Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Sep 1.
Published in final edited form as: Clin Microbiol Infect. 2020 May 5;26(9):1256.e1–1256.e8. doi: 10.1016/j.cmi.2020.04.034

In Pursuit of the Triple Crown: Mechanism-Based Pharmacodynamic Modeling for the Optimization of 3-Drug Combinations against KPC-Producing Klebsiella pneumoniae

Nikolas J Onufrak a,b,#, Nicholas M Smith c, Michael J Satlin d, Jürgen B Bulitta e, Xing Tan h, Patricia N Holden c, Roger L Nation f, Jian Li g, Alan Forrest a,*,, Brian T Tsuji c,*, Zackery P Bulman c,h,*,#
PMCID: PMC7641993  NIHMSID: NIHMS1591016  PMID: 32387437

Abstract

Objectives

Optimal combination therapy for Klebsiella pneumoniae carbapenemase (KPC)-producing K. pneumoniae (KPC-Kp) is unknown. The present study sought to characterize the pharmacodynamics (PD) of polymyxin B (PMB), meropenem (MEM), and rifampin (RIF) alone and in combination using a hollow fiber infection model (HFIM) coupled with mechanism-based modeling (MBM).

Methods

A 10-day HFIM was utilized to simulate human pharmacokinetics (PK) of various PMB, MEM, and RIF dosing regimens against a clinical KPC-Kp isolate, with total and resistant subpopulations quantified to capture PD response. A MBM was developed to characterize bacterial subpopulations and synergy between agents. Simulations using the MBM and published population PK models were employed to forecast the bacterial time-course and the extent of its variability in infected patients for 3-drug regimens.

Results

In the HFIM, a PMB single-dose (‘burst’) regimen of 5.53 mg/kg combined with MEM 8 g using a 3-hour prolonged infusion every 8 h and RIF 600 mg every 24 h resulted in bacterial counts below the quantitative limit within 24 h and remained undetectable throughout the 10-day experiment. The final MBM consisted of two bacterial subpopulations of differing PMB and MEM joint susceptibility and the ability to form a non-replicating, tolerant subpopulation. Synergistic interactions between PMB, MEM, and RIF were well-quantified, with the MBM providing adequate capture of the observed data.

Conclusions

An in vitro-in silico approach answers questions related to PD optimization as well as overall feasibility of combination therapy against KPC-Kp, offering crucial insights in the absence of clinical trials.

Introduction

The rise of multidrug-resistant bacteria is one of humanity’s greatest existential threats [1, 2]. Klebsiella pneumoniae carbapenemase (KPC)-producing K. pneumoniae (KPC-Kp) are a particularly troubling microorganism, maintaining pathogenicity and virulence while eroding the utility of most broad-spectrum antimicrobials [3]. As a result, KPC-Kp infections are associated with an average mortality rate of 41% [4], and maintain the ability to disseminate resistance mechanisms to other Gram-negative organisms [3].

In the search for effective therapies, revival of historic antimicrobial agents and repurposing of others has been met with mixed results [5]. While new options to combat KPC-Kp have emerged [6], polymyxins continue to be the most-prescribed agents [7]. However, numerous in vitro and clinical reports of the low barrier to polymyxin resistance in addition to dose-limiting toxicities have led to the consensus recommendation of combination therapy for KPC-Kp [8]. Such polymyxin-based combinations frequently employ a carbapenem [6, 8], though optimal dosing regimens are unknown, as is the potential for additional benefit with inclusion of a third agent. To this end, the rifamycins are an intriguing option, maintaining a unique mechanism of action and demonstrating synergistic activity in vitro [9]. With robust clinical trial evidence lacking, pharmacodynamic modeling and simulation affords the opportunity to explore therapeutic options using a translational approach, bridging experiments conducted at the bench to inferences made at the bedside [10, 11].

The objectives of the present study were to evaluate the pharmacodynamics (PD) of high-dose meropenem (MEM) regimens alone and in combination with polymyxin B (PMB) and/or rifampin (RIF) against KPC-Kp using the hollow fiber infection model (HFIM); develop a mechanism-based model (MBM) describing the independent and interactive PD of PMB, MEM, and RIF; and to forecast bacterial killing under various clinical scenarios using model-based simulations.

Methods

Bacterial Isolate

An ST258 KPC-producing K. pneumoniae isolate, KPC-Kp 9A, was used in all experiments. This organism was isolated from the blood of a 50 year-old woman who received salvage chemotherapy for refractory acute lymphoblastic leukemia, who subsequently developed septic shock and succumbed to the infection within 24 h. The isolate harbored the beta-lactamase genes blaKPC-2, blaSHV-11, and blaTEM-1, in addition to a premature stop codon in OmpK35 (with OmpK36 remaining wild-type). MIC testing by broth microdilution was performed per CLSI specifications [12], with values for PMB, MEM, and RIF provided in Table 1.

Table 1.

Simulated pharmacokinetics of agents employed in hollow fiber experiments

Agent MIC (mg/L) Dosing Regimen fCmax (mg/L) t1/2 (h)a %fT>MIC %fT>4xMIC fAUC24 (mg·h/L)
PMB 0.5 5.53 mg/kg single-dose 6.0 8 --- --- 61, 7.8b
3.33 mg/kg loading dose, followed by 1.43 mg/kg every 12 h 2.4 --- --- 35.9
MEM 16 2 g every 8 h (3-h infusion) 49 2.5 88 0 706
4 g every 8 h (3-h infusion) 98 100 39 1411
8 g every 8 h (3-h infusion) 196 100 85 2823
RIF 64 600 mg every 24 h 3.5 2.5 --- --- 14

fCmax, free-drug maximum concentration; t1/2, half-life; %fT>MIC, percentage of time free-drug concentrations remain above the MIC for each 24-hour period; %fT>4xMIC, percentage of time free-drug concentrations remain above 4 times the MIC for each 24-hour period; fAUC24, free-drug area under the concentration-time curve for each 24-hour period

a

Derived from [14] (PMB), [15] (MEM), and [15] (RIF)

b

fAUC0–24 = 61 mg·h/L, fAUC24–48 = 7.8 mg·h/L, negligible PMB exposure thereafter

Hollow Fiber Infection Model Experiments

A 10-day in vitro HFIM was used to define the PD activities of PMB, MEM, and RIF alone and in 2- and 3-drug combinations against KPC-Kp 9A, as described previously [9]. PMB and RIF were purchased from Sigma-Aldrich (St. Louis, MO) and MEM was purchased from AK Scientific, Inc. (Union City, CA). HFIM experiments were conducted against a starting inoculum of ~108 CFU/mL over 240 h with bacterial aliquots obtained after 0, 1,2, 3, 4, 6, 24, 26, 28, 30, 48, 50, 52, 54, 72, 74, 76, 78, 96, 144, 192, and 240 h to perform viable bacterial colony counting. Population analysis profiles (PAPs) were used to detect antibiotic-resistant subpopulations using bacterial aliquots obtained after 0, 24, 48, 72, 96, 144, 192, and 240 h of antibiotic exposure in the HFIM. PAPs were performed using Mueller-Hinton agar plates containing varying concentrations of PMB (0.5, 1, 4, and 10 mg/L) or MEM (4, 16, and 64 mg/L) and incubated for 48 h before counting colonies [13]. Plates containing PMB were made fresh prior to each HFIM experiment and plates containing MEM were prepared every 48 h during the experiment; all PAP plates were stored at 4 °C prior to use. Select colonies grown on meropenem PAP plates were confirmed to have meropenem MICs exceeding the plate’s drug concentration, thus supporting the ability of this approach to accurately quantify meropenem-resistant subpopulations. To confirm Pertinent antibiotic regimens used in the HFIM experiments simulated the human pharmacokinetics (PK) of PMB [14], MEM [15], and RIF [15], and are provided in Table 1.

Mechanism-Based Pharmacodynamic Modeling

The HFIM data were modeled in a stepwise fashion using the Monte Carlo Parametric Expectation-Maximization algorithm (PMETHOD = 4) in S-ADAPT (version 1.57), with pre-/post-processing facilitated by S-ADAPT TRAN [17, 18]. Single-drug regimens were initially fit to discern appropriate bacterial characteristics, killing functions, and parameter estimates. Dual-therapy regimens were then introduced to explore synergistic interactions, culminating in the addition of the triple-drug combination. Bacterial growth was parameterized via a two-state lifecycle model assuming transition between vegetative and replicative states [19]. Differing growth rates were explored for differing bacterial subpopulations to account for fitness costs of less-susceptible populations. Hypothetical signal molecules representing communication between bacterial cells were invoked to describe an inoculum effect wherein high bacterial densities slow the rate of replication, thereby mitigating beta-lactam-induced killing [19]. Antimicrobial effects were parameterized using sigmoidal Emax models except in the case of direct PMB killing, where effect was governed by a second-order process [20]. PD interactions between agents were characterized using both subpopulation and mechanistic synergy, where the former describes killing of a subpopulation resistant to one agent but susceptible to another and the latter acknowledges the capacity for an agent to enhance the killing effect of another [21]; in the case of mechanistic synergy, sigmoidal Emax models were utilized. Additional details regarding MBM components are provided in the Supplementary Materials. The final MBM was selected based on a combination of diagnostic plots, objective function value, and biologic plausibility of the parameter estimates.

Model-Based Simulations

Using the final MBM, clinical trial simulations were performed to predict typical microbiologic outcomes in patients receiving PMB-MEM-RIF 3-drug combinations. Published population PK models [14, 15 Table 1, 22] were leveraged to produce free-drug exposures for a population of 500 infected patients receiving the same dosing regimens as those studied in the HFIM, thereby accounting for inter-patient variability in PK. These exposures were simultaneously linked to the developed MBM for prediction of the time-course of bacterial killing over 10 days for each simulated patient. To account for differences in protein binding between patients, a log-normal 20% coefficient of variation (CV) was applied to the unbound fractions utilized for each drug (PMB – 0.42 [14]; MEM – 0.98 [15]; RIF – 0.20 [16]). For each population PK model, covariate values assumed the population mean. All simulations were conducted using the RxODE package in R (version 3.6.1) [23].

Results

Hollow Fiber Infection Model Experiments

Data from the HFIM that included MEM doses at 2 g every 8 h as a 3-h infusion have been published previously, as have corresponding PK validations [9]; experiments including MEM dosed as 4 g or 8 g every 8 h as a 3-h infusion represent new data, with all HFIM arms included in the MBM. Observed HFIM data are shown in Figure 1, with PAPs data provided as Figure 2. Monotherapies of MEM 4 g and 8 g caused bacterial reductions of ~0.5 and ~2.0 log10 CFU/mL within the first 3 h, respectively, though like the 2 g regimen, regrowth was observed by 24 h. PAPs did not display appreciable MEM resistance proliferation despite regrowth. In two-drug combinations with a PMB ‘burst’ regimen (single dose of 5.53 mg/kg), MEM dosed at 4 g caused a maximum bacterial reduction of ~2.7 log10 CFU/mL at 4 h but again regrew within 24 h, with amplification of polymyxin resistance based on PAPs data. When the PMB ‘burst’ regimen was paired with an 8 g MEM dose, bacterial counts were repressed to between ~2 and ~4 log10 CFU/mL throughout the 240-h experiment. The triple-drug combination of a PMB ‘burst’, MEM 2 g every 8 h as a 3-h infusion, and 600 mg RIF every 24 h achieved >5 log10 CFU/mL reductions within the first 6 h, with bacterial counts remaining around 3 log10 CFU/mL throughout the remainder of the experiment. In contrast, no viable bacteria were detected after 24 h following the 3-drug combination with an enhanced MEM dose of 8 g.

Figure 1.

Figure 1.

Observed (circles) and model-fitted (lines) viable total bacterial counts for 1-, 2-, and 3-drug regimens employed in the hollow fiber infection model

Figure 2.

Figure 2.

Population analysis profiles of high-dose meropenem-containing regimens studied within the hollow fiber infection model

Mechanism-Based Pharmacodynamic Modeling

The developed MBM incorporating the killing and regrowth of bacteria following exposure to PMB, MEM, and RIF as 1-, 2-, or 3-drug regimens adequately described the observed data (Figure 1). The final model consisted of two viable bacterial subpopulations (dually PMB- and MEM-susceptible [PMBS-MEMS] and dually PMB- and MEM-resistant [PMBR-MEMR]) in addition to a non-replicating tolerant subpopulation linked to the PMBR-MEMR subpopulation. The direct killing effects of PMB were parameterized as different second-order rate constants for the viable subpopulations, whereas differing maximal killing rates and EC50 values were employed for MEM effects. PMB displayed mechanistic synergy, lowering MEM EC50 values by a maximum of 56.5%, with an EC50 for the synergy estimated as a PMB concentration of 2.52 mg/L. While RIF appeared to exert no direct antimicrobial effect, the observed HFIM data suggested its combination with PMB and MEM provided synergistic benefit, particularly in terms of driving bacterial eradication. This was accounted for in the model via concentration-dependent RIF inhibition of the rate constant for bacterial transition to a tolerant subpopulation. A schematic representation of the MBM is provided as Figure 3, with corresponding parameter definitions, estimates, and standard errors provided in Table 2. Model diagnostics are included as Supplementary Figures S1 and S2.

Figure 3. Mechanism-based model schematic summarizing key elements of the parameterization.

Figure 3.

S, susceptible; R, resistant; RIFinh, rifampin-mediated inhibitory effect; PMBsyn, polymyxin B-mediated synergistic effect; INHkill, sigmol, inhibitory inoculum effect due to hypothetical signaling molecules

Note: parameter definitions and estimates are provided in Table 2, with additional information provided as Supplementary Material.

Table 2.

Parameter definitions and estimates

Parameter Symbol Units Value SE%
Parameters Governing Bacterial Growth

Mean growth time of susceptible sub-population MGTS min 117 5.66
Mean growth time of resistant sub-population MGTR min 79.8 2.91
Log starting inoculum LogCFU0 log10(cfu/mL) 8.28 1.25
Maximum population LogCFUmax log10(cfu/mL) 10.6 0.605
Mutation frequency of resistant sub-population LogMFR - −4.97 1.42
Log rate of tolerant cell formation Logktol log10(1/h) −2.36 15.1
Log rate of tolerant cell reversion Logkrev log10(1/h) −1.85 8.46
Parameters Governing Meropenem Killing Effect

Max first-order killing effect for MEMS Smax,S 1/h 31.1 8.18
Max first-order killing effect for MEMR Smax,R 1/h 1.52 20.0
Meropenem concentration for 50% of Smax,S SC50,S mg/L 46.4 11.0
Meropenem concentration for 50% of Smax,R SC50,R mg/L 75.4 3.16
Shape parameter for meropenem killing Hillm - 2.32 11.0
Max inhibition of MEM killing by signal molecules SIGmax % 97.4 3.74
Log signal molecule concentration for 50% of SIGmax LogSIG50 - 5.26 1.57
Parameters Governing Polymyxin B Killing Effect

Second-order killing effect for PMBS kkill,S L/mg/h 70.1 4.44
Second-order killing effect for PMBR kkill,R L/mg/h 2.13 14.1
Parameters Governing Rifampin Inhibition Effect

Maximum inhibition of tolerant cell formation GRmax,RIF % 97.9 3.61
Rifampin concentration for 50% of max inhibition GR50 mg/L 0.129 16.3
Shape parameter for rifampin effect Hillgr - 3.09 15.5
Parameters Governing Increased Meropenem Permeability by Polymyxin B

Maximum decrease in meropenem SC50 SYNmax % 56.5 45.8
Polymyxin B concentration for 50% of SYNmax SYN50 mg/L 2.52 -
Shape parameter for polymyxin B effect on synergy Hillsyn - 0.0444 22.9
Residual Variance

Additive residual variance on log10 scale σ log10(cfu/mL) 0.578 4.32

Note: Additional information regarding the parameterization is provided as Supplementary Material

Model-Based Simulations

Results of model-based simulations depicting anticipated PD responses in patients for each of the MEM doses employed in 3-drug combinations using PMB as a single-dose ‘burst’, as well as an interpolated 3-drug combination using 4 g MEM are provided in Figure 3. Predicted bacterial killing was similar across the three MEM dosing regimens throughout much of the first 24 h, suggesting rapid initial killing with a subsequent decrease in killing rate for all 3 regimens as the predominant susceptible subpopulation was eliminated, consistent with the rapid bactericidal activity of PMB. The typical (population mean) predictions suggest that the triple-drug combination using 2 g MEM would yield substantial regrowth within 48 h (Figure 3A), whereas using 4 g MEM will delay such regrowth to day 6–7 of therapy, albeit with substantial variability, with the 80% prediction interval ranging from complete eradication to complete regrowth (Figure 3B). Notably, employing 8 g MEM predicts bacterial counts to remain suppressed (<2 log10 CFU/mL) throughout the 10-day period (Figure 3C).

Discussion

Amidst a rising tide of antimicrobial resistance, determination of viable therapeutic options is desperately needed. The present study leveraged state of the art in vitro experimentation and in silico modeling and simulation to quantify the ability of PMB, MEM, and RIF combinations to combat a KPC-Kp clinical isolate. Here it was found that a PMB ‘burst’ regimen, consisting of a single 5.53 mg/kg dose, combined with high-dose MEM (8 g provided as a 3-h prolonged infusion every 8 h) and RIF 600 mg every 24 h is capable of eradicating this highly resistant organism. The developed MBM sufficiently captured the observed data from the HFIM experiments, and exemplifies the difficulty in producing clinically achievable exposures of PMB and MEM necessary to treat high-burden KPC-Kp infections.

This is the first known attempt at mathematically modeling the PD of 3-drug combinations against Gram-negative bacteria. The results of the 1- and 2-drug regimens are in line with prior HFIM reports, albeit with different organisms [24, 25]. Rapid regrowth of PMB-resistant subpopulations has been observed following extensive initial killing of both Acinetobacter baumannii [24] and Escherichia coli [25], which may be attributed to a change in the ionic character of the outer membrane and/or a loss of LPS. The synergistic effect of PMB-MEM combination has also been noted [24], with the hypothesis that PMB permeabilizes the bacterial outer membrane, inducing the uptake of coadministered antimicrobials. The addition of RIF to a combination of PMB and MEM has previously demonstrated enhanced activity in a one-compartment in vitro model against two KPC-Kp isolates, though no mathematical modeling was performed [26].

Unexpectedly, a bacteriostatic phenotype was observed between 2–5 log10 CFU/mL in the HFIM experiments, leading to the incorporation in the MBM of tolerant bacterial cells arising from the PMBR-MEMR subpopulation that could neither replicate nor be killed. As the experimental data suggested that RIF-containing 3-drug regimens were not subject to this dormancy, a RIF-mediated inhibition of the formation of tolerant cells was introduced. Bacterial persisters have been proposed in the literature as a subpopulation of slow-growing or dormant cells that can transiently withstand antimicrobial exposure while otherwise maintaining drug susceptibility upon drug withdrawal, representing a unique entity from resistant cells [27]. Coupled with previous reports of tolerance when KPC-Kp was exposed to carbapenem monotherapy [28], the present study provides a potential mechanistic interpretation of tolerant cell formation and prevention, though these results should be considered hypothesis-generating and warrant additional study.

Stochastic simulations leveraging the developed MBM combined with published population PK indicate an average response that nearly eradicates KPC-Kp when a PMB ‘burst’ is combined with MEM 8 g as a 3 h prolonged infusion and RIF 600 mg. With regimens containing a MEM dosing regimen of 4g as a 3 h prolonged infusion, the corresponding 80% prediction interval infers extreme variability in the extent of bacterial regrowth, emphasizing the inherent difficulty of simultaneously achieving each agent’s optimal exposure given the extent of PK variability in infected patients. As the treating clinician must balance population-level knowledge within the context of their individual patient, this signals the necessity of therapeutic drug management to ensure preclinically and clinically derived PK-PD targets are achieved. Indeed, such practice is advocated in international guidelines for PMB [8], is increasingly recommended yet underutilized for β-lactams [29], and has been shown feasible for RIF [30]. If drug concentration measurements are not available, the developed MBM may still serve as a tool for simulating overall probability of a given bacterial endpoint with a desired PMB, MEM, and/or RIF dosing regimen. Importantly, modeling of the entire bacterial time-course affords predictions of antimicrobial efficacy irrespective of an MIC, allowing great flexibility in application based on the preferred clinical effect. However, direct clinical translation must be cautioned given the high doses of PMB and MEM employed here, which may result in unacceptably high toxicity rates and require additional testing.

There are several limitations to the present analyses. First is the use of a single bacterial isolate. However, isolate KPC-Kp 9A belongs to genetic sequence type ST258 and harbors blaKPC-2, and is thus characteristic of the most prevalent KPC-Kp and of that responsible for pandemic expansion of the organism [31]. Second, it is acknowledged that the HFIM system is devoid of an immune system component, which is expected to exert an intrinsic bacterial killing effect, though it must be noted that the patient from whom the isolate was derived was immunocompromised. Third, a single RIF dosing regimen of 600 mg every 24 h was employed, and while this is the most commonly utilized regimen in the clinic, emerging evidence correlates higher doses with greater antimicrobial effect [32]. Given the present results, future experimentation with higher and varied RIF doses appears warranted. Finally, a single starting inoculum of ~8 log10 CFU/mL was used, likely handicapping antimicrobial efficacy. Combined with an OmpK35 deletion creating a higher barrier to bacterial eradication, these facets coalesce to engender a “worst case” scenario and thus a conservative estimate of each regimen’s clinical adequacy.

In conclusion, KPC-Kp was highly resilient to 3-drug combinations involving PMB, MEM, and RIF. The MBM characterized direct bacterial killing effects and synergy between agents against phenotypically distinct subpopulations, suggesting that resistance suppression was possible, though at dosing regimens beyond those likely to be palatable. In the absence of clinical trials, this integrated approach holds great promise for discovering PD-optimized antimicrobial combination therapy and inferring its feasibility, providing crucial insights for an increasingly drug-resistant world.

Supplementary Material

1

Figure 4.

Figure 4.

Simulated bacterial time-course for triple-drug combinations using the mechanism-based pharmacodynamic model and pharmacokinetic parameters derived from infected patients. Solid lines represent typical (population mean) predictions; shaded regions represent 80% prediction intervals.

Acknowledgements

This project was supported by the National Institutes of Health, National Institute of Allergy and Infectious Diseases grant R01AI111990. Z.P.B. was supported by the National Center for Advancing Translational Sciences, National Institutes of Health, under Grant KL2TR002002. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

This manuscript is dedicated to the memory of Dr. Alan Forrest, whose steadfast commitment to the discipline and passion for teaching has guided many investigators throughout various stages of their careers. He will be remembered as a giant in antimicrobial chemotherapy who worked to improve the care of patients around the world through application of pharmacokinetic and pharmacodynamic principles.

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 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.

References

  • 1.O’Neill J Tackling drug-resistant infections globally: Final report and recommendations. London: The Review on Antimicrobial Resistance; 2016. Available from https://amr-review.org/sites/default/files/160525_Final%20paper_with%20cover.pdf Accessed 12 September, 2019. [Google Scholar]
  • 2.World Health Organization. Global action plan on antimicrobial resistance. Geneva: 2015. Available from https://apps.who.int/iris/bitstream/handle/10665/193736/9789241509763_eng.pdf?sequence=1. Accessed 12 September, 2019. [Google Scholar]
  • 3.Munoz-Price L, Poirel L, Bonomo RA, et al. Clinical epidemiology of the global expansion of Klebsiella pneumoniae carbapenemases. Lancet Infect Dis 13(9):785–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ramos-Castañeda JA, Ruano-Ravina A, Barbosa-Lorenzo R, et al. Mortality due to KPC carbapenemase-producing Klebsiella pneumoniae infections: Systematic review and meta-analysis. J Infect 2018;76:438–48. [DOI] [PubMed] [Google Scholar]
  • 5.Tzouvlekis LS, Markogiannakis A, Psichogiou M, Tassios PT, Daikos GL. Carbapenemases in Klebsiella pneumoniae and other Enterobacteriaceae: An evolving crisis of global dimensions. Clin Microbiol Rev 2012;25(4):682–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bassetti M, Giacobbe DR, Giamarellou H, et al. Management of KPC-producing Klebsiella pneumoniae infections. Clin Microbiol Infect 2018;24(2):133–44. [DOI] [PubMed] [Google Scholar]
  • 7.Carr A, Stringer J. Antibiotic R&D Update 20. Available from https://needham.bluematrix.com/sellside/EmailDocViewer?encrypt=e0df6a7d-6477-4c25-9d2f-09f3fb40d31a&mime=pdf. Accessed 12 September, 2019.
  • 8.Tsuji BT, Pogue JM, Zavascki AP, et al. International consensus guidelines for the optimal use of the polymyxins: Endorsed by the American College of Clinical Pharmacy (ACCP), European Society of Clinical Microbiology and Infectious Diseases (ESCMID), Infectious Diseases Society of America (IDSA), International Society for Antiinfective Pharmacology (ISAP), Society of Critical Care Medicine (SCCM), and Society of Infectious Diseases Pharmacists (SIDP). Pharmacotherapy 2019;39(1):10–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bulman ZP, Satlin MJ, Chen L, et al. New Polymyxin B dosing strategies to fortify old allies in the war against KPC-2-producing Klebsiella pneumoniae. Antimicrob Agents Chemother 2017;61(4):e02023–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Nielsen EI, Friberg LE. Pharmacokinetic-pharmacodynamic modeling of antibacterial drugs. Pharm Rev 2013;65:1053–90. [DOI] [PubMed] [Google Scholar]
  • 11.Tängden T, Ramos Martín V, Felton TW, et al. The role of infection models and PK/PD modelling for optimising care of critically ill patients with severe infections. Intensive Care Med 2017;43:1021–32. [DOI] [PubMed] [Google Scholar]
  • 12.Clinical and Laboratory Standards Institute. Performance standards for antimicrobial susceptibility testing: Fifteenth informational supplement M100-S15. CLSI, Wayne, PA, USA, 2005. [Google Scholar]
  • 13.Pournaras S, Kristo I, Vrioni G, et al. Characteristics of meropenem heteroresistance in Klebsiella pneumonia carbapenemase (KPC)-producing clinical isolates of K. pneumoniae. J Clin Microbiol 2009;48(7):2601–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.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(4):524–531. [DOI] [PubMed] [Google Scholar]
  • 15.Ehmann L, Zoller M, Minichmayr IK, Scharf C, Huisinga W, Zander J, Kloft C. Development of a dosing algorithm for meropenem in critically ill patients based on a population pharmacokinetic/pharmacodynamic analysis. Int J Antimicrob Agents. 2019;54(3):309–17. [DOI] [PubMed] [Google Scholar]
  • 16.Sanofi. Rifadin (R) rifampin [package insert]. Bridgewater, NJ: Sanofi; 2013. [Google Scholar]
  • 17.Bauer RJ. S-ADAPT/MCPEM User’s Guide. Berkeley, CA, USA: Biomedical Simulation Resource, 2011. [Google Scholar]
  • 18.Bulitta JB, Bingolbali A, Shin BS et al. Development of a new pre- and post-processing tool (SADAPT-TRAN) for nonlinearmixed-effects modeling in S-ADAPT. AAPS J 2011;13:201–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bulitta JB, Ly NS, Yang JC, Forrest A, Jusko WJ, Tsuji BT. 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] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bulitta JB, Yang JC, Yohonn L, Ly NS, Brown SV, D’Hondt RE, Jusko WJ, Forrest A, Tsuji BT. Attenuation of colistin bactericidal activity by high inoculum of Pseudomonas aeruginosa characterized by a new mechanism-based population pharmacodynamic model. Antimicrob Agents Chemother 2010;54:2051–2062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Landersdorfer CB, Ly NS Xu H, Tsuji BT, Bulitta JB. Quantifying Subpopulation Synergy for Antibiotic Combinations via Mechanism-Based Modeling and a Sequential Dosing Design. Antimicrob Agents Chemother 2013;57:2343–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Svensson RJ, Aartenoutse RE, Diacon AH, Dawson R, Gillespie SH, Boeree MJ, Simonsson USH. A Population pharmacokinetic model incorporating saturable pharmacokinetics and autoinduction for high rifampicin doses. Clin Pharmacol Ther 2018; 103(4):674–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Wang W, Hallow K, James D. A tutorial on RxODE: Simulating differential equation pharmacometric models in R. CPT Pharmacometrics Syst Pharmacol 2016;5(1):3–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lenhard JR, Bulitta JB, Connell TD, et al. High-intensity meropenem combinations with polymyxin B: new strategies to overcome carbapenem resistance in Acinetobacter baumannii. J Antimicrob Chemother 2017;72(1):153–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Smith NM, Bulman ZP, Sieron, et al. Pharmacodynamics of dose-escalated ‘front-loading’polymyxin B regimens against polymyxin-resistant mcr-1-harbouring Escherichia coli. J Antimicrob Chemother 2017;72(8):2297–2303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Diep JK, Jacobs DM, Sharma R, et al. Polymyxin B in combination with rifampin and meropenem against polymyxin B-resistant KPC-producing Klebsiella pneumoniae. Antimicrob Agents Chemother 2017;61(2):e02121–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Fisher RA, Gollan B, Helaine S. Persistent bacterial infections and persister cells. Nat Rev Microbiol 2017;15(8):453–64. [DOI] [PubMed] [Google Scholar]
  • 28.Cross T, Ransegnola B, Shin JH, et al. Spheroplast-mediated carbapenem tolerance in Gram-negative pathogens. Antimicrob Agents Chemother 2019;63(9):e00756–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Huttner A, Harbarth S, Hope WW, Lipman J, Roberts JA. Therapeutic drug monitoring of the β-lactam antibiotics: What is the evidence and which patients should we be using it for? J Antimicrob Chemother 2015;70(12):3178–83. [DOI] [PubMed] [Google Scholar]
  • 30.Alsultan A, Peloquin CA. Therapeutic drug monitoring in the treatment of tuberculosis: An update. Drugs 2014;74:839–54. [DOI] [PubMed] [Google Scholar]
  • 31.Logan LK, Weinstein RA. The epidemiology of carbapenem-resistant Enterobacteriaceae: The impact and evolution of a global menace. J Infect Dis 2017;215(Suppl 1):S28–S36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Svensson EM, Svensson RJ, te Brake LHM, et al. The potential for treatment shortening With higher rifampicin doses: Relating drug exposure to treatment response in patients with pulmonary tuberculosis. Clin Infect Dis 2018;67(1):34–41. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

RESOURCES