Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Jun 1.
Published in final edited form as: Lancet Infect Dis. 2014 Apr 24;14(6):498–509. doi: 10.1016/S1473-3099(14)70036-2

Challenges and Potential Solutions – Individualised Antibiotic Dosing at the Bedside for Critically Ill Patients: a structured review

Jason A Roberts 1,2, Mohd Hafiz Abdul Aziz 1, Jeffrey Lipman 1,2, Johan W Mouton 3, Alexander A Vinks 4, Timothy W Felton 5, William W Hope 6, Andras Farkas 7, Michael N Neely 8, Jerome J Schentag 9, George Drusano 10, Otto R Frey 11, Ursula Theuretzbacher 12, Joseph L Kuti 13, On behalf of The International Society of Anti-Infective Pharmacology (ISAP) and the PK/PD Study Group of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID)
PMCID: PMC4181663  NIHMSID: NIHMS600115  PMID: 24768475

Summary

Infections in critically ill patients are associated with persistently poor clinical outcomes. These patients have severely altered and variable antibiotic pharmacokinetics and are infected by less susceptible pathogens. Antibiotic dosing that does not account for these features is likely to result in sub-optimal outcomes. In this paper, we review the patient- and pathogen-related challenges that contribute to inadequate antibiotic dosing and discuss how a process for individualised antibiotic therapy, that increases the accuracy of dosing, can be implemented to further optimise care for the critically ill patient. The process for optimised antibiotic dosing firstly requires determination of the physiological derangements in the patient that can alter antibiotic concentrations including altered fluid status, microvascular failure, serum albumin concentrations as well as altered renal and hepatic function. Secondly, knowledge of the susceptibility of the infecting pathogen should be determined through liaison with the microbiology laboratory. The patient and pathogen challenges can then be solved by combining susceptibility data with measured antibiotic concentration data (where possible) into a clinical dosing software. Such software uses pharmacokinetic-pharmacodynamic (PK/PD) models from critically ill patients to accurately predict the dosing requirements for the individual patient with the aim of optimising antibiotic exposure and maximising effectiveness.

Keywords: Antibacterial, Bayesian, Intensive Care Unit, pharmacodynamics, pharmacokinetics

Introduction

Patients in intensive care units (ICU) are markedly different from those in general ward environments and have significantly higher mortality rates. These patients are mostly critically ill and have a much higher level of sickness severity that is associated with profound pathophysiological changes and aggressive medical intervention1, 2. Indeed, the healthcare system is treating increasing numbers of critically ill patients and the clinical outcome for many patient sub-groups is not improving significantly3. In particular, critically ill patients with sepsis, septic shock and/or acute kidney injury are considered to be significant challenge to infectious diseases physicians, critical care physicians, nephrologists and clinical pharmacists and pharmacologists.

In sepsis and septic shock studies, interventions relating to optimisation of antibiotic therapy demonstrate the greatest improvements in clinical outcomes4-9. There is much evidence defining the impact of early and appropriate antibiotic administration on decreased mortality6-8 but less information regarding the impact of appropriate dosage regimens on clinical outcome10. Although robust in vitro and animal in vivo data exist describing exposure-effect relationships of antibiotics and bacterial killing11, 12, the impact of antibiotic exposure on mortality has not been defined as precisely. Nevertheless, some important studies, mostly observational or retrospective in nature, are available.

For aminoglycosides, a randomised controlled trial by Van Lent Evers demonstrated that a dedicated therapeutic drug monitoring (TDM) intervention in a general hospitalised patient cohort resulted in a significantly reduced patient hospital length of stay13. Quinolones10, 14, 15, beta-lactams10, 16-19, glycopeptides20, 21 and linezolid22 all have at least retrospective cohort analyses that demonstrate clinical cure and/or mortality advantages associated with achievement of target pharmacokinetic (PK)/pharmacodynamic (PD) indices. The major challenge for clinicians remains how to ensure that dosing achieves these PK/PD targets in individual patients.

Guidance for effective antibiotic dosing in critically ill patients is usually not included in treatment guidelines. Clinically, use of the Product Information (or Package Insert) for the antibiotic is the mainstay for choosing antibiotic doses for critically ill patients. However, this information is based on dose-finding studies that are performed in non-critically ill patients and then extrapolated to critically ill patients. The presence of the severely altered PK means that many critically ill patients may be at risk for not achieving the PK/PD targets that are known to be associated with an improved likelihood of positive clinical outcomes23, 24. Even general dosing guidelines for ICU patients may not be a satisfactory solution because critically ill patients have significant PK variability. With increasing PK variability, the likelihood of accurately predicting a therapeutic dose in an individual patient decreases, potentially resulting in sub-optimal patient outcomes.

After many years of dosing antibiotics in critically ill patients using a ‘one dose fits all’ strategy, there is a strong rationale to move to an individualised dosing approach. This change in approach to dosing antibiotics is supported by the ubiquitous problem of reduced antibiotic development pipeline that requires better use of existing agents, for which antibiotic resistance is steadily emerging.

In this paper, we describe the challenges of altered PK caused by the pathophysiological changes that are commonly found in critically ill patients as well as the challenges of the reduced bacterial organism susceptibility frequently encountered in the ICU. Either PK or PD, or both, influence the PK/PD ratio and therefore, the magnitude of the PK/PD target. Therefore, we will consider solutions to the above challenges in the form of individualizing dosing strategies supported by different bedside dosing tools based on software packages that can improve the likelihood of effective dosing.

Search strategy and selection criteria

Data for this review were identified by searches of Pubmed (1966 to February 2014), EMBASE (1966 to July 2013) and the Cochrane Controlled Trial Register as well as references from relevant articles. Search terms related to antibiotic pharmacokinetics and pharmacodynamics in critically ill patients and dosing software were included. Numerous articles were identified through searches of the extensive files of the authors. All relevant papers available in English were reviewed for inclusion.

Challenge 1: The effect of critical illness pathophysiology on pharmacokinetics

Dysfunction of one or multiple organ systems occurs in critical illness and may result in significantly different antibiotic concentrations from those observed in noncritically ill patients as shown in Figure 1. Without rational dose adjustment, these changes in drug concentrations can predispose to clinical failure, emergence of antimicrobial resistance or even drug toxicity. Below we review the PK effects caused by dysfunction of the cardiovascular, renal, pulmonary and hepatic systems.

Figure 1.

Figure 1

The spectrum of critical illness-related altered pathophysiology and its effects on drug concentrations.

Legend: CL – clearance; Vd volume of distribution; RRT – renal replacement therapy; ECMO – extracorporeal membrane oxygenation

The cardiovascular system

Fluid shifts, ‘third spacing’ and fluid overload

Critically ill patients frequently demonstrate a systemic inflammatory response syndrome (SIRS) caused by either infectious or non-infectious pathology25. A major consequence of SIRS, particularly in patients with severe sepsis and septic shock, is the extreme fluid extravasation into the interstitial space from endothelial damage and capillary leakage. This phenomenon is commonly described as ‘third spacing’25. In response to the resulting hypotension, clinicians administer large volumes of resuscitation fluids that may also distribute into interstitial fluid thereby significantly increasing interstitial volume. For hydrophilic antibiotics, these processes may lead to a large increase in volume of distribution (Vd)26. In contrast, lipophilic antibiotics (e.g. fluoroquinolones and macrolides) possess an inherently larger Vd and their Vd is often not significantly influenced by such fluid movements and/or administration27.

The increase in Vd for hydrophilic antibiotics such as the aminoglycosides28, 29, beta-lactams26, 30, glycopeptides31 and linezolid32 have been extensively documented in critically ill patients. Increases in Vdthat are up to 2-fold greater than the Vdobserved in non-critically ill patients are common.

Hypoalbuminaemia and altered protein concentrations

Hypoalbuminaemia, defined as a serum albumin concentration < 25 g/L, is a common but frequently neglected condition in the ICU. The incidence is reportedly as high as 40-50%33. Ulldemolins et al. have extensively reviewed this phenomenon and concluded that the influence of hypoalbuminaemia on antibiotic PK in critically ill patients may be clinically important34. With decreasing albumin concentrations, an increase in the unbound fraction of protein bound drugs can occur35. The unbound fraction of such antibiotics is not only available for elimination, but also for distribution. The Vd for moderate to highly-protein bound antibiotics including ceftriaxone36, flucloxacillin37, ertapenem38, 39 and daptomycin40 are found to increase by up to 100% in critically ill patients with hypoalbuminaemia.

Evidently, fluid shiftsand altered protein binding, as commonly seen in mechanically ventilated patients, will both increase Vd. An increased Vd may reduce the peak concentration of drugs which may be problematic for antibiotics exhibit concentration-dependent effects (e.g. aminoglycosides). These drugs require a high unbound peak concentration (Cmax) over MIC (Cmax/MIC) and area-under the curve to MIC (AUC/MIC) ratio for maximal bacterial killing41-43. However, where the drug has high protein binding (e.g. daptomycin), hypoalbuminaemia will likely to lead to a high free fraction in the early part of the dosing interval which may result in higher unbound concentrations which may be advantageous. In contrast, for time-dependent beta-lactam antibiotics, changes in Vd and protein binding can lead to unbound concentrations later in the dosing interval are consistently lower than those observed in non-critically ill patients putting the patient at risk of treatment failure from subtherapeutic concentrations35, 36.

Importantly, increasing sickness severity is associated with increased Vd meaning that the most critically ill patients are likely to have the most reduced antibiotic exposures if standard dosing is used, at least in the first days of treatment29. With recovery from infection, the Vd will return to ‘normal’ and as such, in longer courses of therapy, dose modifications throughout treatment are often required. For all antibiotic classes including concentration-dependent antibiotics, an increased Vdmay delay the time to achievement of therapeutic concentrations.

Tissue perfusion and target site distributions of antibiotics

Effective antibiotic concentrations need to be achieved in the interstitial fluid of tissues as this is the site of most infections44. However, severe infections can cause vascular dysfunction including microvascular failure, which can impair the drug delivery into body tissues45. Data describing impaired tissue penetration exists for various antibiotics including cefpirome46, fosfomycin47 piperacillin45, 48and levofloxacin49 with sub-therapeuticconcentrations in tissue common in the early phase of treatment, particularly in patients with septic shock being treated with vasopressors45. Therefore, at least for the antibioticclasses above, plasma concentrations maybe an imprecise surrogate for tissue concentrations.

The renal system

Many of the commonly used antibiotics in critically ill patients are subject to renal clearance and therefore alterations in renal function will affect concentrations of those antibiotics. While it is standard practice to reduce an antibiotic dose in the presence of renal dysfunction, acute kidney injury (AKI), or renal replacement therapy (RRT) to avoid toxicity in the critically ill patient, it is less well appreciated that glomerular filtration can be increased in some patients as a result of a phenomenon known as augmented renal clearance (ARC). ARC is a potential reason for ‘under-dosing’. Thus some critically ill patients may actually require a more intensive regimen, which may be just as relevant as dosage reduction in the setting of renal impairment.

Augmented renal clearance (ARC)

ARC is driven by pathophysiological responses to infection as well as treatment interventions (e.g. fluid resuscitation and use of vasopressors) that are also associated with early increase in cardiac output and an associated enhanced blood flow to major organs50. For the kidney, this increased perfusion leads to increased drug delivery and therefore significantly increased glomerular filtration and clearance of renally cleared solutes including antibiotics (e.g. aminoglycosides, beta-lactams and glycopeptides)1, 51, 52. ARC is being increasingly observed in critically ill patients with normal serum creatinine concentration. ARC typically occurs in younger males with trauma, sepsis, burns, haematological malignancy or pancreatitis53. ARC is defined as a creatinine clearance (CrCL) ≥130 mL/min. Udy et al, demonstrated up to 82% of patients demonstrating ARC will not achieve therapeutic antibiotic concentrations with standard antibiotic doses54.

Renal dysfunction

A decline in kidney perfusion including microcirculatory failure can occur leading to AKI and reduced clearance of renally-eliminated antibiotics. AKI is identified by elevated serum creatinine concentrations or drop in urine output55 and when present will require appropriate dose decreases to ensure therapeutic and non-toxic antibiotic exposures. Importantly, large dose decreases are not required in the presence of AKI for antibiotics with a wide therapeutic index and where clearance occurs by multiple routes and the proportion of non-renal clearance is moderate to high (e.g. ceftriaxone, flucloxacillin and ciprofloxacin have both hepatic and renal clearance pathways).

Renal replacement therapy

If severe AKI occurs, RRT may be prescribed for clearance of metabolic waste products and/or fluid removal. RRT may take the form of continuous renal replacement therapy (CRRT) or intermittent haemodialysis (IHD) or a hybrid form of both such as sustained low-efficiency dialysis (SLED). CRRT is by far the most common form of RRT in critically ill patients, although hybrid forms of RRT are increasingly common. The principles of antibiotic dosing during RRT and factors that need to be considered have been discussed in significant detail previously56, 57. In general, drugs with a high Vd (> 1 L/kg), lipophilic drugs, and/or drugs with high protein binding (>80%) are poorly eliminated by RRT56.

Sepsis in the presence of RRT is associated with a 50% increased likelihood of mortality compared to RRT alone58. In part, this may be explained by the difficulties in antibiotic dosing in these patients. Because there is no standardised approach to delivering RRT, with the exception of IHD, antibiotic clearance can be highly variable across different RRT modalities and settings. Recent papers have highlighted the dosing challenges for vancomycin, ciprofloxacin and beta-lactams, where anywhere from 10-50% of critically ill patients did not achieve target antibiotic concentrations59, 60. Dosing of antibiotics during RRT should ideally be individualised to the patient and the RRT modality and settings prescribed.

The pulmonary system

Pneumonia, (especially complicating mechanical ventilation), is the most common infection in critically ill patients and is an important cause of morbidity and mortality in ICU patients61. Providing optimal antibiotic exposure for hospital acquired pneumonia in ventilated patients may be challenging especially when clinicians consider the various patient, pathology and drug factors that can each affect drug penetration to the site of infection62. Alveolar compartments such as epithelial lining fluid (ELF) are considered the closest measurable site where extracellular pathogens accumulate and thus, optimal antibiotic concentrations in this area may determine therapeutic success62. Following systemic drug administration, the antibiotic must cross the alveolar capillary barrier before reaching and exerting their activity within the ELF. The passage across this barrier may be influenced by physicochemical (e.g. lipophilicity) and PK (e.g. protein binding) characteristics of the antibiotic and as well as patient specific characteristics (e.g. inflammation and/or chronic lung disease). The degree of ELF penetration for an antibiotic is characterized by the ratio of ELF exposure to the corresponding plasma exposure.

In the context of antibiotic physicochemistry,the more lipophilic antibiotics (e.g. fluoroquinolones, macrolides and oxazolidinones) have demonstrate an ELF: plasma exposure ratio of ≥163, 64. Such high ELF:plasma exposure ratios are not always observed for hydrophilic antibiotics although ratios may be underestimated due to technical errors63, 65, 66. In general, while blood concentrations may appear “therapeutic”, ELF concentrations may be insufficient for optimal antibiotic treatment, especially in case of reduced bacterial susceptibility.For this reason, some authors suggest higher doses should be used in patients with severe nosocomial pneumonia to optimize ELF concentrations, for hydrophilic drugs67, 68. Alternatively, use of different administration approaches such as extended or continuous infusion of beta-lactam antibiotics63, 65, or administration via nebulisation69, may be used to increase antibiotic concentrations in ELF.

The hepatic system

During severe sepsis and septic shock, hepatic dysfunction may also cause a decrease in drug metabolism and clearance70, 71. To date, few data are available to guide antibiotic dose adjustments in critically ill patients with liver dysfunction72, regardless of critical illness.

Challenge 2: Increased incidence of reduced bacterial susceptibility

The minimum inhibitory concentration (MIC) of the antibiotic against the pathogen causing the infection is a critical consideration for antibiotic dosing. The MIC is a critical factor of the PK/PD relationship that defines the drug exposure necessary to ensure a patient achieves a predefined PK/PD target that is associated with maximal efficacy.

Infections in the ICU are often caused by pathogens with higher MICs compared with other clinical settings73, 74. For example, a comparison of carbapenem susceptibility against predominantly Gram negative isolates in Germany, doripenem, meropenem and imipenem demonstrated a MIC90in critically ill patients that was 4-, 8- and 8-fold greater than in non-critically ill patients, respectively75. As MICs increase, thePK exposure that is required to achieve the PK/PD threshold must increase in a proportional manner.To numerically highlight the difference in PK exposure required for an antibiotic in the presence of an elevated MIC, we can consider one example where vancomycin is being administered for a healthcare associated pneumonia for which, a PK/PD target AUC0-24/MIC of 400, may be used76. In this case, if the methicillin resistant Staphylococcus aureus(MRSA) pathogen has an MIC of 0.5 mg/L, then an AUC0-24 of 200 mg.h/L is required, which would be achieved comfortably with a trough concentration exceeding 10 mg/L. However, if the MIC is 2 mg/L, then an AUC0-24 of 800 mg.h/L is required, thus necessitating a target trough concentration > 20-25 mg/L which would dramatically increase the risk of drug-related toxicity. In the latter case, consideration of an alternative antibiotic or combination therapy may be required.

Using the above example as an indicator of the challenges of achieving optimal dosing in the presence of decreased antibiotic susceptibility, it is clear that quantitative knowledge of antibiotic susceptibility will help guide dosing needs in critically ill patients. With a continuing decrease in susceptibility to the commonly used antibiotics in critically ill patients, regular surveillance is required77. These surveillance programs should also report MICs from ICU and non-ICU wards means as separate reports because differences in antibiotic susceptibility are common73, 74, 78, 79. These issues highlight the importance of having numerical data on the MIC of the pathogen causing the infection. At present, most laboratories routinely report susceptibility with the classification ‘S, I and R’ (Susceptible, Intermediate-Susceptible and Resistant) based on MIC breakpoints. While this approach is suitable for many clinical situations because it unambiguously delineates when an antibiotic should not be administered, it may not be suitable for a critically ill patient with altered PK and antibiotic susceptibility close to the breakpoint. In this case, the relevant PK/PD target may still not be achieved despite the organisms have an MIC classified as “S”80. Therefore, knowledge of MIC data of the pathogen in the individual patient is essential to accurately calculate the PK exposure that patient needs such that the PK/PD target can be achieved. The local practitioner aiming to apply the dosage individualization concepts outlined in this paper should make it a priority to engage their microbiology lab to ensure that methods to obtain MIC data are in place.

Another important issue for treatment of infections in the critically ill is that MIC breakpoints published by groups such as the European Committee on Antimicrobial Susceptibility and Testing (EUCAST; available at www.eucast.org) and the Clinical and Laboratory Standards Institute (CLSI; available at www.clsi.org) are frequently determined using antibiotic exposures from non-critically ill patients. It follows that if an individual patient has profoundly altered PK and is infected by a pathogen with an MIC at or near the breakpoint, then use of a standard fixed regimen may increase the probability of under dosing (potential reasons for this PK variability are discussed above).

Given such inherent challenges relating to pathophysiology, PK and decreased bacterial susceptibility, what can be done to increase the likelihood of positive treatment outcomes for critically ill patients?

Solution: Individualised antibiotic dosing

Optimal patient outcomes from treatment of infection are most likely to occur when PK/PD targets that are associated with maximal antibiotic activity are achieved. The advent of in vitro and in vivomathematical PK/PD models over the recent 30 years has allowed accurate description of the PK/PD targets that are associated with maximal antibiotic effect. More recent clinical analyses have attempted to confirm the results of these studies and have, for the most part, described similar PK/PD targets to those observed in the pre-clinical studies81. Table 1 describes various PK/PD targets that have been described in pre-clinical and clinical studies which could be considered as therapeutic targets to optimise dosing in individual patients.

Table 1.

Studies reporting PK/PD indices from pre-clinical and clinical evaluations

Antibiotic
classifications
Optimal PK/PD indices*
Pre-clinical studies Clinical studies
Concentration-
dependent
Aminoglycosides Maximal killing AUC0-24/MIC 80-100 43 Clinical Cure Cmax/MIC 8-10
AUC/MIC >70
82-86
Resistance suppression Cmax/MIC 10-30 87 Microbiological Cure -
Time-dependent Carbapenems Maximal killing 40% T>MIC 88 Clinical Cure 75% T>MIC
Cmin/MIC 5
89
Resistance suppression 16 × MIC
Cmin/MIC >6.2
90
91
Microbiological Cure 54% T>MIC 17
Cephalosporins Maximal killing 60-70% T>MIC 11 Clinical Cure 100% T>MIC 92
Resistance suppression - Microbiological Cure 60-100% T>MIC
95% T>4.3×MIC
16
93
Penicillins Maximal killing 40-50% T>MIC 11 Clinical Cure -
Resistance suppression 40-50% T>MIC 94 Microbiological Cure 40-50% T>MIC 95
Concentration
with time-
dependence
Fluoroquinolones Maximal killing AUC0-24/MIC >30-100 11, 96 Clinical Cure AUC0-24/MIC ≥125-250
Cmax/MIC ≥8
15, 86, 96
86,97, 98
Resistance suppression AUC0-24/MIC >160
AUC0-24/MPC ≥22
99, 100
101
Microbiological Cure AUC0-24/MIC ≥34-125
Cmax/MIC ≥8
14, 102
86
Vancomycin Maximal killing AUC0-24/MIC 86-460 103 Clinical Cure AUC0-24/MIC ≥400-450 20, 21
Resistance suppression AUC0-24/MIC >200 104 Microbiological Cure AUC0-24/MIC ≥400 20
Linezolid Maximal killing - Clinical Cure AUC0-24/MIC ≥85
85% T>MIC
22
Resistance suppression - Microbiological Cure AUC0-24/MIC 80-120
85% T>MIC
22
Tigecycline Maximal killing 50% T>MIC 105 Clinical Cure AUC0-24/MIC >12.8-17.9
f AUC0-24/MIC ≥0.9
106, 107
108
Resistance suppression - Microbiological Cure AUC0-24/MIC 6.9-17.9 109, 110
Daptomycin Maximal killing AUC0-24/MIC 38-442 111, 112 Clinical Cure -
Resistance suppression AUC0-24/MIC >200 104 Microbiological Cure -
Colistin Maximal killing AUC0-24/MIC 7-23 113, 114 Clinical Cure -
Resistance suppression - Microbiological Cure -
*

Where the index is reported as a range, the data included may have been derived from different infection models with different bacteria. Specific data about the contributing values can be found in the associated references.

**

Data for the various indices has been reported in different studies according to total and free (unbound) drug. Where free concentrations have been reported, this is denoted using ‘f

Legend: MIC – minimum inhibitory concentration; AUC0-24/MIC – ratio of area under the concentration time curve from 0-24 hours to MIC; Cmax/MIC – ratio of maximum concentration of antibiotic in a dosing interval to MIC; T>MIC – percentage of dosing interval that the antibiotic concentration is maintained above the MIC; AUC0-24/MPC; ratio of the AUC0-24 to mutant prevention concentration; Cmin – minimum concentration of antibiotic in a dosing interval; f – free or fraction of drug not bound to plasma proteins

To increase the likelihood of achieving therapeutic targets for systemically administered antibiotics, there are two main approaches to adjusting standard regimens: 1) altered administration techniques such as once-daily dosing or prolonged infusion which is often based on published studies of the specific dosing regimen and/or 2) dose adjustment that is guided by therapeutic drug monitoring (TDM).

Altered administration techniques

There are numerous PK studies that apply dosing simulations to identify optimised regimens to achieve PK/PD targets for infections caused by organisms with higher MICs or in the setting of altered PK. Such an approach to dosing is not an individualised approach per se when it is employed on a population level, but is a form of therapeutic adaptation designed to enhance improve antibiotic efficacy. PK/PD-based dosing has changed the way aminoglycosides are prescribed clinically from thrice to once daily dosing and has improved the safety and efficacy of these compounds115. As such, extended interval dosing for aminoglycosides is widely considered the standard of care116.

Studies for beta-lactams are particularly numerous. Collectively, these studies suggest that prolongation of the duration of infusion (either for 40-50% of the dosing interval (i.e., 3-4 hours), or as a continuous infusion) achieves a greater likelihood of achieving PK/PD targets than standard bolus dosing in critically ill patients68, 117-124. Recent studies have sought to investigate the clinical value of use of these prolonged infusions with prospective randomised controlled trials suggesting a potential advantage of continuous infusions for critically ill patients with severe sepsis125, 126. While some meta-analyses of these studies have not been able to quantify definitive advantages for either intermittent or prolonged infusions of beta-lactams, these studies have often not been stratified for patients with altered PK or reduced susceptibility126, 127. For example, in one study, changing the approach to dosing beta-lactams at an ICU-wide level from intermittent to extended infusion was not found to be clinically advantageous – this was likely because of a high proportion of susceptible pathogens in that ICU. These low MICs meant that standard infusions had already obtained requisite PK/PD thresholds in the vast majority of patients128. A clinician must determine whether the patient and/or pathogen is at risk of failing a standard fixed regimen. Changing the approach to dosing for all patients may not be necessary and not confer any therapeutic advantage. This study from Arnold et al128, adds support to the important findings from Lodise et al. of the clinical effectiveness of administering piperacillin/tazobactam as an extended infusion for Pseudomonas aeruginosa infections in patients with higher levels of sickness severity129.

For vancomycin, a few studies have compared continuous infusion versus intermittent dosing and have largely demonstrated equivalence130. Only one study, by Rello et al, has shown potential clinical outcome advantages for continuous infusion131. The value of continuous infusion for vancomycin is likely related to more consistent achievement of PK/PD targets31. The influence on emergence of resistance has not been addressed in these studies.

In general, the above concepts can be implemented at the ICU level when MIC data are available to justify a change at the ICU level empirically.

Therapeutic Drug Monitoring

TDM (also described as therapeutic drug management) is traditionally used to minimise toxicities for drugs, but in critically ill patients, is being increasingly used to optimise dosing in the presence of severely deranged PK10, 132. TDM relies on direct measurement of serum antibiotic concentrations with timely feedback to the clinician who would then interpret that result in the context of a therapeutic range. The adequacy of the measured concentration can be interpreted by directly comparing a single concentration value to a therapeutic target, or by describing antibiotic exposure using non-linear regression or Bayesian techniques. Doses can then be increased or decreased as predicted by the clinician or the dosing software (therapeutic drug management).

Notably, an understanding of the importance of the unbound concentration of the assayed drug in the blood sample is vital for accuracy when interpreting drug exposure because only free drug concentrations are microbiologically active. Knowledge of free concentrations is most important for highly bound antibiotics133. The concentration result from the assay should also be made available in a timely manner so that rapid dose adjustment can be made. Given the dynamic nature of PK in these patients, delays that are too long, can result in inappropriate dose adjustment. Importantly, for ideal TDM, the antibiotic MIC of the organism and TDM target should also be available (i.e., Challenge 2).

To date, various studies reporting TDM for antibiotics in critically ill patients have been reported including aminoglycosides134, glycopeptides135, beta-lactams10, 19, 136, linezolid137and quinolones10. Unfortunately, few of these studies have sought to measure clinical outcome advantages of using TDM compared with not applying TDM and a rigorous prospective evaluation of the benefit of individualised therapy should be considered essential.

Dosing nomograms

Dosing nomograms for dose adjustment of antibiotics are in common usage across many clinical areas138. These nomograms function by comparing a measured concentration value for the prescribed antibiotic at the time point that the sample was taken with a graph that defines the therapeutic range of concentrations at that stated time point. The dose of antibiotic can then be increased or decreased as necessary to ensure the next measured concentration is in the therapeutic range. These nomograms are simple to apply even for those without an advanced understanding of PK/PD and remain popular, with nomograms for vancomycin and aminoglycosides most widely available138-140 simply because these agents have toxicity thresholds (i.e., low therapeutic index), have drug assays available and are often targets for TDM by pharmacists and physicians. A limitation of many nomograms, however, is that they are rarely designed with PK/PD targets from critically-ill patients and rely on the clinician’s experience to make appropriate dose adjustments141.

Non-linear regression based dose adaptation

Application of non-linear regression analysis to a series of concentration values at different time points can be useful to calculate basic PK parameters such as area under the curve (AUC), clearance, elimination rate constant, maximum concentration in the dosing interval (Cmax) and trough concentration (Cmin). From a drug dosing perspective, the measured or calculated values for AUC, Cmax and/or Cmin can be compared against the PK/PD targets for the prescribed antibiotic and the dose empirically increased or decreased as needed.

Bayesian dose estimation and adaptation

Population PK models for antibiotics in critically ill patients have been developed for many antibiotics. Problematic to many of these models are that although there is increased PK variability in these patients, the sample size used for many of these studies remains small (~10-20 patients) and so not all of the PK variability for the population is likely to be captured by these models142, 143. Nevertheless, applying these population models is likely to be more accurate than using a model derived from another patient group144. The process for how dose individualisation could occur in a critically ill patient is described in the section, The process of how dose individualisation could occur for a patient.

To ensure greatest accuracy of dose adaptation based on drug concentration data, use of a stochastic control approach can be applied to define the timing and number of drug concentrations that should be taken from the patient and then used in the dose prediction145. This approach is particularly useful for drugs with high PK variability72. With more antibiotic concentration data over different dosing intervals, the accuracy of dose prediction improves72.

How are patient-specific PK parameters calculated?

In Bayesian dose adaptation, the dose of the drug is adjusted to ensure the individual patient’s exposure meets PK/PD targets. Information about the specific patient, in the form of serum drug concentrations, and a population PK model, from the relevant population, are included. This population PK model contains a series of mathematical equations including parameter estimates and their distribution for clearance and Vd.

The process of how dose individualisation could occur for a patient

A critically ill patient would receive a first dose at the clinician’s discretion, preferably using a strategy associated with an increased likelihood of achieving PK/PD targets, e.g. vancomycin loading dose31 or extended infusion beta-lactam68, 117-124. During the first or a subsequent dosage interval, one or more blood sample/s could be taken to estimate the patient’s individual PK parameters for the antibiotic. The samples would then be assayed in a timely manner (e.g., within 6 hours) and the dosing history, drug concentrations and necessary patient data (e.g., weight or creatinine clearance) would be provided to the chosen software package. Meanwhile, a pathogen MIC is derived for the antibiotic by the microbiology lab. The software package could then combine the patient’s observed data plus the population PK model to estimate the Bayesian posterior PK parameter values for the individual. The appropriate dose that achieves the PK/PD targets required for a critically ill patient could then be calculated and used in next dosing interval.

What antibiotic dosing software is available?

Numerous programs exist which apply different approaches to calculating individualised antibiotic doses for patients. A summary of various programs has been provided in Table 2, with other programs also identified by Fuchs et al146. Importantly, not all programs contain all relevant antibiotics, although the developers of most programs state that additional PK models for antibiotics can be included in these programs on request or by the user. To ensure robust bedside antibiotic dosing is possible, many of these programs have, or are developing electronic medical record interfaces and smart phone applications which can be literally used at the patient’s bedside.

Table 2.

Characteristics of various antibiotic dosing programs

BestDose v1.0 ID-ODS MWPharm DoseMe TCI Works First-dose WinAUIC CADDy
Program v4.e
PK Method Bayesian non-
parametric
approach
Bayesian
parametric
approach
Bayesian
parametric
approach
Bayesian
parametric
approach
Bayesian
parametric
approach
Population PK
parametric
approach
Non-linear
regression
Non-linear
regression
Adaptive feedback? Yes Yes Yes Yes Yes No No No
Web, server or
terminal based?
Terminal Terminal and
Server
Terminal and
Server
Terminal and
Server
Terminal and
Server
Web Terminal Web or Server
Compatibility Windows Mac, Windows,
Linux, Android,
IOS
Windows Mac, Windows,
Linux, Android,
IOS
Mac, Windows Mac, Windows,
Linux, Android,
IOS
Windows Mac, Windows,
Linux, Android,
IOS
Smart phone
application?
No Yes No Yes No Yes No No
Patient covariates in
dose predictions?
Yes Yes Yes Yes Yes Yes Yes Yes
Output from
program?
Doses and PK
parameter
estimates
Dosing regimens,
PK parameter
estimates and
PTAs
Doses and PK
parameter
estimates
Doses and PK
parameter
estimates
Doses and PK
parameter
estimates
Doses PK parameter
estimates
Doses
IT support available
within 24 hours? *
Yes Yes No Yes Yes No No Yes
Capacity for ICU
and non-ICU
dosing?
Yes Yes Yes Yes Yes No No No
Further information http://www.lapk.org http://www.optimum-dosingstrategies.org http://www.mediware.cz/index_en.html www.doseme.com.au www.tciworks.info http://www.firstdose.org/ Contact the
developers (Dr J
Schentag)
www.jjpreisenberger.de
Cost Current
version is free.
Free 1250 euros per
license
Dependent on
requirements
Free Free Free Free
*

Response time often depends on the severity of the problem for the user (for non-urgent issues, responses may exceed 24 hours)

Legend – ID-ODS: Individually Designed Optimum Dosing Strategies; WinAUIC: Windows Antibiotic Utilisation Information and Consultation; PK: pharmacokinetic; RRT: renal replacement therapy; AUIC: area under the inhibitory curve; MIC: minimum inhibitory concentration; ICU: intensive care unit; IT: information technology; PTA: probability of target attainment

What might a future scenario for antibiotic dosing in a critically ill patient look like?

Based on best available evidence, we would suggest that a robust process for dose individualisation in a critically ill patient would be as below. Note that part of this process would require baseline data of local bacterial susceptibility in the clinician’s ICU.

  1. Diagnosis of infection and selection of antibiotic is made.

  2. The patient’s physiological characteristics would be determined such that data of relevant covariate descriptors of the prescribed drug are known (e.g. weight, sex, creatinine clearance, serum albumin concentration, fluid overload status, presence of extracorporeal circuits).

  3. First dose estimated based on patient characteristics and local susceptibility data preferably using dosing decision support software.

  4. Dose is administered to the patient in a timely manner after diagnosis

  5. Blood samples are taken at pre-determined time points and are assayed within a timely period.

  6. The clinician can then enter the concentration-time data from the blood samples and the patient covariate data into the software which is in a smartphone or computer terminal at the patient’s bedside. The output from the software is a personalised dosing regimen for the patient that can achieve an evidence-based PK/PD target. When pathogen specific susceptibility data is available, this should be incorporated into the dose estimation process.

Conclusions

Critically ill patients have dramatically varied PK compared with non-critically ill patients and are more likely to be infected by less susceptible bacteria. Traditional antibiotic dosing strategies are unlikely to consistently achieve PK/PD targets associated with maximal antibiotic activity thereby putting the patient at risk of clinical failure, the development of resistance, or both. Optimization of antibiotic dosing in the ICU, therefore, requires an individualized approach for the patient that considers the MIC of the infecting pathogen and selects a dosing regimen that provides a high likelihood of obtaining the requisite PK/PD index predictive of success. The challenges are clearly here, but so too are the solutions. Pro-active therapeutic management for antibiotics other than vancomycin and aminoglycosides is possible, but needs to be escalated to the next level and made available to all hospitals. The ability to acquire individualized antibiotic concentrations, combined with access to available software programs as described here will increase dosing accuracy and the likelihood of achieving PK/PD targets in our patients.

Acknowledgements

We wish to acknowledge Judit Preisenberger, Prof Alexander Brinkman, Dr Robert McLeay, Prof Nick Holford and Dr Sam Holford for providing information about specific dosing software programs.

Jason Roberts is funded by a Career Development Fellowship from the National Health and Medical Research Council of Australia (APP1048652).

Tim Felton is a MRC Clinical Training Fellow supported by the North West England Medical Research Council Fellowship Scheme in Clinical Pharmacology and Therapeutics, which is funded by the Medical Research Council (grant number G1000417/94909), ICON, GlaxoSmithKline, AstraZeneca and the Medical Evaluation Unit.

William Hope is supported by a National Institute of Health (NIHR) Clinician Scientist Fellowship.

Financial support: Nil

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.

Potential conflicts of Interest: Dr Farkas is involved in the development of the software Individually Designed Optimum Dosing Strategies(ID-ODS); A/Prof Neely is involved in the development of the software BestDose; Prof Schentag is involved in the development of the software WinAUIC; Dr Frey is involved in the development of the software CADDy Program. We do not intentionally provide recommendations about preferred software in this paper.

References

  • 1.Lipman J, Udy AA, Roberts JA. Do we understand the impact of altered physiology, consequent interventions and resultant clinical scenarios in the intensive care unit? The antibiotic story. Anaesth Intensive Care. 2011;39:999–1000. doi: 10.1177/0310057X1103900602. [DOI] [PubMed] [Google Scholar]
  • 2.Roberts JA, Lipman J. Pharmacokinetic issues for antibiotics in the critically ill patient. Crit Care Med. 2009;37(3):840–51. doi: 10.1097/CCM.0b013e3181961bff. [DOI] [PubMed] [Google Scholar]
  • 3.Dombrovskiy VY, Martin AA, Sunderram J, Paz HL. Rapid increase in hospitalization and mortality rates for severe sepsis in the United States: a trend analysis from 1993 to 2003. Crit Care Med. 2007;35(5):1244–50. doi: 10.1097/01.CCM.0000261890.41311.E9. [DOI] [PubMed] [Google Scholar]
  • 4.Bellomo R, Cass A, Cole L, et al. Intensity of continuous renal-replacement therapy in critically ill patients. N Engl J Med. 2009;361(17):1627–38. doi: 10.1056/NEJMoa0902413. [DOI] [PubMed] [Google Scholar]
  • 5.Finfer S, Chittock DR, Su SY, et al. Intensive versus conventional glucose control in critically ill patients. N Engl J Med. 2009;360(13):1283–97. doi: 10.1056/NEJMoa0810625. [DOI] [PubMed] [Google Scholar]
  • 6.Kollef MH, Sherman G, Ward S, Fraser VJ. Inadequate antimicrobial treatment of infections: a risk factor for hospital mortality among critically ill patients. Chest. 1999;115(2):462–74. doi: 10.1378/chest.115.2.462. [DOI] [PubMed] [Google Scholar]
  • 7.Kumar A, Roberts D, Wood KE, et al. Duration of hypotension before initiation of effective antimicrobial therapy is the critical determinant of survival in human septic shock. Crit Care Med. 2006;34(6):1589–96. doi: 10.1097/01.CCM.0000217961.75225.E9. [DOI] [PubMed] [Google Scholar]
  • 8.MacArthur RD, Miller M, Albertson T, et al. Adequacy of early empiric antibiotic treatment and survival in severe sepsis: experience from the MONARCS trial. Clin Infect Dis. 2004;38(2):284–8. doi: 10.1086/379825. [DOI] [PubMed] [Google Scholar]
  • 9.Ranieri VM, Thompson BT, Barie PS, et al. Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med. 2012;366(22):2055–64. doi: 10.1056/NEJMoa1202290. [DOI] [PubMed] [Google Scholar]
  • 10.Scaglione F, Esposito S, Leone S, et al. Feedback dose alteration significantly affects probability of pathogen eradication in nosocomial pneumonia. Eur Respir J. 2009;34(2):394–400. doi: 10.1183/09031936.00149508. [DOI] [PubMed] [Google Scholar]
  • 11.Craig WA. Pharmacokinetic/pharmacodynamic parameters: rationale for antibacterial dosing of mice and men. Clin Infect Dis. 1998;26(1):1–10. doi: 10.1086/516284. quiz 1-2. [DOI] [PubMed] [Google Scholar]
  • 12.Drusano GL. Antimicrobial pharmacodynamics: critical interactions of ‘bug and drug’. Nat Rev Microbiol. 2004;2(4):289–300. doi: 10.1038/nrmicro862. [DOI] [PubMed] [Google Scholar]
  • 13.van Lent-Evers NA, Mathot RA, Geus WP, van Hout BA, Vinks AA. Impact of goal-oriented and model-based clinical pharmacokinetic dosing of aminoglycosides on clinical outcome: a cost-effectiveness analysis. Ther Drug Monit. 1999;21(1):63–73. doi: 10.1097/00007691-199902000-00010. [DOI] [PubMed] [Google Scholar]
  • 14.Ambrose PG, Grasela DM, Grasela TH, Passarell J, Mayer HB, Pierce PF. Pharmacodynamics of fluoroquinolones against Streptococcus pneumoniae in patients with community-acquired respiratory tract infections. Antimicrob Agents Chemother. 2001;45(10):2793–7. doi: 10.1128/AAC.45.10.2793-2797.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Forrest A, Nix DE, Ballow CH, Goss TF, Birmingham MC, Schentag JJ. Pharmacodynamics of intravenous ciprofloxacin in seriously ill patients. Antimicrob Agents Chemother. 1993;37(5):1073–81. doi: 10.1128/aac.37.5.1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Crandon JL, Bulik CC, Kuti JL, Nicolau DP. Clinical pharmacodynamics of cefepime in patients infected with Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2010;54(3):1111–6. doi: 10.1128/AAC.01183-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Li C, Du X, Kuti JL, Nicolau DP. Clinical pharmacodynamics of meropenem in patients with lower respiratory tract infections. Antimicrob Agents Chemother. 2007;51(5):1725–30. doi: 10.1128/AAC.00294-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Muller AE, Punt N, Mouton JW. Optimal exposures of ceftazidime predict the probability of microbiological and clinical outcome in the treatment of nosocomial pneumonia. J Antimicrob Chemother. 2013;68(4):900–6. doi: 10.1093/jac/dks468. [DOI] [PubMed] [Google Scholar]
  • 19.Roberts JA, Ulldemolins M, Roberts MS, et al. Therapeutic drug monitoring of beta-lactams in critically ill patients: proof of concept. Int J Antimicrob Agents. 2010;36(4):332–9. doi: 10.1016/j.ijantimicag.2010.06.008. [DOI] [PubMed] [Google Scholar]
  • 20.Moise-Broder PA, Forrest A, Birmingham MC, Schentag JJ. Pharmacodynamics of vancomycin and other antimicrobials in patients with Staphylococcus aureus lower respiratory tract infections. Clin Pharmacokinet. 2004;43(13):925–42. doi: 10.2165/00003088-200443130-00005. [DOI] [PubMed] [Google Scholar]
  • 21.Zelenitsky S, Rubinstein E, Ariano R, et al. Vancomycin pharmacodynamics and survival in patients with methicillin-resistant Staphylococcus aureus-associated septic shock. Int J Antimicrob Agents. 2013;41(3):255–60. doi: 10.1016/j.ijantimicag.2012.10.015. [DOI] [PubMed] [Google Scholar]
  • 22.Rayner CR, Forrest A, Meagher AK, Birmingham MC, Schentag JJ. Clinical pharmacodynamics of linezolid in seriously ill patients treated in a compassionate use programme. Clin Pharmacokinet. 2003;42(15):1411–23. doi: 10.2165/00003088-200342150-00007. [DOI] [PubMed] [Google Scholar]
  • 23.Roberts JA, De Waele JJ, Dimopoulos G, et al. DALI: Defining Antibiotic Levels in Intensive care unit patients: a multi-centre point of prevalence study to determine whether contemporary antibiotic dosing for critically ill patients is therapeutic. BMC infectious diseases. 2012;12:152. doi: 10.1186/1471-2334-12-152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Roberts JA, Paul SK, Akova M, et al. DALI: Defining Antibiotic Levels in Intensive care unit patients: Are current beta-lactam antibiotic doses sufficient for critically ill patients? Clin Infect Dis. 2013 doi: 10.1093/cid/ciu027. in press (Pubmed ID 24429437) [DOI] [PubMed] [Google Scholar]
  • 25.van der Poll T. Immunotherapy of sepsis. Lancet Infect Dis. 2001;1:165–74. doi: 10.1016/S1473-3099(01)00093-7. [DOI] [PubMed] [Google Scholar]
  • 26.Goncalves-Pereira J, Povoa P. Antibiotics in critically ill patients: a systematic review of the pharmacokinetics of beta-lactams. Crit Care. 2011;15(5):R206. doi: 10.1186/cc10441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Gous A, Lipman J, Scribante J, et al. Fluid shifts have no influence on ciprofloxacin pharmacokinetics in intensive care patients with intra-abdominal sepsis. Int J Antimicrob Agents. 2005;26(1):50–5. doi: 10.1016/j.ijantimicag.2005.04.005. [DOI] [PubMed] [Google Scholar]
  • 28.Conil JM, Georges B, Breden A, et al. Increased amikacin dosage requirements in burn patients receiving a once-daily regimen. Int J Antimicrob Agents. 2006;28(3):226–30. doi: 10.1016/j.ijantimicag.2006.04.015. [DOI] [PubMed] [Google Scholar]
  • 29.Marik PE. Aminoglycoside volume of distribution and illness severity in critically ill septic patients. Anaesth Intensive Care. 1993;21(2):172–3. doi: 10.1177/0310057X9302100206. [DOI] [PubMed] [Google Scholar]
  • 30.Sime FB, Roberts MS, Peake SL, Lipman J, Roberts JA. Does Beta-lactam Pharmacokinetic Variability in Critically Ill Patients Justify Therapeutic Drug Monitoring? A Systematic Review. AnnIntens Care. 2012;2(1):35. doi: 10.1186/2110-5820-2-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Roberts JA, Taccone FS, Udy AA, Jacobs F, Louis J-L, Lipman J. Vancomycin dosing in critically ill patients – robust methods for improved continuous infusion regimens. Antimicrob Agents Chemother. 2011;55(6):2704–9. doi: 10.1128/AAC.01708-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Buerger C, Plock N, Dehghanyar P, Joukhadar C, Kloft C. Pharmacokinetics of unbound linezolid in plasma and tissue interstitium of critically ill patients after multiple dosing using microdialysis. Antimicrob Agents Chemother. 2006;50(7):2455–63. doi: 10.1128/AAC.01468-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Finfer S, Bellomo R, Boyce N, French J, Myburgh J, Norton R. A comparison of albumin and saline for fluid resuscitation in the intensive care unit. New Eng J Med. 2004;350(22):2247–56. doi: 10.1056/NEJMoa040232. [DOI] [PubMed] [Google Scholar]
  • 34.Ulldemolins M, Roberts JA, Rello J, Paterson DL, Lipman J. The effects of hypoalbuminaemia on optimising antibiotic dosing in critically ill patients. Clin Pharmacokinet. 2011;50(2):1–12. doi: 10.2165/11539220-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 35.Roberts JA, Pea F, Lipman J. The clinical relevance of plasma protein binding changes. Clin Pharmacokinet. 2013;52(1):1–8. doi: 10.1007/s40262-012-0018-5. [DOI] [PubMed] [Google Scholar]
  • 36.Joynt GM, Lipman J, Gomersall CD, Young RJ, Wong EL, Gin T. The pharmacokinetics of once-daily dosing of ceftriaxone in critically ill patients. J Antimicrob Chemother. 2001;47(4):421–9. doi: 10.1093/jac/47.4.421. [DOI] [PubMed] [Google Scholar]
  • 37.Ulldemolins M, Roberts JA, Wallis SC, Rello J, Lipman J. Flucloxacillin dosing in critically ill patients with hypoalbuminaemia: special emphasis on unbound pharmacokinetics. J Antimicrob Chemother. 2010;65(8):1771–8. doi: 10.1093/jac/dkq184. [DOI] [PubMed] [Google Scholar]
  • 38.Brink AJ, Richards GA, Schillack V, Kiem S, Schentag J. Pharmacokinetics of once-daily dosing of ertapenem in critically ill patients with severe sepsis. Int J Antimicrob Agents. 2009;33(5):432–6. doi: 10.1016/j.ijantimicag.2008.10.005. [DOI] [PubMed] [Google Scholar]
  • 39.Burkhardt O, Kumar V, Katterwe D, et al. Ertapenem in critically ill patients with early-onset ventilator-associated pneumonia: pharmacokinetics with special consideration of free-drug concentration. J Antimicrob Chemother. 2007;59(2):277–84. doi: 10.1093/jac/dkl485. [DOI] [PubMed] [Google Scholar]
  • 40.Dvorchik B, Arbeit RD, Chung J, Liu S, Knebel W, Kastrissios H. Population pharmacokinetics of daptomycin. Antimicrob Agents Chemother. 2004;48(8):2799–807. doi: 10.1128/AAC.48.8.2799-2807.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Mouton JW, Jacobs N, Tiddens H, Horrevorts AM. Pharmacodynamics of tobramycin in patients with cystic fibrosis. Diagn Microbiol Infect Dis. 2005;52(2):123–7. doi: 10.1016/j.diagmicrobio.2005.02.011. [DOI] [PubMed] [Google Scholar]
  • 42.Mouton JW, van Ogtrop ML, Andes D, Craig WA. Use of pharmacodynamic indices to predict efficacy of combination therapy in vivo. Antimicrob Agents Chemother. 1999;43(10):2473–8. doi: 10.1128/aac.43.10.2473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Vogelman B, Gudmundsson S, Leggett J, Turnidge J, Ebert S, Craig WA. Correlation of antimicrobial pharmacokinetic parameters with therapeutic efficacy in an animal model. J Infect Dis. 1988;158:831–47. doi: 10.1093/infdis/158.4.831. [DOI] [PubMed] [Google Scholar]
  • 44.Ryan DM. Pharmacokinetics of antibiotics in natural and experimental superficial compartments in animals and humans. J Antimicrob Chemother. 1993;31(Suppl D):1–16. doi: 10.1093/jac/31.suppl_d.1. [DOI] [PubMed] [Google Scholar]
  • 45.Joukhadar C, Frossard M, Mayer BX, et al. Impaired target site penetration of beta-lactams may account for therapeutic failure in patients with septic shock. Crit Care Med. 2001;29(2):385–91. doi: 10.1097/00003246-200102000-00030. [DOI] [PubMed] [Google Scholar]
  • 46.Joukhadar C, Klein N, Mayer BX, et al. Plasma and tissue pharmacokinetics of cefpirome in patients with sepsis. Crit Care Med. 2002;30(7):1478–82. doi: 10.1097/00003246-200207000-00013. [DOI] [PubMed] [Google Scholar]
  • 47.Joukhadar C, Klein N, Dittrich P, et al. Target site penetration of fosfomycin in critically ill patients. J Antimicrob Chemother. 2003;51(5):1247–52. doi: 10.1093/jac/dkg187. [DOI] [PubMed] [Google Scholar]
  • 48.Roberts JA, Roberts MS, Robertson TA, Dalley AJ, Lipman J. Piperacillin penetration into tissue of critically ill patients with sepsis--bolus versus continuous administration? Crit Care Med. 2009;37(3):926–33. doi: 10.1097/CCM.0b013e3181968e44. [DOI] [PubMed] [Google Scholar]
  • 49.Zeitlinger MA, Dehghanyar P, Mayer BX, et al. Relevance of soft-tissue penetration by levofloxacin for target site bacterial killing in patients with sepsis. Antimicrob Agents Chemother. 2003;47(11):3548–53. doi: 10.1128/AAC.47.11.3548-3553.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Di Giantomasso D, May CN, Bellomo R. Vital organ blood flow during hyperdynamic sepsis. Chest. 2003;124(3):1053–9. doi: 10.1378/chest.124.3.1053. [DOI] [PubMed] [Google Scholar]
  • 51.Lipman J, Wallis SC, Boots RJ. Cefepime versus cefpirome: the importance of creatinine clearance. Anesth Analg. 2003;97(4):1149–54. doi: 10.1213/01.ANE.0000077077.54084.B0. [DOI] [PubMed] [Google Scholar]
  • 52.Udy AA, Roberts JA, Lipman J. Implications of augmented renal clearance in critically ill patients. Nat Rev Nephrol. 2011;7(9):539–43. doi: 10.1038/nrneph.2011.92. [DOI] [PubMed] [Google Scholar]
  • 53.Fuster-Lluch O, Geronimo-Pardo M, Peyro-Garcia R, Lizan-Garcia M. Glomerular hyperfiltration and albuminuria in critically ill patients. Anaesth Intensive Care. 2008;36(5):674–80. doi: 10.1177/0310057X0803600507. [DOI] [PubMed] [Google Scholar]
  • 54.Udy AA, Varghese JM, Altukroni M, et al. Sub-therapeutic initial beta-lactam concentrations in select critically ill patients: association between augmented renal clearance and low trough drug concentrations. Chest. 2011 doi: 10.1378/chest.11-1671. [DOI] [PubMed] [Google Scholar]
  • 55.Bagshaw SM, George C, Dinu I, Bellomo R. A multi-centre evaluation of the RIFLE criteria for early acute kidney injury in critically ill patients. Nephrol Dial Transplant. 2008;23(4):1203–10. doi: 10.1093/ndt/gfm744. [DOI] [PubMed] [Google Scholar]
  • 56.Choi G, Gomersall CD, Tian Q, Joynt GM, Freebairn R, Lipman J. Principles of antibacterial dosing in continuous renal replacement therapy. Crit Care Med. 2009;37(7):2268–82. doi: 10.1097/CCM.0b013e3181aab3d0. [DOI] [PubMed] [Google Scholar]
  • 57.Jamal JA, Economou CJ, Lipman J, Roberts JA. Improving antibiotic dosing in special situations in the ICU: burns, renal replacement therapy and extracorporeal membrane oxygenation. Curr Opin Crit Care. 2012;18(5):460–71. doi: 10.1097/MCC.0b013e32835685ad. [DOI] [PubMed] [Google Scholar]
  • 58.Bagshaw SM, Uchino S, Bellomo R, et al. Septic acute kidney injury in critically ill patients: clinical characteristics and outcomes. Clin J Am Soc Nephrol. 2007;2(3):431–9. doi: 10.2215/CJN.03681106. [DOI] [PubMed] [Google Scholar]
  • 59.Roberts DM, Roberts JA, Roberts MS, et al. Variability of antibiotic concentrations in critically ill patients receiving continuous renal replacement therapy – a multicentre pharmacokinetic study. Crit Care Med. 2012;40(5):1523–8. doi: 10.1097/CCM.0b013e318241e553. [DOI] [PubMed] [Google Scholar]
  • 60.Seyler L, Cotton F, Taccone FS, et al. Recommended beta-lactam regimens are inadequate in septic patients treated with continuous renal replacement therapy. Crit Care. 2011;15(3):R137. doi: 10.1186/cc10257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Vincent JL, Rello J, Marshall J, et al. International study of the prevalence and outcomes of infection in intensive care units. JAMA. 2009;302(21):2323–9. doi: 10.1001/jama.2009.1754. [DOI] [PubMed] [Google Scholar]
  • 62.Rodvold KA, George JM, Yoo L. Penetration of anti-infective agents into pulmonary epithelial lining fluid: focus on antibacterial agents. ClinPharmacokinet. 2011;50(10):637–64. doi: 10.2165/11594090-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 63.Boselli E, Breilh D, Duflo F, et al. Steady-state plasma and intrapulmonary concentrations of cefepime administered in continuous infusion in critically ill patients with severe nosocomial pneumonia. Crit Care Med. 2003;31(8):2102–6. doi: 10.1097/01.CCM.0000069734.38738.C8. [DOI] [PubMed] [Google Scholar]
  • 64.Kontou P, Chatzika K, Pitsiou G, Stanopoulos I, Argyropoulou-Pataka P, Kioumis I. Pharmacokinetics of ciprofloxacin and its penetration into bronchial secretions of mechanically ventilated patients with chronic obstructive pulmonary disease. Antimicrob Agents Chemother. 2011;55(9):4149–53. doi: 10.1128/AAC.00566-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Boselli E, Breilh D, Rimmele T, et al. Alveolar concentrations of piperacillin/tazobactam administered in continuous infusion to patients with ventilator-associated pneumonia. Crit Care Med. 2008;36(5):1500–6. doi: 10.1097/CCM.0b013e318170ba21. [DOI] [PubMed] [Google Scholar]
  • 66.Boselli E, Breilh D, Rimmele T, et al. Plasma and lung concentrations of ceftazidime administered in continuous infusion to critically ill patients with severe nosocomial pneumonia. Intensive Care Med. 2004;30:989–91. doi: 10.1007/s00134-004-2171-2. [DOI] [PubMed] [Google Scholar]
  • 67.Boselli E, Breilh D, Cannesson M, et al. Steady-state plasma and intrapulmonary concentrations of piperacillin/tazobactam 4 g/0.5 g administered to critically ill patients with severe nosocomial pneumonia. IntensCare Med. 2004;30(5):976–9. doi: 10.1007/s00134-004-2222-8. [DOI] [PubMed] [Google Scholar]
  • 68.Lodise TP, Sorgel F, Melnick D, Mason B, Kinzig M, Drusano GL. Penetration of meropenem into epithelial lining fluid of patients with ventilator-associated pneumonia. Antimicrob Agents Chemother. 2011;55(4):1606–10. doi: 10.1128/AAC.01330-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lu Q, Luo R, Bodin L, et al. Efficacy of high-dose nebulized colistin in ventilator-associated pneumonia caused by multidrug-resistant Pseudomonas aeruginosa and Acinetobacter baumannii. Anesthesiology. 2012;117(6):1335–47. doi: 10.1097/ALN.0b013e31827515de. [DOI] [PubMed] [Google Scholar]
  • 70.McKindley DS, Hanes S, Boucher BA. Hepatic drug metabolism in critical illness. Pharmacotherapy. 1998;18(4):759–78. [PubMed] [Google Scholar]
  • 71.Ulldemolins M, Roberts JA, Lipman J, Rello J. Antibiotic dosing in multiple organ dysfunction syndrome. Chest. 2011;139(5):1210–20. doi: 10.1378/chest.10-2371. [DOI] [PubMed] [Google Scholar]
  • 72.Hope WW, Vanguilder M, Donnelly JP, et al. Software for dosage individualization of voriconazole for immunocompromised patients. Antimicrob Agents Chemother. 2013;57(4):1888–94. doi: 10.1128/AAC.02025-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Rhomberg PR, Fritsche TR, Sader HS, Jones RN. Antimicrobial susceptibility pattern comparisons among intensive care unit and general ward Gram-negative isolates from the Meropenem Yearly Susceptibility Test Information Collection Program (USA) Diagn Microbiol Infect Dis. 2006;56(1):57–62. doi: 10.1016/j.diagmicrobio.2005.12.009. [DOI] [PubMed] [Google Scholar]
  • 74.Samtani MN, Flamm R, Kaniga K, Nandy P. Pharmacokinetic pharmacodynamic-model-guided doripenem dosing in critically ill patients. Antimicrob Agents Chemother. 2010;54(6):2360–4. doi: 10.1128/AAC.01843-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Valenza G, Seifert H, Decker-Burgard S, Laeuffer J, Morrissey I, Mutters R. Comparative Activity of Carbapenem Testing (COMPACT) study in Germany. Int J Antimicrob Agents. 2012;39(3):255–8. doi: 10.1016/j.ijantimicag.2011.10.015. [DOI] [PubMed] [Google Scholar]
  • 76.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(3):325–7. doi: 10.1086/600877. [DOI] [PubMed] [Google Scholar]
  • 77.Patzer JA, Dzierzanowska D, Turner PJ. Trends in antimicrobial susceptibility of Gram-negative isolates from a paediatric intensive care unit in Warsaw: results from the MYSTIC programme (1997-2007) J Antimicrob Chemother. 2008;62(2):369–75. doi: 10.1093/jac/dkn184. [DOI] [PubMed] [Google Scholar]
  • 78.Binkley S, Fishman NO, LaRosa LA, et al. Comparison of unit-specific and hospital-wide antibiograms: potential implications for selection of empirical antimicrobial therapy. Infect Control Hosp Epidemiol. 2006;27(7):682–7. doi: 10.1086/505921. [DOI] [PubMed] [Google Scholar]
  • 79.Lee K, Kim MN, Kim JS, et al. Further increases in carbapenem-, amikacin-, and fluoroquinolone-resistant isolates of Acinetobacter spp. and P. aeruginosa in Korea: KONSAR study 2009. Yonsei Med J. 2011;52(5):793–802. doi: 10.3349/ymj.2011.52.5.793. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Gillespie EL, Kuti JL, Nicolau DP. When “S” does not mean success: the importance of choice of antibiotic and dose on clinical and economic outcomes of severe infection. Connecticut Med. 2005;69(4):203–10. [PubMed] [Google Scholar]
  • 81.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(1):79–86. doi: 10.1086/510079. [DOI] [PubMed] [Google Scholar]
  • 82.Kashuba ADM, Nafiziger AN, Drusano GL, Bertino JSJ. Optimizing aminoglycoside therapy for nosocomial pneumonia caused by gram-negative bacteria. Antimicrob Agents Chemother. 1999;43:623–9. doi: 10.1128/aac.43.3.623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Moore RD, Lietman PS, Smith CR. Clinical response to aminoglycoside therapy: importance of the ratio of peak concentration to minimal inhibitory concentration. J Infect Dis. 1987;155:93–9. doi: 10.1093/infdis/155.1.93. [DOI] [PubMed] [Google Scholar]
  • 84.Drusano GL, Ambrose PG, Bhavnani SM, Bertino JS, Nafziger AN, Louie A. Back to the future: using aminoglycosides again and how to dose them optimally. Clin Infect Dis. 2007;45(6):753–60. doi: 10.1086/520991. [DOI] [PubMed] [Google Scholar]
  • 85.Highet VS, Forrest A, Ballow CH, Schentag JJ. Antibiotic dosing issues in lower respiratory tract infection: population-derived area under inhibitory curve is predictive of efficacy. J Antimicrob Chemother. 1999;43(Suppl A):55–63. doi: 10.1093/jac/43.suppl_1.55. [DOI] [PubMed] [Google Scholar]
  • 86.Zelenitsky SA, Harding GK, Sun S, Ubhi K, Ariano RE. Treatment and outcome of Pseudomonas aeruginosa bacteraemia: an antibiotic pharmacodynamic analysis. J Antimicrob Chemother. 2003;52(4):668–74. doi: 10.1093/jac/dkg403. [DOI] [PubMed] [Google Scholar]
  • 87.Tam VH, Ledesma KR, Vo G, Kabbara S, Lim TP, Nikolaou M. Pharmacodynamic modeling of aminoglycosides against Pseudomonas aeruginosa and Acinetobacter baumannii: identifying dosing regimens to suppress resistance development. Antimicrob Agents Chemother. 2008;52(11):3987–93. doi: 10.1128/AAC.01468-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ong CT, Tessier PR, Li C, Nightingale CH, Nicolau DP. Comparative in vivo efficacy of meropenem, imipenem, and cefepime against Pseudomonas aeruginosa expressing MexA-MexB-OprM efflux pumps. Diagn Microbiol Infect Dis. 2007;57(2):153–61. doi: 10.1016/j.diagmicrobio.2006.06.014. [DOI] [PubMed] [Google Scholar]
  • 89.Ariano RE, Nyhlen A, Donnelly JP, Sitar DS, Harding GK, Zelenitsky SA. Pharmacokinetics and pharmacodynamics of meropenem in febrile neutropenic patients with bacteremia. Ann Pharmacother. 2005;39(1):32–8. doi: 10.1345/aph.1E271. [DOI] [PubMed] [Google Scholar]
  • 90.Tam VH, Schilling AN, Melnick DA, Coyle EA. Comparison of beta-lactams in counter-selecting resistance of Pseudomonas aeruginosa. Diagn Microbiol Infect Dis. 2005;52(2):145–51. doi: 10.1016/j.diagmicrobio.2005.02.010. [DOI] [PubMed] [Google Scholar]
  • 91.Tam VH, Schilling AN, Neshat S, Poole K, Melnick DA, Coyle EA. Optimization of meropenem minimum concentration/MIC ratio to suppress in vitro resistance of Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2005;49(12):4920–7. doi: 10.1128/AAC.49.12.4920-4927.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.McKinnon PS, Paladino JA, Schentag JJ. Evaluation of area under the inhibitory curve (AUIC) and time above the minimum inhibitory concentration (T>MIC) as predictors of outcome for cefepime and ceftazidime in serious bacterial infections. Int J Antimicrob Agents. 2008;31(4):345–51. doi: 10.1016/j.ijantimicag.2007.12.009. [DOI] [PubMed] [Google Scholar]
  • 93.Tam VH, McKinnon PS, Akins RL, Rybak MJ, Drusano GL. Pharmacodynamics of cefepime in patients with Gram-negative infections. J Antimicrob Chemother. 2002;50(3):425–8. doi: 10.1093/jac/dkf130. [DOI] [PubMed] [Google Scholar]
  • 94.Knudsen JD, Odenholt I, Erlendsdottir H, et al. Selection of resistant Streptococcus pneumoniae during penicillin treatment in vitro and in three animal models. Antimicrob Agents Chemother. 2003;47(8):2499–506. doi: 10.1128/AAC.47.8.2499-2506.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Craig WA, Andes D. Pharmacokinetics and pharmacodynamics of antibiotics in otitis media. Pediatr Infect Dis J. 1996;15(3):255–9. doi: 10.1097/00006454-199603000-00015. [DOI] [PubMed] [Google Scholar]
  • 96.Zelenitsky SA, Ariano RE. Support for higher ciprofloxacin AUC 24/MIC targets in treating Enterobacteriaceae bloodstream infection. J Antimicrob Chemother. 2010;65(8):1725–32. doi: 10.1093/jac/dkq211. [DOI] [PubMed] [Google Scholar]
  • 97.Preston SL, Drusano GL, Berman AL. Pharmaco-dynamics of levofloxacin: a new paradigm for early clinical trials. JAMA. 1998;279:125–9. doi: 10.1001/jama.279.2.125. [DOI] [PubMed] [Google Scholar]
  • 98.Peloquin CA, Cumbo TJ, Nix DE, Sands MF, Schentag JJ. Evaluation of intravenous ciprofloxacin in patients with nosocomial lower respiratory tract infections. Impact of plasma concentrations, organism, minimum inhibitory concentration, and clinical condition on bacterial eradication. Arch Intern Med. 1989;149(10):2269–73. [PubMed] [Google Scholar]
  • 99.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(2):275–85. doi: 10.1172/JCI16814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Tam VH, Louie A, Deziel MR, Liu W, Leary R, Drusano GL. Bacterial-population responses to drug-selective pressure: examination of garenoxacin’s effect on Pseudomonas aeruginosa. J Infect Dis. 2005;192(3):420–8. doi: 10.1086/430611. [DOI] [PubMed] [Google Scholar]
  • 101.Olofsson SK, Marcusson LL, Komp Lindgren P, Hughes D, Cars O. Selection of ciprofloxacin resistance in Escherichia coli in an in vitro kinetic model: relation between drug exposure and mutant prevention concentration. J Antimicrob Chemother. 2006;57(6):1116–21. doi: 10.1093/jac/dkl135. [DOI] [PubMed] [Google Scholar]
  • 102.Drusano GL, Preston SL, Fowler C, Corrado M, Weisinger B, Kahn J. Relationship between fluoroquinolone area under the curve: minimum inhibitory concentration ratio and the probability of eradication of the infecting pathogen, in patients with nosocomial pneumonia. J Infect Dis. 2004;189(9):1590–7. doi: 10.1086/383320. [DOI] [PubMed] [Google Scholar]
  • 103.Craig WA. Basic pharmacodynamics of antibacterials with clinical applications to the use of b-lactams, glycopeptides, and linezolid. Infect Dis Clin N Am. 2003;17:479–501. doi: 10.1016/s0891-5520(03)00065-5. [DOI] [PubMed] [Google Scholar]
  • 104.Firsov AA, Smirnova MV, Lubenko IY, Vostrov SN, Portnoy YA, Zinner SH. Testing the mutant selection window hypothesis with Staphylococcus aureus exposed to daptomycin and vancomycin in an in vitro dynamic model. J Antimicrob Chemother. 2006;58(6):1185–92. doi: 10.1093/jac/dkl387. [DOI] [PubMed] [Google Scholar]
  • 105.van Ogtrop ML, Andes D, Stamstad TJ, et al. In vivo pharmacodynamic activities of two glycylcyclines (GAR-936 and WAY 152,288) against various gram-positive and gram-negative bacteria. Antimicrob Agents Chemother. 2000;44(4):943–9. doi: 10.1128/aac.44.4.943-949.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Meagher AK, Ambrose PG, Grasela TH, Ellis-Grosse EJ. Pharmacokinetic/pharmacodynamic profile for tigecycline-a new glycylcycline antimicrobial agent. Diagn Microbiol Infect Dis. 2005;52(3):165–71. doi: 10.1016/j.diagmicrobio.2005.05.006. [DOI] [PubMed] [Google Scholar]
  • 107.Rubino CM, Bhavnani SM, Forrest A, et al. Pharmacokinetics pharmacodynamics of tigecycline in patients with community-acquired pneumonia. Antimicrob Agents Chemother. 2012;56(1):130–6. doi: 10.1128/AAC.00277-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Bhavnani SM, Rubino CM, Hammel JP, et al. Pharmacological and patient-specific response determinants in patients with hospital-acquired pneumonia treated with tigecycline. Antimicrob Agents Chemother. 2012;56(2):1065–72. doi: 10.1128/AAC.01615-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Meagher AK, Ambrose PG, Grasela TH, Ellis-Grosse EJ. The pharmacokinetic and pharmacodynamic profile of tigecycline. Clin Infect Dis. 2005;41(Suppl 5):S333–40. doi: 10.1086/431674. [DOI] [PubMed] [Google Scholar]
  • 110.Passarell JA, Meagher AK, Liolios K, et al. Exposure-response analyses of tigecycline efficacy in patients with complicated intra-abdominal infections. Antimicrob Agents Chemother. 2008;52(1):204–10. doi: 10.1128/AAC.00813-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Dandekar PK, Tessier PR, Williams P, Nightingale CH, Nicolau DP. Pharmacodynamic profile of daptomycin against Enterococcus species and methicillin-resistant Staphylococcus aureus in a murine thigh infection model. J Antimicrob Chemother. 2003;52(3):405–11. doi: 10.1093/jac/dkg337. [DOI] [PubMed] [Google Scholar]
  • 112.Safdar N, Andes D, Craig WA. In vivo pharmacodynamic activity of daptomycin. Antimicrob Agents Chemother. 2004;48(1):63–8. doi: 10.1128/AAC.48.1.63-68.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.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(3):1117–24. doi: 10.1128/AAC.01114-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Dudhani RV, Turnidge JD, Nation RL, Li J. 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(9):1984–90. doi: 10.1093/jac/dkq226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Marik PE, Lipman J, Kobilski S, Scribante J. A prospective randomized study comparing once-versus twice-daily amikacin dosing in critically ill adult and paediatric patients. J Antimicrob Chemother. 1991;28(5):753–64. doi: 10.1093/jac/28.5.753. [DOI] [PubMed] [Google Scholar]
  • 116.Munckhof WJ, Grayson ML, Turnidge JD. A meta-analysis of studies on the safety and efficacy of aminoglycosides given either once daily or as divided doses. J Antimicrob Chemother. 1996;37(4):645–63. doi: 10.1093/jac/37.4.645. [DOI] [PubMed] [Google Scholar]
  • 117.Crandon JL, Ariano RE, Zelenitsky SA, Nicasio AM, Kuti JL, Nicolau DP. Optimization of meropenem dosage in the critically ill population based on renal function. Intensive Care Med. 2011;37(4):632–8. doi: 10.1007/s00134-010-2105-0. [DOI] [PubMed] [Google Scholar]
  • 118.Drusano GL. Prevention of resistance: a goal for dose selection for antimicrobial agents. Clin Infect Dis. 2003;36(Suppl 1):S42–50. doi: 10.1086/344653. [DOI] [PubMed] [Google Scholar]
  • 119.Lodise TP, Jr., Lomaestro B, Rodvold KA, Danziger LH, Drusano GL. Pharmacodynamic profiling of piperacillin in the presence of tazobactam in patients through the use of population pharmacokinetic models and Monte Carlo simulation. Antimicrob Agents Chemother. 2004;48(12):4718–24. doi: 10.1128/AAC.48.12.4718-4724.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Lomaestro BM, Drusano GL. Pharmacodynamic evaluation of extending the administration time of meropenem using a Monte Carlo simulation. Antimicrob Agents Chemother. 2005;49(1):461–3. doi: 10.1128/AAC.49.1.461-463.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Nicasio AM, Ariano RE, Zelenitsky SA, et al. Population pharmacokinetics of high-dose, prolonged-infusion cefepime in adult critically ill patients with ventilator-associated pneumonia. Antimicrob Agents Chemother. 2009;53(4):1476–81. doi: 10.1128/AAC.01141-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Nicolau DP, McNabb J, Lacy MK, Quintiliani R, Nightingale CH. Continuous versus intermittent administration of ceftazidime in intensive care unit patients with nosocomial pneumonia. Int J Antimicrob Agents. 2001;17(6):497–504. doi: 10.1016/s0924-8579(01)00329-6. [DOI] [PubMed] [Google Scholar]
  • 123.Roberts JA, Kirkpatrick CM, Roberts MS, Dalley AJ, Lipman J. First-dose and steady-state population pharmacokinetics and pharmacodynamics of piperacillin by continuous or intermittent dosing in critically ill patients with sepsis. Int J Antimicrob Agents. 2010;35(2):156–63. doi: 10.1016/j.ijantimicag.2009.10.008. [DOI] [PubMed] [Google Scholar]
  • 124.Roberts JA, Lipman J. Optimal doripenem dosing simulations in critically ill nosocomial pneumonia patients with obesity, augmented renal clearance, and decreased bacterial susceptibility. CritCare Med. 2013;41(2):489–95. doi: 10.1097/CCM.0b013e31826ab4c4. [DOI] [PubMed] [Google Scholar]
  • 125.Dulhunty JM, Roberts JA, Davis JS, et al. Continuous infusion of beta-lactam antibiotics in severe sepsis: a multicenter double-blind, randomized controlled trial. Clin Infect Dis. 2013;56(2):236–44. doi: 10.1093/cid/cis856. [DOI] [PubMed] [Google Scholar]
  • 126.Roberts JA, Webb S, Paterson D, Ho KM, Lipman J. A systematic review on clinical benefits of continuous administration of beta-lactam antibiotics. Crit Care Med. 2009;37(6):2071–8. doi: 10.1097/CCM.0b013e3181a0054d. [DOI] [PubMed] [Google Scholar]
  • 127.Kasiakou SK, Sermaides GJ, Michalopoulos A, Soteriades ES, Falagas ME. Continuous versus intermittent intravenous administration of antibiotics: a meta-analysis of randomised controlled trials. Lancet Infect Dis. 2005;5(9):581–9. doi: 10.1016/S1473-3099(05)70218-8. [DOI] [PubMed] [Google Scholar]
  • 128.Arnold HM, Hollands JM, Skrupky LP, et al. Prolonged infusion antibiotics for suspected gram-negative infections in the ICU: a before-after study. Ann Pharmacother. 2013;47(2):170–80. doi: 10.1345/aph.1R523. [DOI] [PubMed] [Google Scholar]
  • 129.Lodise TP, Jr., Lomaestro B, Drusano GL. Piperacillin-tazobactam for Pseudomonas aeruginosa infection: clinical implications of an extended-infusion dosing strategy. Clin Infect Dis. 2007;44(3):357–63. doi: 10.1086/510590. [DOI] [PubMed] [Google Scholar]
  • 130.Wysocki M, Delatour F, Faurisson F, et al. Continuous versus intermittent infusion of vancomycin in severe Staphylococcal infections: prospective multicenter randomized study. Antimicrob Agents Chemother. 2001;45(9):2460–7. doi: 10.1128/AAC.45.9.2460-2467.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Rello J, Sole-Violan J, Sa-Borges M, et al. Pneumonia caused by oxacillin-resistant Staphylococcus aureus treated with glycopeptides. Crit Care Med. 2005;33(9):1983–7. doi: 10.1097/01.ccm.0000178180.61305.1d. [DOI] [PubMed] [Google Scholar]
  • 132.Roberts JA, Norris R, Paterson DL, Martin JH. Therapeutic Drug Monitoring of antimicrobials. Br J Clin Pharmacol. 2012;73(1):27–36. doi: 10.1111/j.1365-2125.2011.04080.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Wong G, Briscoe S, Adnan S, et al. Protein Binding of beta-Lactam Antibiotics in Critically Ill Patients: Can We Successfully Predict Unbound Concentrations? Antimicrob Agents Chemother. 2013;57(12):6165–70. doi: 10.1128/AAC.00951-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Gauthier T, Lacarelle B, Marre F, Villard PH, Catalin J, Durand A. Predictive performance of two software packages (USC*PACK PC and Abbott PKS system) for the individualization of amikacin dosage in intensive care unit patients. IntJBiomedComp. 1994;36(1-2):131–4. doi: 10.1016/0020-7101(94)90104-x. [DOI] [PubMed] [Google Scholar]
  • 135.Pea F, Brollo L, Viale P, Pavan F, Furlanut M. Teicoplanin therapeutic drug monitoring in critically ill patients: a retrospective study emphasizing the importance of a loading dose. J Antimicrob Chemother. 2003;51(4):971–5. doi: 10.1093/jac/dkg147. [DOI] [PubMed] [Google Scholar]
  • 136.Delattre IK, Musuamba FT, Verbeeck RK, et al. Empirical models for dosage optimization of four beta-lactams in critically ill septic patients based on therapeutic drug monitoring of amikacin. Clin Biochem. 2010;43(6):589–98. doi: 10.1016/j.clinbiochem.2009.12.007. [DOI] [PubMed] [Google Scholar]
  • 137.Pea F, Furlanut M, Cojutti P, et al. Therapeutic drug monitoring of linezolid: a retrospective monocentric analysis. Antimicrob Agents Chemother. 2010;54(11):4605–10. doi: 10.1128/AAC.00177-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Finnell DL, Davis GA, Cropp CD, Ensom MH. Validation of the Hartford nomogram in trauma surgery patients. Ann Pharmacother. 1998;32(4):417–21. doi: 10.1345/aph.17243. [DOI] [PubMed] [Google Scholar]
  • 139.Pea F, Furlanut M, Negri C, et al. Prospectively validated dosing nomograms for maximizing the pharmacodynamics of vancomycin administered by continuous infusion in critically ill patients. Antimicrob Agents Chemother. 2009;53(5):1863–7. doi: 10.1128/AAC.01149-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Pea F, Viale P, Cojutti P, Furlanut M. Dosing nomograms for attaining optimum concentrations of meropenem by continuous infusion in critically ill patients with severe gram-negative infections: a pharmacokinetics/pharmacodynamics-based approach. Antimicrob Agents Chemother. 2012;56(12):6343–8. doi: 10.1128/AAC.01291-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Mouton JW, Vinks AA. Is continuous infusion of beta-lactam antibiotics worthwhile?--efficacy and pharmacokinetic considerations. J Antimicrob Chemother. 1996;38(1):5–15. doi: 10.1093/jac/38.1.5. [DOI] [PubMed] [Google Scholar]
  • 142.Muller AE, Schmitt-Hoffmann AH, Punt N, Mouton JW. Monte Carlo simulations based on phase 1 studies predict target attainment of ceftobiprole in nosocomial pneumonia patients: a validation study. Antimicrob Agents Chemother. 2013;57(5):2047–53. doi: 10.1128/AAC.02292-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Tam VH, Kabbara S, Yeh RF, Leary RH. Impact of Sample Size on the Performance of Multiple-Model Pharmacokinetic Simulations. Antimicrob Agents Chemother. 2006;50(11):3950–2. doi: 10.1128/AAC.00337-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Roberts JA, Kirkpatrick CM, Lipman J. Monte Carlo simulations: maximizing antibiotic pharmacokinetic data to optimize clinical practice for critically ill patients. J Antimicrob Chemother. 2011;66(2):227–231. doi: 10.1093/jac/dkq449. [DOI] [PubMed] [Google Scholar]
  • 145.Bayard DS, Milman MH, Schumitzky A. Design of dosage regimens: a multiple model stochastic control approach. Int J Biomed Comp. 1994;36(1-2):103–15. doi: 10.1016/0020-7101(94)90100-7. [DOI] [PubMed] [Google Scholar]
  • 146.Fuchs A, Csajka C, Thoma Y, Buclin T, Widmer N. Benchmarking therapeutic drug monitoring software: a review of available computer tools. ClinPharmacokinet. 2013;52(1):9–22. doi: 10.1007/s40262-012-0020-y. [DOI] [PubMed] [Google Scholar]

RESOURCES