clin Infect Dis 2012;55:534-42.

49
53. Pruul H, McDonald PJ. Potentiation of antibacterial activity of azithromycin and

other macrolides by normal human serum. Antimicrob Agents Chemother 1992;36:10-6.

54. Ericsson HM, Sherris JC. Antibiotic sensitivity testing. Report of an international

collaborative study. Acta Pathol Microbiol Scand B: Microbiol Immunol 1971;217:Suppl

217:1+.

55. Aliabadi FS, Landoni MF, Lees P. Pharmacokinetics (PK), pharmacodynamics


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

(PD), and PK-PD integration of danofloxacin in sheep biological fluids. Antimicrob

Agents Chemother 2003;47:626-35.

56. Blaser J, Stone BB, Zinner SH. Two compartment kinetic model with multiple

artificial capillary units. J Antimicrob Chemother 1985;15 Suppl A:131-7.

57. Crandon JL, Nicolau DP. Human simulated studies of aztreonam and aztreonam-

avibactam to evaluate activity against challenging gram-negative organisms, including

metallo-beta-lactamase producers. Antimicrob Agents Chemother 2013;57:3299-306.

58. Bulitta JB, Ly NS, Yang JC, et al. Development and qualification of a

pharmacodynamic model for the pronounced inoculum effect of ceftazidime against

Pseudomonas aeruginosa. Antimicrob Agents Chemother 2009;53:46-56.

59. Sy SK, Beaudoin ME, Nichols WW, et al. Avibactam and ceftazidime combination

against multidrug resistant organisms in an in vitro pharmacokinetic/pharmacodynamic

model. 54th Intersci Conf Antimicrob Agents Chemother; 2014; Washington, DC; 2014.

p. abstr A-1344.

60. Sy SK, Beaudoin ME, Schuck VJ, et al. Modeling the potentiation of in vitro

aztreonam activities by avibactam against four beta-lactam-resistant bacterial strains.

50
53rd Intersci Conf Antimicrob Agents Chemother; 2013; Denver, CO; 2013. p. abstr A-

1014.

61. Gershenfeld NA. The nature of mathematical modeling. Cambridge: Cambridge

University Press, 1999.

62. Udekwu KI, Parrish N, Ankomah P, et al. Functional relationship between

bacterial cell density and the efficacy of antibiotics. J Antimicrob Chemother


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

2009;63:745-57.

63. Mouton JW, Vinks AA. Pharmacokinetic/pharmacodynamic modelling of

antibacterials in vitro and in vivo using bacterial growth and kill kinetics: the minimum

inhibitory concentration versus stationary concentration. Clin Pharmacokinet

2005;44:201-10.

64. Tam VH, Ledesma KR, Vo G, et al. Pharmacodynamic modeling of

aminoglycosides against Pseudomonas aeruginosa and Acinetobacter baumannii:

identifying dosing regimens to suppress resistance development. Antimicrob Agents

Chemother 2008;52:3987-93.

65. Treyaprasert W, Schmidt S, Rand KH, et al. Pharmacokinetic/pharmacodynamic

modeling of in vitro activity of azithromycin against four different bacterial strains. Int J

Antimicrob Agents 2007;29:263-70.

66. Zhuang L, Sy SK, Xia H, et al. Evaluation of in vitro synergy between vertilmicin

and ceftazidime against Pseudomonas aeruginosa using a semi-mechanistic

pharmacokinetic/pharmacodynamic model. Int J Antimicrob Agents 2015;45:151-60.

** This article is one of the first to successfully characterize whether a

combination is synergistic, antabonistic or additive using modeling.

51
67. Greco WR, Bravo G, Parsons JC. The search for synergy: a critical review from a

response surface perspective. Pharmacol Rev 1995;47:331-85.

*This article provides the mathematical foundation for qualifying synergistic

activities of combination therapy.

68. Maiwald T, Timmer J. Dynamical modeling and multi-experiment fitting with

PottersWheel. Bioinformatics 2008;24:2037-43.


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

69. Hengl S, Kreutz C, Timmer J, et al. Data-based identifiability analysis of non-

linear dynamical models. Bioinformatics 2007;23:2612-8.

70. Kohanski MA, Dwyer DJ, Collins JJ. How antibiotics kill bacteria: from targets to

networks. Nat Rev Microbiol 2010;8:423-35.

71. Topfer N, Jozefczuk S, Nikoloski Z. Integration of time-resolved transcriptomics

data with flux-based methods reveals stress-induced metabolic adaptation in

Escherichia coli. BMC Syst Biol 2012;6:148.

72. Muller M, dela Pena A, Derendorf H. Issues in pharmacokinetics and

pharmacodynamics of anti-infective agents: distribution in tissue. Antimicrob Agents

Chemother 2004;48:1441-53.

73. Plock N, Kloft C. Microdialysis--theoretical background and recent

implementation in applied life-sciences. Eur J Pharm Sci 2005;25:1-24.

74. Barbour A, Schmidt S, Rout WR, Ben et al. Soft tissue penetration of cefuroxime

determined by clinical microdialysis in morbidly obese patients undergoing abdominal

surgery. Int J Antimicrob Agents 2009;34:231-5.

52
75. Sauermann R, Burian B, Burian A, et al. Tissue pharmacokinetics of ertapenem

at steady-state in diabetic patients with leg infections. J Antimicrob Chemother

2013;68:895-9.

76. Hutschala D, Skhirtladze K, Kinstner C, et al. In vivo microdialysis to measure

antibiotic penetration into soft tissue during cardiac surgery. Ann Thorac Surg

2007;84:1605-10.
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

77. Velissaris D, Karamouzos V, Marangos M, et al. Pharmacokinetic changes and

dosing modification of aminoglycosides in critically ill obese patients: a literature review.

J Clin Med Res 2014;6:227-33.

78. de Kock L, Sy SK, Rosenkranz B, et al. Pharmacokinetics of para-aminosalicylic

acid in HIV-uninfected and HIV-coinfected tuberculosis patients receiving antiretroviral

therapy, managed for multidrug-resistant and extensively drug-resistant tuberculosis.

Antimicrob Agents Chemother 2014;58:6242-50.

79. Sy SK, de Kock L, Diacon AH, Werely CJ, et al. N-acetyltransferase genotypes

and the pharmacokinetics and tolerability of para-aminosalicylic acid in patients with

drug-resistant pulmonary tuberculosis. Antimicrob Agents Chemother 2015;59:4129-38.

80. Goff DA, Nicolau DP. When pharmacodynamics trump costs: an antimicrobial

stewardship program's approach to selecting optimal antimicrobial agents. Clin Ther

2013;35:766-71.

81. Mouton JW, Punt N, Vinks AA. Concentration-effect relationship of ceftazidime

explains why the time above the MIC is 40 percent for a static effect in vivo. Antimicrob

Agents Chemother 2007;51:3449-51.

53
82. Kong X, Wen JQ, Qi RF, et al. Diffuse interstitial brain edema in patients with

end-stage renal disease undergoing hemodialysis: a tract-based spatial statistics study.

Medicine 2014;93:e313.

83. Gupta V, Yassin MH. Infection and hemodialysis access: an updated review.

Infect Disord Drug Targets 2013;13:196-205.

84. Sowinski KM, Magner SJ, Lucksiri A, et al.. Influence of hemodialysis on


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

gentamicin pharmacokinetics, removal during hemodialysis, and recommended dosing.

Clin J Am Soc Nephrol 2008;3:355-61.

85. Teigen MM, Duffull S, Dang L, et al. Dosing of gentamicin in patients with end-

stage renal disease receiving hemodialysis. J Clin Pharmacol 2006;46:1259-67.

86. Begg EJ, Barclay ML, Duffull SB. A suggested approach to once-daily

aminoglycoside dosing. Br J Clin Pharmacol 1995;39:605-9.

87. Plajer SM, Chin PK, Vella-Brincat JW, et al. Gentamicin and renal function:

lessons from 15 years' experience of a pharmacokinetic service for extended interval

dosing of gentamicin. Ther Drug Monit 2015;37:98-103.

88. Zhuang L, Xia H, He Y, et al. Derendorf H. Evaluation of dosing strategy of

gentamicin in patients with end-stage renal disease receiving hemodialysis using semi-

mechanistic pharmacokinetic/pharmacodynamic model. J Antimicrob Chemother 2015

(in press).

89. Crandon JL, Schuck VJ, Banevicius MA, et al. Comparative in vitro and in vivo

efficacies of human simulated doses of ceftazidime and ceftazidime-avibactam against

Pseudomonas aeruginosa. Antimicrob Agents Chemother 2012;56:6137-46.

54
90. Housman ST, Crandon JL, Nichols WW, et al. Efficacies of ceftazidime-

avibactam and ceftazidime against Pseudomonas aeruginosa in a murine lung infection

model. Antimicrob Agents Chemother 2014;58:1365-71.

91. Li J, Knebel W, Riggs M, Zhou D, et al. Population pharmacokinetic modeling of

ceftazidime (CAZ) and avibactam (AVI) in healthy volunteers and patients with

complicated intra-abdominal infection (cIAI). 52nd Annual Interscience Conference on


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

Antimicrobial Agents and Chemotherapy; 2012 September 8-12, 2012; San Francisco,

CA; 2012. p. abstr A-634.

92. Baquero F. Resistance to quinolones in gram-negative microorganisms:

mechanisms and prevention. Eur Urol 1990;17 Suppl 1:3-12.

93. Baquero F, Negri MC. Strategies to minimize the development of antibiotic

resistance. J Chemother 1997 May;9 Suppl 3:29-37.

94. Khachman D, Conil JM, Georges B, et al. Optimizing ciprofloxacin dosing in

intensive care unit patients through the use of population pharmacokinetic-

pharmacodynamic analysis and Monte Carlo simulations. J Antimicrob Chemother

2011;66:1798-809.

95. Lee HH, Molla MN, Cantor CR, et al. Bacterial charity work leads to population-

wide resistance. Nature 2010;467:82-5.

96. Tam VH, Louie A, Deziel MR, et al. The relationship between quinolone

exposures and resistance amplification is characterized by an inverted U: a new

paradigm for optimizing pharmacodynamics to counterselect resistance. Antimicrob

Agents Chemother 2007;51:744-7.

55
**This article explains the relationship between resistance amplification and drug

dose, which provides a basis for high initial antibiotic dose in treatment regimens

to eradicate infection quickly before bacteria has the chance to develop drug

resistance.

97. Juan C, Macia MD, Gutierrez O, et al. mechanisms of beta-lactam resistance

mediated by AmpC hyperproduction in Pseudomonas aeruginosa clinical strains.


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

Antimicrob Agents Chemother 2005;49:4733-8.

98. Dong Y, Zhao X, Domagala J, et al. Effect of fluoroquinolone concentration on

selection of resistant mutants of Mycobacterium bovis BCG and Staphylococcus

aureus. Antimicrob Agents Chemother 1999;43:1756-8.

99. Dong Y, Zhao X, Kreiswirth BN, et al. Mutant prevention concentration as a

measure of antibiotic potency: studies with clinical isolates of Mycobacterium

tuberculosis. Antimicrob Agents Chemother 2000;44:2581-4.

100. Zhao X, Drlica K. Restricting the selection of antibiotic-resistant mutants: a

general strategy derived from fluoroquinolone studies. Clin Infect Dis 2001;33 Suppl

3:S147-56.

101. Firsov AA, Vostrov SN, Lubenko IY, et al. In vitro pharmacodynamic evaluation of

the mutant selection window hypothesis using four fluoroquinolones against

Staphylococcus aureus. Antimicrob Agents Chemother 2003;47:1604-13.

102. Drlica K. The mutant selection window and antimicrobial resistance. J Antimicrob

Chemother 2003;52:11-7.

103. Andersson DI. Persistence of antibiotic resistant bacteria. Curr Opin Microbiol

2003;6:452-6.

56
104. Andersson DI. The biological cost of mutational antibiotic resistance: any

practical conclusions? Curr Opin Microbiol 2006;9:461-5.

105. Andersson DI, Hughes D. Antibiotic resistance and its cost: is it possible to

reverse resistance? Nat Rev Microbiol 2010;8:260-71.

106. Jumbe LnN, Drusano GL. A model-based PK/PD antimicrobial chemotherapy

drug development platform to simultaneously combat infectious diseases and drug


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

resistance. In: Kimko HHC, Peck CC, eds. Clinical Trial Simulations: Springer 2011:251-

79.

107. Nielsen EI, Viberg A, Lowdin E, et al. Semimechanistic

pharmacokinetic/pharmacodynamic model for assessment of activity of antibacterial

agents from time-kill curve experiments. Antimicrob Agents Chemother 2007;51:128-36.

108. Mohamed AF, Nielsen EI, Cars O, et al. Pharmacokinetic-pharmacodynamic

model for gentamicin and its adaptive resistance with predictions of dosing schedules in

newborn infants. Antimicrob Agents Chemother 2012;56:179-88.

109. Nguyen TT, Guedj J, Chachaty E, et al. Mathematical modeling of bacterial

kinetics to predict the impact of antibiotic colonic exposure and treatment duration on

the amount of resistant enterobacteria excreted. PLoS Comput Biol 2014;10:e1003840.

110. Tam VH, Louie A, Deziel MR, et al. Bacterial-population responses to drug-

selective pressure: examination of garenoxacin's effect on Pseudomonas aeruginosa. J

Infect Dis 2005;192:420-8.

111. Tam VH, Louie A, Fritsche TR, et al. Impact of drug-exposure intensity and

duration of therapy on the emergence of Staphylococcus aureus resistance to a

quinolone antimicrobial. J Infect Dis 2007;195:1818-27.

57
112. Jumbe N, Louie A, Leary R, et al. Application of a mathematical model to prevent

in vivo amplification of antibiotic-resistant bacterial populations during therapy. J Clin

Invest 2003;112:275-85.

113. Bulitta JB, Yang JC, Yohonn L, et al. Attenuation of colistin bactericidal activity by

high inoculum of Pseudomonas aeruginosa characterized by a new mechanism-based

population pharmacodynamic model. Antimicrob Agents Chemother 2010;54:2051-62.


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

114. Sandri AM, Landersdorfer CB, Jacob J, et al. Population pharmacokinetics of

intravenous polymyxin B in critically ill patients: implications for selection of dosage

regimens. Clin Infect Dis 2013;57:524-31.

115. Nation RL, Li J, Cars O, et al. Framework for optimisation of the clinical use of

colistin and polymyxin B: the Prato polymyxin consensus. Lancet Infect Dis

2015;15:225-34.

116. Sandri AM, Landersdorfer CB, Jacob J, et al. Pharmacokinetics of polymyxin B in

patients on continuous venovenous haemodialysis. J Antimicrob Chemother

2013;68:674-7.

117. Asin-Prieto E, Rodriguez-Gascon A, Troconiz IF, et al. Population

pharmacokinetics of piperacillin and tazobactam in critically ill patients undergoing

continuous renal replacement therapy: application to

pharmacokinetic/pharmacodynamic analysis. J Antimicrob Chemother 2014;69:180-9.

118. Johnson CA, Halstenson CE, Kelloway JS, et al. Single-dose pharmacokinetics

of piperacillin and tazobactam in patients with renal disease. Clin Pharmacol Ther

1992;51:32-41.

58
119. Udy AA, Lipman J, Jarrett P, et al. Are standard doses of piperacillin sufficient for

critically ill patients with augmented creatinine clearance? Crit Care 2015;19:28.

120. Chung EK, Cheatham SC, Fleming MR, et al. Population pharmacokinetics and

pharmacodynamics of piperacillin and tazobactam administered by prolonged infusion

in obese and nonobese patients. J Clin Pharmacol 2015;55:899-908.

121. Sturm AW, Allen N, Rafferty KD, et al. Pharmacokinetic analysis of piperacillin
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

administered with tazobactam in critically ill, morbidly obese surgical patients.

Pharmacotherapy 2014;34:28-35.

122. Deman H, Verhaegen J, Willems L, et al. Dosing of piperacillin/tazobactam in a

morbidly obese patient. J Antimicrob Chemother 2012;67:782-3.

123. Drusano GL. Antimicrobial pharmacodynamics: critical interactions of 'bug and

drug'. Nat Rev Microbiol 2004;2:289-300.

124. Singh R, Kim A, Tanudra MA, et al. Pharmacokinetics/pharmacodynamics of a

beta-lactam and beta-lactamase inhibitor combination: a novel approach for

aztreonam/avibactam. J Antimicrob Chemother 2015;70:2618-26.

125. Safdar N, Andes D, Craig WA. In vivo pharmacodynamic activity of daptomycin.

Antimicrob Agents Chemother 2004;48:63-8.

126. Greenberg RN, Benes CA. Time-kill studies with oxacillin, vancomycin, and

teicoplanin versus Staphylococcus aureus. J Infect Dis 1990;161:1036-7.

127. Brink AJ, Richards GA, Cummins RR, Lambson J, et al. Recommendations to

achieve rapid therapeutic teicoplanin plasma concentrations in adult hospitalised

patients treated for sepsis. Int J Antimicrob Agents 2008;32:455-8.

59
128. Lodise TP, Butterfield JM, Hegde SS, et al. Telavancin pharmacokinetics and

pharmacodynamics in patients with complicated skin and skin structure infections and

various degrees of renal function. Antimicrob Agents Chemother 2012;56:2062-6.

129. Sakoulas G, Moise-Broder PA, Schentag J, et al. Relationship of MIC and

bactericidal activity to efficacy of vancomycin for treatment of methicillin-resistant

Staphylococcus aureus bacteremia. J Clin Microbiol 2004;42:2398-402.


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

130. Burgess DS, Frei CR, Lewis Ii JS, et al. The contribution of pharmacokinetic-

pharmacodynamic modelling with Monte Carlo simulation to the development of

susceptibility breakpoints for Neisseria meningitidis. Clin Microbiol Infect 2007;13:33-9.

131. Noreddin AM, El-Khatib WF, Aolie J, et al. Pharmacodynamic target attainment

potential of azithromycin, clarithromycin, and telithromycin in serum and epithelial lining

fluid of community-acquired pneumonia patients with penicillin-susceptible,

intermediate, and resistant Streptococcus pneumoniae. IntJ infect Dis 2009;13:483-7.

132. Matsumoto K, Shigemi A, Takeshita A, et al. Analysis of thrombocytopenic

effects and population pharmacokinetics of linezolid: a dosage strategy according to the

trough concentration target and renal function in adult patients. Int J Antimicrob Agents

2014;44:242-7.

133. Schentag JJ. Pharmacokinetic and pharmacodynamic surrogate markers: studies

with fluoroquinolones in patients. Am J Health Syst Pharm 1999;56:S21-4.

134. Aminimanizani A, Beringer P, Jelliffe R. Comparative pharmacokinetics and

pharmacodynamics of the newer fluoroquinolone antibacterials. Clin Pharmacokinet

2001;40:169-87.

60
135. Andes D, Craig WA. Pharmacodynamics of the new fluoroquinolone gatifloxacin

in murine thigh and lung infection models. Antimicrob Agents Chemother 2002;46:1665-

70.

136. Forrest A, Nix DE, Ballow CH, et al. Pharmacodynamics of intravenous

ciprofloxacin in seriously ill patients. Antimicrob Agents Chemother 1993;37:1073-81.

137. Ernst EJ, Klepser ME, Petzold CR, et al. Evaluation of survival and
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

pharmacodynamic relationships for five fluoroquinolones in a neutropenic murine model

of pneumococcal lung infection. Pharmacotherapy 2002;22:463-70.

138. Meagher AK, Passarell JA, Cirincione BB, et al. Exposure-response analyses of

tigecycline efficacy in patients with complicated skin and skin-structure infections.

Antimicrob Agents Chemother 2007;51:1939-45.

61
Table 1: Pharmacodynamic indices and their target values by antimicrobial class and

agents. Table modified from Asin-Prieto et al. (2015)1.

Antimicrobial Agent PD index Target value Ref


86
Aminoglycosides Cmax/MIC 10
β-lactams
123
Penicillins fT>MIC 50-60%
123
Cephalosporins fT>MIC 60-70%
123
Carbapenems fT>MIC 40-50%
124
Monobactams fT>MIC 60-70%
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

Clindamycin
27-30
Colistin fAUC/MIC 6 – 46 (variable depending on
bacteria type and strain)
Glycopeptides/lipopeptides
125
Daptomycin AUC/MIC 666
126, 127
Teicoplanin Cmin/MIC ≥10 for serious Gram-positive
≥20
128
Televancin AUC/MIC 219
129
Vancomycin AUC/MIC 400
Macrolides
130
Azithromycin fAUC/MIC 25
130
Clarithromycin fAUC/MIC 25
131
Teilithromycin fAUC/MIC 30
Oxazolidinones
132
Linezolid AUC/MIC 100
133, 134
Quinolones
135, 21
Gatifloxacin AUC/MIC 140
136, 22
Ciprofloxacin AUC/MIC 125
135, 21
Levofloxacin AUC/MIC 100
137, 23
Moxifloxacin AUC/MIC 95
Tetracyclines
138
Tigecycline fT>MIC 25

62
Table 2: Molecular classification (Ambler) of β-lactamases. Table modified from Bush et al. (2010)35.

Molecular β-lactam substrates Inhibitors Representative Comments


class (active enzymes
site)
A (serine) Penicillins, extended Clavulanic TEM, SHV, Hydrolysis of benzylpenicillin and
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

spectrum acid, CTX, KPC cephalosporins, TEM-30, TEM-50 and


cephalosporins, tazobactam, SHV-10 are resistant to sulbactam,
monobactams, sulbactam, tazobactam and clavulanic acid
carbenicillin avibactam
B (zinc) Carbapenems None NDM, IMP, VIM, Monobactams are impervious to IMP-1,
IND VIM-1 and IND-1
C (serine) Cephalosporins Avibactam AmpC, CMY Increased hydrolysis of cephalosporins
than benzylpenicillin, oxyimino-β-
lactams
D (serine) Carbapenems, Variable‡ OXA Hydrolyzes cloxacillin or oxacillin,
extended-spectrum oxyimino-β-lactams and carbapenems
cephalosporins,
cloxacillin

The commercially available β-lactamase inhibitors have limited coverage of class D enzymes.

63
Table 3: Summary of methodologies in characterizing antimicrobial agents PK-PD properties
Method/Approach Purpose Advantages/Disadvantages Applications to Dose Optimization
MIC and PD To determine PD Advantages: the methodology is relatively straight forward and the PD surrogate index is an important factor to
surrogate index surrogate index of processes to characterize PD surrogate index are well established. consider for dosing intervals, infusion
an agent Disadvantages: the empirical PD surrogate indices for an durations, as well as the dose of the
antimicrobial agent often show co-linearity, which can make dose antimicrobial agent. For newer antimicrobial
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

selection quite difficult. MIC in broth may not be representative of true agents, the characterization of their PD
MIC in biological fluids. surrogate index is mandatory.
Static time-kill To characterize the Advantages: The static time-kill curves are used to develop the PK- PK-PD models are developed from the
kinetics time-course of PD model, utilizing the information on changes in bacterial density in information from static time-kill kinetics for
bacterial response response to an antibiotic challenge maintained at constant the purpose of simulating bacterial
to antimicrobial concentration. response to non-static antibiotic
challenge at fixed Disadvantages: The static time-kill curves are very labor intensive. In concentration that mimics the kinetic
drug concentration combination studies, the number of these curves to characterize well behavior of the drug in vivo.
the combined drug effect can increase exponentially.
Dynamic time-kill To evaluate the Advantages: The dynamic time-kill kinetic studies mimic human Several dosing regimens can be evaluated
kinetics time-course of pharmacokinetic behavior of the drug. This set-up allows various with the dynamic time-kill kinetics.
bacterial density dosing regimens to be simulated in vitro. The characteristic PD
change when the surrogate index for a drug can be determined by evaluating several
antibiotic dosing regimens in the dynamic time-kill experiments.
concentration is Disadvantages: The hollow fiber equipment that automates the
changing according process of dynamic time-kill experiment is very expensive. Mere
to its half-life in information from dynamic time-kill experiments is insufficient for
humans model development. Thus, the data from dynamic time-kill kinetics
are often modeled together with data from the static time-kill kinetics.
Additionally, dynamic time-kill kinetics requires information on drug
half-life in humans. Consequently, the human pharmacokinetics of
the drug has to be available by the time the dynamic time-kill kinetic
studies are planned.
Microdialysis To determine time- Advantages: Microdialysis allows for direct measurement of the free There are a number of literatures that
course of free drug concentration at the site of infection. PD surrogate index based looked at the free drug concentration at the
unbound drug on microdialysis-collected information is more relevant especially if tissue sites. Evaluating the PD surrogate
concentration at the the PD surrogate index is determined from time-kill kinetic study. index based on the free drug concentration
tissue site Disadvantages: Validating the microdialysis procedure for the specific at the site of infection is a more direct
antimicrobial agent is a tedious process. Feasibility study is required method to evaluate whether sufficient
prior to running the microdialysis trial in humans. antimicrobial activities are occurring at the
site of infection.

64
Animal models of To determine the in Advantages: Humanized dosing in neutropenic or immunocompetent This procedure can be used to determine
humanized dose vivo efficacy using mouse thigh- or lung-infected model is pertinent to evaluate in vivo the duration of therapy as well as to
human-like drug efficacy of antimicrobial agents. One can evaluate human-like validate the dose and dosing interval
disposition pharmacokinetics in the animal model of infection. This procedure determined to achieve optimized PD
can provide information on the duration and length of therapy surrogate index target.
required to clear the infection.
Disadvantages: The procedure is predictive for drugs that are renally
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

cleared. For drugs with extensive hepatic metabolism, the human-like


pharmacokinetic may be difficult to mimic.
Modeling To develop PK-PD Advantages: Once the population pharmacokinetic model is This is a translational research application
model that developed in the relevant human population, predictions of the time- utilizing the information of human
describes the course of different dosing regimens and the extent of variability pharmacokinetic and linking in vitro
relationship between individuals can be done. The development of the PK-PD information of bacteria response to
between drug model can provide insights into how drug disposition in humans is pertinent drug concentrations.
concentration and affecting the dynamics of bacteria growth and kill.
its effect on Disadvantages: Human pharmacokinetic data is required for building
bacterial density the population pharmacokinetic model. The pharmacokinetics in
diseased patients may not be the same as healthy volunteers.
Model simulations To predict the Advantages: Simulations allow for investigation of the effect of Simulations are performed to provide better
bacteria dynamics various dosage regimens on the hypothetical growth and kill of confidence of the dosage regimen and
in various proposed bacteria in vivo, before designing or running a clinical trial. insights of the trial design, especially for a
dosing regimens Simulations can provide better confidence on the decided dosage phase III trial. In the clinic and hospital
regimen and to some extent the required duration of therapy before setting, simulations are used to predict the
running a trial. Simulations are also utilized to determine the best next dose for therapeutic drug
optimized “next” dose for a patient in a therapeutic drug monitoring monitoring of patients to achieve a specific
setting. target drug concentration.
Disadvantages: Prior to running a simulation, one needs to have a
model developed.
Probability of target To determine the Advantages: PTA provides statistical quantification of whether a Once the human pharmacokinetics is
attainment (PTA) percentage of the dosage regimen can achieve the PD surrogate index for the target determined including interindividual
patient population population. One can also evaluate the PTA for increasing MIC values variability, the utility of several dosage
that will achieve the and understand the limit of the dosage regimen against drug resistant regimens in attaining the required PD
required PD bacteria population. surrogate index can be evaluated and
surrogate index for Disadvantages: There are some indications that PTA values are not compared. PTA of a dosage regimen is
a specific dosage representative of the clinical outcomes. plotted against increasing MIC values.
regimen

65
Figure 1: Pharmacodynamic indices (24-hr AUC/MIC, Peak/MIC and Time above MIC) for ceftazidime in relationship to
the 24-hour log10 cfu of K. pneumoniae in the lung-infected neutropenic mice model. AUC, area under the curve; MIC,
minimum inhibitory concentration; cfu, colony forming unit.

Adapted with permission from Andes and Craig (2002)16.


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

66
Figure 2: Patterns of bactericidal activity of tobramycin, ciprofloxacin and ticarcillin in 6-hr constant concentration time-kill

kinetic studies. cfu, colony forming unit; MIC, minimum inhibitory concentration.

Adapted with permission from Craig and Ebert (1990)17.


Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

67
Figure 3: Determination of pharmacodynamic index of linezolid by fitting a sigmoidal Emax model and determining the

highest R2 value for the relationship between microbiological outcome and pharmacokinetic parameter. MIC, minimum

inhibitory concentration; cfu, colony forming unit; AUC, area under the curve; %T>MIC, percentage of the dosing interval
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

wherein drug concentration is above MIC.

Adapted with permission from Andes et al. (2002)18.

68
Figure 4: Prediction of antibacterial activity of vertilmicin, an aminoglycoside in development, under three dosing

regimens. The top graphs represent drug concentration of vertilmicin and the bottom graphs are hypothetical P.
Downloaded by [University of Nebraska, Lincoln] at 21:43 13 December 2015

aeruginosa response to the three dosing regimens. QD, once daily dosing; BID, twice daily dosing.

Adapted with permission from Zhuang et al (2015)66.

69

S-ar putea să vă placă și