Abstract
Translational PK/PD modeling has emerged as a critical technique for quantitative analysis of the relationship between dose, exposure and response of antibiotics. By combining model components for pharmacokinetics, bacterial growth kinetics and concentration-dependent drug effects, these models are able to quantitatively capture and simulate the complex interplay between antibiotic, bacterium and host organism. Fine-tuning of these basic model structures allows to further account for complicating factors such as resistance development, combination therapy, or host responses. With this tool set at hand, mechanism-based PK/PD modeling and simulation allows to develop optimal dosing regimens for novel and established antibiotics for maximum efficacy and minimal resistance development.
Introduction
The treatment options available to combat infectious diseases are becoming increasingly limited due to a rising incidence of resistance among bacteria against the currently used antibiotics. Additionally, the discovery and development of new antibiotics has slowed down because of limited incentives for the pharmaceutical industry in this therapeutic area and a high benefit-risk ratio of existing antibiotics [1]. This necessitates prudent use of the currently clinically available as well as newly developed antibiotics. One of the key requirements for preserving clinical efficacy of antibiotics and avoiding tolerance development is to identify the optimum dosing schedule for maximum bacterial kill and minimal emergence of resistance [2]. Although standard PK/PD indices have been used for this purpose with some clinical success, this approach is associated with several drawbacks. One of them is the high dependence upon the minimum inhibitory concentration (MIC), which is limited to a single efficacy endpoint that does not take into account the entire time course of effect and may vary between treated individuals and bacterial strains [3].
Translational PK/PD modeling and simulation has emerged as an alternative strategy to characterize the relationship between dose, exposure and response for identifying the most suitable dosing regimen. For example, infections with Mycobacterium tuberculosis are usually characterized by the presence of both multiplying and non-multiplying bacteria, and most antibiotics are usually more effective in killing multiplying rather than non-multiplying microorganisms. Mechanistic PK/PD models have successfully been applied to delineate the independent drug effects associated with killing of the multiplying and the non-multiplying bacterial populations [4]. In another example, different dosing regimens with similar exposure to colistin resulted in greater emergence of resistance with longer dosing intervals. Based on this observation, PK/PD models have subsequently been used for simulating untested dosing scenarios and developing an optimum dosing regimen for colistin [5].
It is the purpose of this review to provide an overview on the mechanism-based PK/PD models applied in translational PK/PD modelling for antibiotics.
PK/PD indices
For decades, the PK/PD relationships of antibiotics have been categorized with three different PK/PD indices, which rely on a summary measurement of in vivo drug exposure relative to the MIC, determined in vitro using serial dilution steps [6]. Usually, only free rather than total drug concentrations are considered as only free drug which is not bound to plasma proteins exerts the pharmacological activity. Standardized notations for these PK/PD indices are AUC/MIC, Cmax/MIC and T>MIC. AUC/MIC is the ratio of area under the free plasma concentration-time curve relative to MIC, Cmax/MIC is the ratio of free peak plasma concentration relative to MIC, and T>MIC is the cumulative percentage of a time period that the free concentration is above MIC. Antibiotics that are classified as having their efficacy driven by T>MIC show time-dependent killing which suggests that the antibacterial effect is at its maximum with concentrations just above the MIC and no further improvement in killing is obtained by further increasing the concentrations. Cmax/MIC best describes antibiotics which follow concentration-dependent killing and require maximum peak antibiotic concentration for maximum efficacy, whereas AUC/MIC is representative of the antibiotics which elicit both time- and concentration-dependent killing [3]. Although the MIC based approach has been widely used by clinicians for designing dosing regimens and can be useful to predict the effect of dose fractionation on antibacterial activity, this approach is associated with several limitations.
PK/PD indices rely heavily on MIC. Measurement of MIC is associated with substantial uncertainty and variability because of the two-fold dilution technique used for its assessment and the interpretation as implied binary response, which considers efficacy only above MIC and no killing below MIC. Furthermore, MIC varies across patient populations and bacterial strains, and may change with time in the same patient [7]. Thus, MIC-based approaches ignore the dynamics of bacterial killing with time. It is typically assumed that a PK/PD index determined in a preclinical species or population can be extrapolated to another patient population. However, PK/PD indices have been found to vary among different patient populations, species, and disease conditions, and one of the reasons is associated with differences in the pharmacokinetics of the antibiotic in these groups [8,9]. For example, in case of β-lactam antibiotics, the PK/PD index has been observed to change from T>MIC to AUC/MIC as the half-life increases in renally impaired patients. Similarly, decreases in half-life will shift the PK/PD index from AUC/MIC to T>MIC [10]. Thus, the value for PK/PD indices as guiding tool for dosage regimen design seems questionable.
One approach to overcome these drawbacks is the use of mechanism-based PK/PD modeling. Rather than relying on point estimates that integrate PK and PD factors of the host, the drug and the microorganism into one index, a mechanism-based PK/PD model has sufficient granularity to allow adjustment to changes in one of several of the PK and PD parameters as needed by changing clinical situations. In addition, PK/PD models reflect the graduate killing effect on bacteria with changing drug exposure rather than dichotomous all-or-nothing effects as implied by MIC values. Finally, PK/PD models consider the time-courses of bacterial growth and killing in relation to time courses of drug concentrations rather than time-integrated measures as used for the indices. Overall, these advantages allow for a more flexible, realistic and dynamic interplay of drug, host and microorganism-related factors that are ultimately predictive of bacterial killing and therapeutic efficacy.
Mechanism based PK/PD modeling
Mechanism based PK/PD models take into consideration the time course of bacterial count based on in vitro, in vivo and clinical information. These models are composed of three basic components: (i) a component that describes the bacterial growth kinetics, (ii) a component that describes the pharmacokinetics of the drug, and (iii) a component that integrates the effect of the drug concentration on bacterial turnover.
Bacterial Growth Kinetics
The simplest model component used for describing the bacterial growth kinetics consists of a single bacterial compartment with a first-order rate constant for bacterial replication (Krep) and a first order rate constant for death of the bacteria (Kdt). Eq. 1 and Fig. 1A describe the mono-exponential increase in microbial number seen in the absence of drug treatment [11], where N is the number of bacteria.
(1) |
Quite often only the net process between bacterial growth and kill is quantified, with a first-order rate constant Kgrowth = Krep − Kdt, also called Knet, since it is difficult to separately estimate both the replication and death rate constants with limited data [12]. The underlying assumption of this model is that the microbial population is homogenous having the same turnover rate constants, which is an oversimplification of the real scenario as microbial populations are usually known to be heterogeneous with multiple subpopulations with distinct metabolic states and drug resistance profiles [7].
In the absence of antibiotics, the bacterial number N eventually approaches a maximum Nmax when limitation in nutrients and bacterial density limit further growth. In this steady-state condition there is no net change in the microbial population. There are three ways to describe these capacity limited growth curves:
- b) Using a saturable, non-linear function [15]:
where VGmax is the bacterial maximum growth in bacterial count per time, and N50 is N at which the bacterial growth is half-maximal.(3) -
c) Using a model where transformation from the growing (S) to the resting stage (R) occurs when the total microbial count approaches the stationary phase [16] (Fig. 1B).
(4)
where S and R are the number of growing and resting bacteria, respectively. A high total bacterial load is assumed to potentiate the transformation from S to R. The transfer back to the growing stage (Krs) is usually assumed to be negligible during in vitro time-kill experiments and therefore in those cases fixed to 0.(5)
In some instances, additional delay functions have been introduced to characterize the growth delay observed during the first hours after the initiation of growth experiments when the microbes have not yet reached the logarithmic growth phase. For example, inverse mono-exponential functions with asymptotic increase such as 1 − e−δ×t have been used as multiplier for Krep or Kgrowth, where δ is a first-order rate constant characterizing the dissipation of the delay [17,18].
Pharmacokinetics
The pharmacokinetic model component is used for characterizing the concentration profile of a drug over a time period. Traditional compartmental modeling approaches are generally used to describe the pharmacokinetics with one- or multi-compartment systems. For example, in an in vitro chemostat-based PK/PD model system [18], the drug concentration can be described with a one-compartment model with mono-exponential decrease with time according to
(6) |
where C is the concentration of the Ke drug and is the first-order elimination rate constant of the drug.
The pharmacokinetic profile of a new drug is usually established in healthy subjects during the initial clinical phase of drug development. The pharmacokinetics may however be altered under pathophysiologic conditions in a diseased population. For example, drugs which are primarily renally cleared may reach higher systemic exposure in patients with renal impairment compared to healthy individuals. In these cases, creatinine clearance is typically found to be a significant covariate and can be used to guide patient customized dosing [19]. Disease conditions can also cause changes in the body fluids and protein binding which may ultimately affect tissue distribution and the free fraction of the drug [20], and may thus need to be considered in the pharmacokinetic model component of mechanism based PK/PD models.
Many microbial infections are localized in extravascular tissues rather than blood or plasma [21]. The concentration at the site of infection may in these cases be different from plasma concentrations due to drug distribution processes and may thereby complicate the accurate assessment of optimal dosing regimens [22]. For example, concentration-time profiles of antibiotics in epithelial lining fluid in the lungs was found to be different from plasma with concentration ratios ranging from <0.1 to 3 [23]. In addition to efficacy, organ or tissue concentrations may also be of interest with regard to toxicity [24]. Techniques like microdialysis have been used for measurement of drug concentrations in tissue/organs [25]. Tissue distribution of antibiotics may be predicted using physiologically-based pharmacokinetic (PBPK) modelling [26,27]. Recently, a PBPK model incorporating a multi-compartment permeability-limited lung model was used to simulate the pharmacokinetics of anti-tuberculosis agents in plasma, lungs and its sub-compartments where the mycobacteria reside in the host [28]. This model also provides a framework for predicting the lung concentrations of novel anti-tuberculosis agents.
Pharmacodynamics
The drug effect E can be modeled to either inhibit bacterial replication or potentiate bacterial killing. The relationship between drug concentration and its antimicrobial effect is usually described using an ordinary (γ = 1) or a sigmoidal (γ≠1) inhibitory Emax model. The general expression for the bacterial growth kinetics is then modified to Eq. 7:
(7) |
Drug effect decreasing the replication rate can then be modeled as
(8) |
Drug effect increasing the death rate can be modeled as
(9) |
Here Emax and Imax represent the maximum increase or decrease in effect, EC50 and IC50 represent the concentrations of drug that produce half of Emax and Imax , and γ1 and γ2 are the sigmoidicity coefficients. In Eq. 8 and 9, the effect corresponds to a unitless fractional change in the rate constant with Emax and Imax representing the maximum achievable fractional change.
Alternatively to these proportional effect models, the increase in the death rate can also be modeled as an additive effect (Fig. 2A):
(10) |
In Eq. 10, the effect corresponds to an additional kill-rate constant contributed by the drug with Imax representing the maximum achievable drug-imposed kill-rate constant having the unit 1/time.
Combination Therapy
Combination therapy of antibiotics has been found to be useful as it may improve efficacy, increase patient compliance by optimizing dosing schedules, reduce toxicity, and suppress the emergence of resistance [29]. There is high potential for pharmacodynamic drug-drug interactions when antibiotics of a combination therapy act on the same or different molecular targets (serial or parallel) in the same pathogen, resulting in additive, synergistic or antagonistic interaction [30]. The concentration-effect relationship for two-drug combinations results in a three dimensional response surface which is usually characterized quantitatively using the response surface analysis as described by Eq. 11 [31,32]:
(11) |
where C1 and C2 are the concentrations of drug 1 and drug 2 in the combination therapy; IC50,1 is the concentration for which the effect is half maximal for drug 1 when present alone; IC50,2 is the concentration for which the effect is half maximal for drug 2 when present alone; γ1 and γ2 are the sigmoidicity coefficients for drug 1 and drug 2, respectively; Imax is the maximum bacterial killing rate constant; ω is the interaction parameter; and I is the killing rate constant resulting from the combination therapy. The summation of the first and second term on the right side of the equation defines the additive effect whereas the third term is the drug interaction term. The combination effect is additive when ω = 0, is synergistic when ω >> 0, and is antagonistic when ω << 0.
The kinetics of bacterial killing for the combination therapy can then be described using Eq. 12 with modification of the drug effect term by taking a similar approach as described above for the response surface analysis [33].
(12) |
ω is the interaction parameter; and γ is the sigmoidicity coefficient for the combination therapy, under the assumption that in the response surface analysis, γ1 = γ2.
Antibiotic resistance
Antibiotic resistance can be mathematically explained either by an increase in IC50 or decrease in Imax. Prior knowledge about the mechanism of resistance can be used as a rationale for choosing the appropriate model. When reduced drug sensitivity can completely be overcome with a higher dose, an increase in IC50 is suggested as the probable mechanism for resistance. If even high doses are unable to achieve the maximum effect, however, then a decrease in Imax might be the more appropriate mechanism [12].
A variety of PK/PD models have been proposed for describing antibiotic resistance. These models can be broadly categorized into subpopulation approaches and adaptation approaches.
Subpopulation Approaches
The more commonly used subpopulation approaches assume that the total bacterial population is composed of several discrete subpopulations with different drug susceptibility as shown in Fig. 2B. These subpopulations are considered to be present even in the initial inoculum. Subsequent treatment with antibiotic leads to predominant killing of susceptible subpopulations along with selective replication of the less susceptible subpopulations resulting overall in regrowth. More than one differential equation is used simultaneously with each equation representing one bacterial subpopulation (often with different drug susceptibilities) [34]. A variation of the subpopulation model is one in which new drug resistant mutants are formed from the growing subpopulation with a first-order mutation rate during the experiment resulting in regrowth [35].
A modification of the sub-population approach could potentially be used to facilitate development of new generation anti-infectives which do not necessarily kill the bacteria. For example, Bezlotoxumab, a fully human monoclonal antibody, was found to decrease the rate of recurrence of Clostridium difficile infection in patients treated with standard-of-care antibiotics [36]. Bezlotoxumab acts by neutralizing the exotoxin B thereby averting its interaction with colonic cells and the associated inflammation. One of the possible approaches to characterize the PK/PD in this scenario could be a modified subpopulation model where the Bezlotoxumab induced toxin neutralization is modeled in parallel with the killing effect imposed by the standard-of-care treatment on the susceptible Clostridium difficile population.
Adaptation Approaches
An alternative to the subpopulation model in which the total bacteria are assumed to be a mixture of different populations is the adaptive resistance mechanism in which the initial drug susceptible population is gradually considered to evolve into a drug resistant population as shown in Fig. 2C. Only one differential equation is used to describe the rate of change of the total bacterial population. An adaptation factor (α) is introduced which may be dependent on time and concentration:
(13) |
where β describes the maximum adaptation factor and τ represents the rate of adaptation. Dependent on how the adaptation function is implemented in the PK/PD model, a gradual increase in time and concentration may result in an increase in IC50, decrease in Imax, or decrease in bacterial replication rate [37].
Identification of the true resistance mechanism based only on total bacterial counts and statistical modelling criteria have failed in majority of cases. Quantification of resistant populations will help in distinguishing between competing models and support the selection of the most appropriate model for bacterial resistance [38].
Inoculum Effect
The phenomenon that when a higher bacterial density of initial inoculum in an experiment leads to a reduced antibacterial effect is referred to as inoculum effect [39]. Some of the potential mechanisms for this phenomenon are (i) enzymatic inactivation of the drug, (ii) non-specific binding to the bacteria, and (iii) increased likelihood of the preexistence of subpopulations of resistant bacteria if the density of the inoculum is higher than the natural mutation rate of the bacteria. For example, the pharmacodynamic effect of ceftazidime, a β-lactam antibiotic, changed from time-dependent killing to concentration-dependent killing when inoculum density was increased [40]. This conversion in the PK/PD-index has been linked to the distribution and accumulation of β-lactamase in the biofilm of high density inocula which can hydrolyze β-lactam antibiotics.
Bacterial burden shows a wide variation in patients and therefore a PK/PD model which incorporates an inoculum effect might be more predictive in clinical scenarios. One of the models proposed to describe inoculum effects is based on the concept of quorum sensing. It assumes that all bacteria release signal molecules to communicate with each other which consequently results in reduced drug sensitivity [41].
Host Responses
In vivo models of bacterial infection are oftentimes established in immuno-compromised animals as they allow for proper estimation of antibiotic efficacy without any interference from the immune system. In many cases, the PK/PD indices needed to be achieved for efficacy in these models are higher compared to those obtained in immuno-competent animals [42]. In vivo infection models in animals with functional immune system, although less common, have also been used for assessing the efficacy of antibiotics. As the immune system of immunocompetent animals has the ability to kill bacteria, it is important to study the time course of this effect independently from antibiotic therapy in order to be able to delineate the intrinsic activity of the drug from that of the immune system.
The immune system effect can be assessed by three different approaches:
- a) Modulation of bacterial challenge: In this approach inocula with different initial density were used in the same type of animal infection model to quantify the antimicrobial effect associated with the immune system [43]. The immune system was successful in reducing the bacterial load when the initial inoculum was low; at higher initial inocula, however, the immune system’s capacity was overwhelmed resulting in net bacterial growth. This impact of immune system has been described quantitatively using mathematical models where the rate of change in bacterial count is equal to the difference between the growth rate of bacteria and the kill rate contributed by the antibacterial effect of the immune system [43]:
Here, Kir is the maximal kill rate induced by the immune system and N50 is the number of bacteria per g of tissue at which the immune-system mediated kill rate is half-maximal.(14) - b) Modulation of immune reactivity: An alternative strategy to quantify the contribution of the immune system towards bacterial reduction is to use animal infection models with different levels of immunosuppression [44]. A pneumonia mouse model with functional immune system was treated with different escalating doses of the immunosuppressant cyclophosphamide, thereby reducing the neutrophil counts by 20, 70 and 90%, respectively. The rate of change in the bacterial count was equal to the difference between the first order growth rate of bacteria and the saturable kill rate attributed to the number of available neutrophils:
ANC is the absolute neutrophil count and ANC50 is the ANC required to achieve 50% of maximal kill rate.(15) - c) Natural progression of disease: The effect of the immune system can also be included as a time-dependent first-order kill rate, in addition to the natural death rate as described by Eq.16, for example when immunocompetent animals are treated with vehicle control and establish adaptive immunity over time. This assessment may result in a slight overestimation of the impact of the immune system, however, if comparative data in immunocompetent and immune-compromised animals are not available to delineate the real immune system effect from natural bacterial death:
Here Kir is the maximum death rate constant induced by the immune system, t is the time, t50 is the time when the death rate constant is half of its maximum value, and γ is the sigmoidicity factor that determines the shape of the curve. The effect of the immune system is thus modeled as a gradual, time-dependent increase of the death rate constant until it reaches its maximum Kir.(16)
Bench to Bedside Translation of PK/PD Models for Anti-Infectives
The current paradigm for translation of antibacterial PK/PD from preclinical species to humans requires integration of the PK/PD index determined in animal models with human pharmacokinetics [45]. Stochastic simulation of human drug exposure using population pharmacokinetic models generates concentration-time profiles which are then used to compute the PK/PD index for each simulated subject. Based on the proportion of subjects with the desired value of the PK/PD index, probability of attaining the therapeutic target is predicted. This approach helps in making decisions regarding the choice of dosing regimens to be implemented in the clinic.
The use of mechanistic PK/PD models for translation from preclinical to clinical scenarios has been limited. PK/PD models based on in vitro static and dynamic kill curves have been successful in the prediction of in vivo effects in animal models [8]. However, there is still a knowledge gap regarding translation of PK/PD parameters from preclinical to clinical settings. For example, recently a multistate tuberculosis pharmacometric model describing different bacterial states of Mycobacterium tuberculosis was developed based on in vitro data [4]. For clinical implementation of this model [46], most of the parameters pertaining to the natural bacterial growth were fixed to the in vitro estimates; however the exposure-response parameters related to drug effect had to be estimated from clinical data and were different from the in vitro drug effect parameters. More research efforts are needed in this area to better facilitate the quantitative translation of mechanistic PK/PD models to clinical situations.
Development of a Translational PK/PD model for Antibiotics
Mechanism-based models for antibiotics are usually established in a stepwise fashion, in which the different model components are individually developed and integrated (Fig. 3). The typical steps include the following:
Development of a pharmacokinetic model in the studied population (animal models or humans), that captures the time course of free, pharmacologically active concentrations of the antibiotic, preferably in the relevant target tissue.
Development of a bacterial growth model that captures the bacterial growth kinetics and potential host response effects.
Integration of the bacterial growth model and the pharmacokinetic model component into a PK/PD model with a pharmacodynamic model component that links the dynamics of drug concentrations to bacterial turnover. This may include drug combination models, inocula effects, and resistance development.
Once the model has been established, it can be used for simulations to interpolate and extrapolate the observed experimental data to other, untested scenarios, such as different dosing regimens, or different patient populations [3,47]. It can also be used for quantitative comparisons of multiple drug candidates to identify lead candidates and support go-no go decisions [7].
Conclusions
Mechanism-based PK/PD models are increasingly been used in developing dosing strategies for antibiotics in drug development and clinical application. Although these models are simplified depictions of rather complex interactions of drug, microbe and host physiologic processes, they are able to capture the time course and magnitude of antibacterial effect in relation to bacterial growth, the pharmacokinetic and pharmacodynamic properties of the drug, and the chosen dosing regimen. Complicating factors such as resistance development, combination therapy, or host responses can be incorporated in the modeling approach. In situations where only limited data are available, system specific parameters describing for example growth characteristics may also be implemented based on prior knowledge [4]. With these tools at hand, translational PK/PD modelling and simulation may play a pivotal role in identifying the right balance between bacterial killing, adverse effects, and appearance of resistance, and may help identifying and optimizing dosing regimens for novel and established antibacterial agents.
Acknowledgements
This work was supported by grants R01AI120670, R01AI090810, and R01AI111449 by the National Institute of Allergy and Infectious Diseases of the National Institutes of Health, and by the American Lebanese Syrian Associated Charities (ALSAC).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest
The authors have no conflict of interest to declare.
References
- 1.Ventola CL. The antibiotic resistance crisis: part 1: causes and threats. P T. 2015;40:277–283. [PMC free article] [PubMed] [Google Scholar]
- 2.Ventola CL. The antibiotic resistance crisis: part 2: management strategies and new agents. P T. 2015;40:344–352. [PMC free article] [PubMed] [Google Scholar]
- 3.Nielsen EI, Friberg LE. Pharmacokinetic-pharmacodynamic modeling of antibacterial drugs. Pharmacol Rev. 2013;65:1053–1090. doi: 10.1124/pr.111.005769. [DOI] [PubMed] [Google Scholar]
- 4.Clewe O, et al. A multistate tuberculosis pharmacometric model: a framework for studying antitubercular drug effects in vitro. J Antimicrob Chemother. 2016;71:964–974. doi: 10.1093/jac/dkv416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bergen PJ, et al. Comparison of once-, twice- and thrice-daily dosing of colistin on antibacterial effect and emergence of resistance: studies with Pseudomonas aeruginosa in an in vitro pharmacodynamic model. J Antimicrob Chemother. 2008;61:636–642. doi: 10.1093/jac/dkm511. [DOI] [PubMed] [Google Scholar]
- 6.Trivedi A, et al. Applications of pharmacometrics in the clinical development and pharmacotherapy of anti-infectives. Expert Rev Clin Pharmacol. 2013;6:159–170. doi: 10.1586/ecp.13.6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wu B, et al. Principles of Applied Pharmacokinetic–Pharmacodynamic Modeling. In: Vinks AA, et al., editors. In Fundamentals of Antimicrobial Pharmacokinetics and Pharmacodynamics. Springer; New York: 2014. pp. 63–79. [Google Scholar]
- 8.Nielsen EI, et al. Pharmacokinetic/pharmacodynamic (PK/PD) indices of antibiotics predicted by a semimechanistic PKPD model: a step toward model-based dose optimization. Antimicrob Agents Chemother. 2011;55:4619–4630. doi: 10.1128/AAC.00182-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Craig WA, et al. Pharmacodynamics of amikacin in vitro and in mouse thigh and lung infections. J Antimicrob Chemother. 1991;27(Suppl C):29–40. doi: 10.1093/jac/27.suppl_c.29. [DOI] [PubMed] [Google Scholar]
- 10.Kristoffersson AN, et al. Simulation-Based Evaluation of PK/PD Indices for Meropenem Across Patient Groups and Experimental Designs. Pharm Res. 2016;33:1115–1125. doi: 10.1007/s11095-016-1856-x. [DOI] [PubMed] [Google Scholar]
- 11.Vaddady PK, et al. In vitro pharmacokinetic/pharmacodynamic models in anti-infective drug development: focus on TB. Future Med Chem. 2010;2:1355–1369. doi: 10.4155/fmc.10.224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Czock D, Keller F. Mechanism-based pharmacokinetic-pharmacodynamic modeling of antimicrobial drug effects. J Pharmacokinet Pharmacodyn. 2007;34:727–751. doi: 10.1007/s10928-007-9069-x. [DOI] [PubMed] [Google Scholar]
- 13.Campion JJ, et al. Pharmacodynamic modeling of the evolution of levofloxacin resistance in Staphylococcus aureus. Antimicrob Agents Chemother. 2005;49:2189–2199. doi: 10.1128/AAC.49.6.2189-2199.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.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;44:201–210. doi: 10.2165/00003088-200544020-00005. [DOI] [PubMed] [Google Scholar]
- 15.Meagher AK, et al. Novel pharmacokinetic-pharmacodynamic model for prediction of outcomes with an extended-release formulation of ciprofloxacin. Antimicrob Agents Chemother. 2004;48:2061–2068. doi: 10.1128/AAC.48.6.2061-2068.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Nielsen EI, et al. Semimechanistic pharmacokinetic/pharmacodynamic model for assessment of activity of antibacterial agents from time-kill curve experiments. Antimicrob Agents Chemother. 2007;51:128–136. doi: 10.1128/AAC.00604-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Nolting A, et al. Pharmacokinetic-pharmacodynamic modeling of the antibiotic effect of piperacillin in vitro. Pharm Res. 1996;13:91–96. doi: 10.1023/a:1016085402278. [DOI] [PubMed] [Google Scholar]
- 18.Budha NR, et al. A simple in vitro PK/PD model system to determine time-kill curves of drugs against Mycobacteria. Tuberculosis (Edinb) 2009;89:378–385. doi: 10.1016/j.tube.2009.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Patel N, et al. Determination of antibiotic dosage adjustments in patients with renal impairment: elements for success. J Antimicrob Chemother. 2010;65:2285–2290. doi: 10.1093/jac/dkq323. [DOI] [PubMed] [Google Scholar]
- 20.Gonzalez D, et al. The effect of critical illness on drug distribution. Curr Pharm Biotechnol. 2011;12:2030–2036. doi: 10.2174/138920111798808211. [DOI] [PubMed] [Google Scholar]
- 21.Zeitlinger MA, et al. Plasma concentrations might lead to overestimation of target site activity of piperacillin in patients with sepsis. J Antimicrob Chemother. 2005;56:703–708. doi: 10.1093/jac/dki284. [DOI] [PubMed] [Google Scholar]
- 22.Mueller M, et al. Issues in pharmacokinetics, and pharmacodynamics of anti-infective agents: kill curves versus MIC. Antimicrob Agents Chemother. 2004;48:369–377. doi: 10.1128/AAC.48.2.369-377.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Rodvold KA, et al. Penetration of anti-infective agents into pulmonary epithelial lining fluid: focus on antifungal, antitubercular, and miscellaneous anti-infective agents. Clin Pharmacokinet. 2011;50:689–704. doi: 10.2165/11592900-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 24.Mingeot-Leclercq MP, Tulkens PM. Aminoglycosides: nephrotoxicity. Antimicrob Agents Chemother. 1999;43:1003–1012. doi: 10.1128/aac.43.5.1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Joukhadar C, et al. Microdialysis. A novel tool for clinical studies of anti-infective agents. Eur J Clin Pharmacol. 2001;57:211–219. doi: 10.1007/s002280100301. [DOI] [PubMed] [Google Scholar]
- 26.Rowland M, et al. Physiologically-based pharmacokinetics in drug development, and regulatory science. Annu Rev Pharmacol Toxicol. 2011;51:45–73. doi: 10.1146/annurev-pharmtox-010510-100540. [DOI] [PubMed] [Google Scholar]
- 27.Edginton AN, et al. Defining the role of macrophages in local moxifloxacin tissue concentrations using biopsy data, and whole-body physiologically based pharmacokinetic modelling. Clin Pharmacokinet. 2009;48:181–187. doi: 10.2165/00003088-200948030-00004. [DOI] [PubMed] [Google Scholar]
- 28.Gaohua L, et al. Development of a Multicompartment Permeability-Limited Lung PBPK Model, and Its Application in Predicting Pulmonary Pharmacokinetics of Antituberculosis Drugs. CPT Pharmacometrics Syst Pharmacol. 2015;4:605–613. doi: 10.1002/psp4.12034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Jumbe NL, Drusano GL. A Model-Based PK/PD Antimicrobial Chemotherapy Drug Development Platform to Simultaneously Combat Infectious Diseases, and Drug Resistance. In: Kimko CHH, Peck CC, editors. In Clinical Trial Simulations: Applications, and Trends. Springer; New York: 2011. pp. 251–279. [Google Scholar]
- 30.Upton RN, Mould DR. Basic concepts in population modeling, simulation,, and model-based drug development: part 3-introduction to pharmacodynamic modeling methods. CPT Pharmacometrics Syst Pharmacol. 2014;3:e88. doi: 10.1038/psp.2013.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Greco WR, et al. Application of a new approach for the quantitation of drug synergism to the combination of cis-diamminedichloroplatinum, and 1-beta-D-arabinofuranosylcytosine. Cancer Res. 1990;50:5318–5327. [PubMed] [Google Scholar]
- 32.Greco WR, et al. The search for synergy: a critical review from a response surface perspective. Pharmacol Rev. 1995;47:331–385. [PubMed] [Google Scholar]
- 33.Zhuang L, 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;45:151–160. doi: 10.1016/j.ijantimicag.2014.09.017. [DOI] [PubMed] [Google Scholar]
- 34.Tam VH. Suppressing Resistance Development. In: Vinks AA, et al., editors. In Fundamentals of Antimicrobial Pharmacokinetics, and Pharmacodynamics. Springer; New York: 2014. pp. 135–151. [Google Scholar]
- 35.Li RC, et al. Pharmacodynamic modeling of bacterial kinetics: beta-lactam antibiotics against Escherichia coli. J Pharm Sci. 1994;83:970–975. doi: 10.1002/jps.2600830711. [DOI] [PubMed] [Google Scholar]
- 36.Yang Z, et al. Mechanisms of protection against Clostridium difficile infection by the monoclonal antitoxin antibodies actoxumab, and bezlotoxumab. Infect Immun. 2015;83:822–831. doi: 10.1128/IAI.02897-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Tam VH, et al. Modelling time-kill studies to discern the pharmacodynamics of meropenem. J Antimicrob Chemother. 2005;55:699–706. doi: 10.1093/jac/dki086. [DOI] [PubMed] [Google Scholar]
- 38.Jacobs M, et al. Distinguishing Antimicrobial Models with Different Resistance Mechanisms via Population Pharmacodynamic Modeling. PLoS Comput Biol. 2016;12:e1004782. doi: 10.1371/journal.pcbi.1004782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Udekwu KI, et al. Functional relationship between bacterial cell density, and the efficacy of antibiotics. J Antimicrob Chemother. 2009;63:745–757. doi: 10.1093/jac/dkn554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hengzhuang W, et al. High beta-lactamase levels change the pharmacodynamics of beta-lactam antibiotics in Pseudomonas aeruginosa biofilms. Antimicrob Agents Chemother. 2013;57:196–204. doi: 10.1128/AAC.01393-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bulitta JB, 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. doi: 10.1128/AAC.00489-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Andes D, Craig WA. In vivo pharmacodynamic activity of the glycopeptide dalbavancin. Antimicrob Agents Chemother. 2007;51:1633–1642. doi: 10.1128/AAC.01264-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Drusano GL, et al. Impact of burden on granulocyte clearance of bacteria in a mouse thigh infection model. Antimicrob Agents Chemother. 2010;54:4368–4372. doi: 10.1128/AAC.00133-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Guo B, et al. Quantitative impact of neutrophils on bacterial clearance in a murine pneumonia model. Antimicrob Agents Chemother. 2011;55:4601–4605. doi: 10.1128/AAC.00508-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Drusano GL, et al. Use of preclinical data for selection of a phase II/III dose for evernimicin, and identification of a preclinical MIC breakpoint. Antimicrob Agents Chemother. 2001;45:13–22. doi: 10.1128/AAC.45.1.13-22.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Svensson RJ, Simonsson U. Application of the Multistate Tuberculosis Pharmacometric Model in Patients With Rifampicin-Treated Pulmonary Tuberculosis. CPT Pharmacometrics Syst Pharmacol. 2016;5:264–273. doi: 10.1002/psp4.12079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Danhof M, et al. Mechanism-based pharmacokinetic-pharmacodynamic (PK-PD) modeling in translational drug research. Trends Pharmacol Sci. 2008;29:186–191. doi: 10.1016/j.tips.2008.01.007. [DOI] [PubMed] [Google Scholar]