You are on page 1of 16

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/267045847

Antibiotic Dosing in Patients With Acute Kidney Injury: "Enough But Not Too
Much"

Article  in  Journal of Intensive Care Medicine · October 2014


DOI: 10.1177/0885066614555490 · Source: PubMed

CITATIONS READS

19 572

2 authors, including:

Bruce A Mueller
University of Michigan
168 PUBLICATIONS   2,971 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Pharmacogenetics View project

All content following this page was uploaded by Bruce A Mueller on 22 December 2014.

The user has requested enhancement of the downloaded file.


Journal of Intensive Care Medicine
http://jic.sagepub.com/

Antibiotic Dosing in Patients With Acute Kidney Injury: ''Enough But Not Too Much''
Susan J. Lewis and Bruce A. Mueller
J Intensive Care Med published online 16 October 2014
DOI: 10.1177/0885066614555490

The online version of this article can be found at:


http://jic.sagepub.com/content/early/2014/10/15/0885066614555490

Published by:

http://www.sagepublications.com

Additional services and information for Journal of Intensive Care Medicine can be found at:

Email Alerts: http://jic.sagepub.com/cgi/alerts

Subscriptions: http://jic.sagepub.com/subscriptions

Reprints: http://www.sagepub.com/journalsReprints.nav

Permissions: http://www.sagepub.com/journalsPermissions.nav

>> OnlineFirst Version of Record - Oct 16, 2014

What is This?

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


Journal of Intensive Care Medicine
1-14
Antibiotic Dosing in Patients With ª The Author(s) 2014
Reprints and permission:
sagepub.com/journalsPermissions.nav
Acute Kidney Injury: ‘‘Enough But DOI: 10.1177/0885066614555490
jic.sagepub.com
Not Too Much’’

Susan J. Lewis, PharmD1 and Bruce A. Mueller, PharmD, FCCP, FASN, FNKF1

Abstract
Increasing evidence suggests that antibiotic dosing in critically ill patients with acute kidney injury (AKI) often does not achieve
pharmacodynamic goals, and the continued high mortality rate due to infectious causes appears to confirm these findings.
Although there are compelling reasons why clinicians should use more aggressive antibiotic dosing, particularly in patients
receiving aggressive renal replacement therapies, concerns for toxicity associated with higher doses are real. The presence of
multisystem organ failure and polypharmacy predispose these patients to drug toxicity. This article examines the pharmacokinetic
and pharmacodynamic consequences of critical illness, AKI, and renal replacement therapy and describes potential solutions to
help clinicians give ‘‘enough but not too much’’ in these very complicated patients.

Keywords
antibiotics, pharmacokinetics, pharmacodynamics, acute kidney injury, renal replacement therapy

Introduction conservative management are unlikely to be able to remove


15 L of extra volume in a timely manner.
A 68-year-old female (height 5 ft 2 in, weight 95 kg) was
The Surviving Sepsis Guidelines emphasize the significance
admitted for complaint of increasing abdominal pain and
of ‘‘appropriate antimicrobial therapy,’’ which is defined as
altered mental status. Her past medical history is extensive
‘‘initial empiric anti-infective therapy of one or more drugs that
including recurrent urosepsis caused by b-lactamase-positive
have activity against all likely pathogens and that penetrate in
Escherichia coli. In the intensive care unit (ICU), she has not adequate concentrations into tissue presumed to be the source
yet received empiric antibiotic therapy but did get aggressive
of sepsis.’’1 However, even in the straightforward case men-
fluid resuscitation in the emergency department. The patient’s
tioned earlier, numerous barriers exist to achieve ‘‘adequate
oxygen saturation dropped rapidly and she developed respira-
concentrations’’ at the infection site. Recent evidence suggests
tory failure and septic shock, requiring mechanical ventilation
that clinicians prescribe antibiotics with doses that are higher
with oxygen and vasopressor support. The laboratory results
than what is recommended for the patients’ glomerular filtra-
include serum creatinine (SCr) 2.2 mg/dL and blood urea nitro-
tion rate in elderly patients with chronic kidney disease stages
gen (BUN) 45 mg/dL (baseline SCr 1.1 mg/dL, BUN 23 mg/dL
IV and V,7,8 increasing the risk of antibiotic toxicity. However,
1 month ago). The patient’s weight in the nursing home chart 1 in critically ill patients with AKI, higher doses may be war-
week ago was 80 kg. Susceptibility results are pending. The
ranted. Appropriate antibiotic therapy requires a balance
ICU team needs to prescribe antibiotic therapy.
between a need for doses high enough to account for pharma-
This complicated scenario is not unlike those faced regu-
cokinetic alterations in these patients, pathogen resistance that
larly by clinicians. The Surviving Sepsis Guidelines provide
very straightforward guidance on how to treat patients with
sepsis,1 yet the patient in the previous case comes with some
1
challenges that are not readily apparent. Much of the dosing Department of Clinical, Social, and Administrative Sciences, University of
of her antibiotics is predicated on knowledge of her renal func- Michigan College of Pharmacy, Ann Arbor, MI, USA
tion, and it is apparent in this case that she has acute kidney Received March 13, 2014, and in revised form August 4, 2014. Accepted
injury (AKI), which is a common complication of sepsis in the for publication August 25, 2014.
ICU2 and is associated with high morbidity and mortality
(60%-77%).3-5 About 70% of patients with AKI in the ICU Corresponding Author:
Bruce A. Mueller, Department of Clinical, Social and Administrative Sciences,
require some type of renal replacement therapy (RRT),6 and University of Michigan College of Pharmacy, 428 Church St, Ann Arbor, MI
with the degree of fluid overload manifested by this patient, 48109, USA.
it is likely that RRT will be necessary as diuretics and Email: muellerb@med.umich.edu

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


2 Journal of Intensive Care Medicine

target attainment for the infecting pathogens in this patient pop-


ulation.19-22 Many factors influence antibiotic concentrations at
the infection site, beginning with the numerous pharmacokinetic
changes seen in critically ill patients.

Pharmacokinetic Alterations in Critically Ill


Patients With AKI
Absorption
With the occasional exception of oral fluoroquinolones and line-
zolid, critically ill patients with AKI do not generally receive oral
antibiotics. It is well documented that oral drug absorption is
decreased in the critically ill patients.23-26 Although not com-
pletely understood, underlying causes of gastrointestinal dysmo-
tility may include postoperative ileus, opioid use, mechanical
ventilation, trauma, head injury, and sepsis.25,26 In a retrospective
study, gastrointestinal dysmotility was observed in 60% of ICU
patients, which can have profound effects on drug absorption
from gastrointestinal tract.27 Antibiotics may adhere to the feed-
ing tubes or interact with coadministered nutritional products and
Figure 1. Enough but not too much. The clinician’s decision to
choose higher versus lower antibiotic doses in critically ill patients other drugs, resulting in reduced bioavailability.28,29 Elevated
with acute kidney disease requires balancing often conflicting factors. gastric pH by histamine 2 receptor antagonists or proton pump
inhibitors is associated with the absorption of weak bases such
as ketoconazole.30-32 Absorption from intravenous drugs is not
is common in the ICU,9 and extracorporeal drug clearance ver-
usually altered, but absorption of subcutaneous drugs may be
sus concerns of antibiotic toxicity in these vulnerable patients
affected by impaired peripheral circulation by sepsis and use of
with impaired drug clearance (Figure 1). The purpose of this
vasopressors and peripheral edema during intravenous fluid
article is to explore the challenges of providing appropriate
resuscitation, reducing their bioavailabilities.33,34 Fortunately,
antibiotic doses to attain adequate concentrations at the site
antibiotics are not administered by this route obviating this con-
of infection and to suggest alternative strategies balancing
cern, although many other drugs in the ICU are administered sub-
necessity and risk of antibiotic higher doses to maximize anti-
cutaneously (heparins, insulin, etc).
biotic efficacy while minimizing toxicity.

Distribution
Mortality Related to Inappropriate Antibiotic
The pathogenesis of sepsis involves endothelial abnormalities,
Dosing increase in capillary permeability, and the fluid accumulation
Mortality rates in critically ill patients with sepsis reported in interstitial spaces, resulting in increased antibiotic volume
from various regions around the world range from 20% to of distribution.35 Expanded volume of distribution may be
60%.10-14 In the United States, severe sepsis and septic shock especially prominent with hydrophilic drugs such as aminogly-
account for approximately 10% of all mortality, representing cosides, b-lactams, glycopeptides, and daptomycin. Many
a substantial health care burden.10 Consequently, appropriate researchers have demonstrated decreased serum levels of anti-
antibiotic therapy is essential for both humanistic and eco- biotics in critically ill patients secondary to increased volume
nomic reasons. However, available data suggest that inap- of distribution, suggesting the need for higher doses than stan-
propriate antibiotic therapy in patients with sepsis in the ICU dard dosing recommendations to maintain suitable serum con-
is prevalent and is the most important independent risk factor centrations.22,36-44 The increased volume of distribution is
for hospital mortality.15-18 The primary reasons for inap- more pronounced in severe sepsis and in the early stages of sep-
propriate antibiotic therapy have been identified as antibiotic sis. Patients with hyperdynamic sepsis have a significantly
resistance and absence of efficacious therapy in most larger volume of distribution of gentamicin (0.48 L/kg), com-
studies.15 However, attention should be paid to the other evi- pared with those who have hypodynamic sepsis (0.32 L/kg) and
dence showing that even in the case where pathogens are sus- nonseptic patients receiving postsurgical prophylactic doses of
ceptible to the prescribed antibiotics, hospital mortality still gentamicin (0.29 L/kg).38 Taccone et al noted that the first con-
remains high.9 It is not enough to select the appropriate anti- ventional dose of broad-spectrum b-lactams administered in
biotic, but one must also prescribe doses sufficient to deliver suf- the early stage of severe sepsis and septic shock failed to
ficient antibiotic concentrations at the site of infection. achieve adequate concentrations due to increased volume of
Conventional antibiotic dosing may not yield pharmacodynamic distribution.19 Significantly elevated volume of distribution is

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


Lewis and Mueller 3

also observed in critically ill patients with AKI receiving con- free drug is responsible for pharmacologic effects, protein bind-
tinuous RRT (CRRT) after the administration of the first dose ing changes strongly influence efficacy and toxicity.55 Unbound
of daptomycin which possesses both hydrophilic and lipophilic drugs will distribute into tissues, thus increasing volume of dis-
properties (0.23 L/kg42,45 vs 0.08-0.15 L/kg in healthy volun- tribution. Further, unbound drugs will be cleared by kidneys and/
teers46). However, the initially elevated volume of distribution or RRT, thereby increasing drug clearance. Consequently,
tends to decline over the treatment course36,45 possibly due to protein-binding changes in critical illness can have a significant
physiologic changes or correction of fluid overload by RRTs. effect on highly protein-bound antibiotics. For example, ertape-
Taken together, the increased volume of distribution of hydro- nem, a highly protein-bound (85%-95%) carbapenem, yielded
philic antibiotics in the early stage of sepsis or severe sepsis markedly larger volume of distribution (0.21-0.4 L/kg vs 0.07
necessitates a higher dose or a loading dose to rapidly achieve L/kg) and higher total clearance (43 mL/min vs 20 mL/min) in
adequate concentrations to meet Surviving Sepsis Guidelines. patients with hypoalbuminemia and sepsis, compared with
However, increased volume of distribution will normalize after healthy individuals.56,57 Pharmacokinetic changes in this magni-
a few days of treatment, therefore, clinicians may need to move tude should result in dosing increases to maintain therapeutic
to more conventional dosing over the course of therapy to avoid ertapenem plasma concentrations. However, as renal function
toxicity. Not only can pathophysiologic changes related to sep- improves, gradual normalization of protein-binding capacity is
sis influence volume of distribution in critically ill patients, but observed in patients with AKI.58,59
fluid therapy to resuscitate patients with sepsis can alter it as
well. Volume of distribution of amikacin has been shown to
range widely in a patient receiving aggressive fluid support Metabolism
(0.27 to 0.61 L/kg), compared to that of healthy individuals Hepatic metabolism depends on primarily hepatic blood flow,
(0.27 + 0.06 L/kg).47 Other sources of fluid administration enzymatic activity, and protein binding.60,61 Alteration in one
in these patients, parenteral nutrition and intravenous drug or more of these physiologic processes will translate into
administration, add even more volume in this population, changes in hepatic drug metabolism. The effect of AKI on
although their impact may not be readily appreciated. The vol- hepatic blood flow is uncertain, but different stages of sep-
ume of distribution of aminoglycosides is increased substan- sis and fluid status can affect hepatic blood flow.24 Use
tially in critically ill patients receiving both total parenteral of vasoconstrictors can decrease total hepatic blood flow,
nutrition and fluid therapy, compared with those receiving only while inotropes like dobutamine and dopamine have been
fluid therapy.48,49 Clinicians must recognize the influence of all shown to increase hepatic perfusion by enhancing cardiac
the prescribed volume via fluid, blood products, parenteral output.24 Acute kidney injury may also affect Cytochrome
nutrition, and drug infusions on antibiotic volume of distribu- P450 (CYP) enzyme activity. Animal studies indicate that
tion and may need to use higher antibiotic doses to compensate. effects of AKI on hepatic metabolism are drug specific, but one
The influence of fluid overload on antibiotic concentrations human study demonstrated AKI reduces the activity of CYP 3A
and outcomes in critically ill patients has not been demon- enzymes,62 which play a significant metabolic role for more than
strated definitively. Several observational studies have high- 50% of all drugs. Drugs metabolized by the liver may exert pro-
lighted that fluid overload is associated with an increased longed therapeutic efficacy by sustaining elevated drug serum
mortality rate in critically ill patients with AKI.50-53 The Fluids concentrations. On the other hand, delayed pharmacologic
And Catheters Treatment Trial (FACTT) study found that the response may be seen with prodrugs requiring activation via
risk of mortality was increased 1.6-fold per L/d of accumulated hepatic enzymes. The mechanisms of altered hepatic metabo-
fluid.52 In a post hoc analysis from The Program to Improve lism in AKI is complicated and poorly understood,60 but clin-
Care in Acute Renal Disease (PICARD) trial among nondia- icians should recognize these potential changes and monitor
lyzed patients, the percentage of fluid overload at the time of drug serum concentrations to make appropriate dosage modifi-
AKI diagnosis was twice as high in nonsurvivors compared cation and to avoid toxicity whenever feasible.
with survivors, and the adjusted odds ratio for mortality related
to fluid overload at AKI diagnosis was 3.14.51 Theoretically,
fluid overload could dilute the serum concentrations of antibio-
Elimination
tics resulting in decreased efficacy and contribute to the Renal drug clearance is a dynamic process involving glomeru-
increased mortality rate. Further studies need to explore the lar filtration, tubular secretion, and reabsorption. In AKI,
impact of hemodilution of antibiotics mediated by fluid over- diminished glomerular filtration and impaired tubular secretion
load on mortality and the necessity of higher antibiotic doses secondary to decreased drug transporter activity can cause
to account for excessive fluid. accumulation of renally eliminated antibiotics.63 Antibiotic
Drug serum protein binding changes observed in critical ill- dosages and dosing frequency may need to be reduced to avoid
ness can have a profound effect on pharmacokinetics. Hypoal- accumulation and toxicity. This is particularly important with
buminemia is reported in 40% to 50% of critically ill antibiotics with a narrow therapeutic index, such as aminogly-
patients.54 As serum protein concentrations decrease, the cosides and vancomycin. These drugs require frequent drug
amount of drug binding to protein will decrease, resulting in level monitoring and subsequent adjustment of dosing
an increased ‘‘free’’ or unbound fraction of drug. Because only schemes. However, it should be noted that compensation of

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


4 Journal of Intensive Care Medicine

decreased renal clearance by increasing other elimination path- the extended duration of PIRRT (8-12 hours) contributes to
ways may occur during AKI. For these drugs, dosage adjust- substantial antibiotic clearance, increasing the risk of pro-
ment on a basis of SCr alone may potentially lead to longed subtherapeutic antibiotic concentrations.76,77 In addi-
subtherapeutic concentrations. Jones et al found that unmodi- tion, the extended duration of PIRRT may result in some
fied ciprofloxacin dosage (400 mg intravenously twice daily) doses administered while dialysis is running, which will affect
did not result in significantly different serum concentrations delivered drug dose. This can further complicate efficacy of
in patients with severe sepsis with renal impairment (creatinine antibiotic therapy.
clearance < 20 mL/min), compared to those with creatinine Appropriate antibiotic dosing in critically ill patients with
clearance > 20 mL/min.64 Enhanced transintestinal and biliary RRT requires understanding of the influences of RRT variables
elimination to compensate for reduced renal clearance may and non-RRT clearance on antibiotic therapy to determine opti-
account for these findings. mal dosing regimens. As evidenced in numerous reports, cur-
Alteration in nonrenal clearances of several antibiotics such rent antibiotic dosing recommendations in RRT often fail to
as imipenem and vancomycin are also noted in patients with reach pharmacodynamic goals.20,44,67,85-90 These findings have
AKI. Overall, estimated nonrenal clearances of these antibiotics prompted several clinicians to consider antibiotic doses higher
decline in patients in AKI compared to that seen in patients with than what are traditionally recommended to attain pharmaco-
normal renal function but tend to be higher than those observed dynamic target.20,44,67,85-90
in patients with end-stage renal disease (ESRD).65-68 However,
as AKI persists, vancomycin nonrenal clearance rates progres-
sively decline and approach that of patients with ESRD.65 Pharmacodynamic Target Attainment
The addition of RRT can markedly increase drug clearance, Although pharmacokinetics predicts how patient physiology
adding another layer to the complexity of antibiotic dosing in cri- affects antibiotic concentrations, pharmacodynamics integrates
tically ill patients. Physicochemical properties of drugs such as pharmacokinetic characteristics with antibacterial effect. In
molecular weight, protein binding, and volume of distribution order to optimize antibiotic therapy, pharmacokinetic para-
will all influence on drug clearance via RRT. Determining drug meters must be accounted for if pharmacodynamic targets are
clearance can be complicated, given the heterogeneity in modal- to be met. This complex pharmacokinetic/pharmacodynamic
ities of RRT.69,70 Duration and intensity of RRT can increase relationship has been explained by characterizing antibiotics
solute clearance,71 and different modes of fluid replacement into 2 different categories, concentration-dependent activity
(predilution or postdilution) will affect solute clearance.69 The or time-dependent activity. For concentration-dependent anti-
details of pharmacokinetic studies of antibiotics in different RRT biotics (aminoglycosides, daptomycin, and fluoroquinolones),
settings are evaluated in several recent reviews.72-77 therapeutic outcome will be maximized when a high peak
With the benefits of hemodynamic stability and fluid con- serum concentration (Cmax) relative to the organism’s mini-
trol, CRRT is often preferred in critically ill patients with AKI, mum inhibitory concentration (MIC) is achieved. With time-
but many antibiotic dosing recommendations in patients dependent activity antibiotics (b-lactams, carbapenems), the
receiving CRRT are based on in vitro and very small pharma- time of the serum concentration above the MIC (T > MIC) is
cokinetic studies using obsolete techniques or are extrapolated the most important determinant of their efficacy. For several
from pharmacokinetic data in noncritically ill patients or antibiotics (vancomycin, linezolid) displaying both characteris-
patients with ESRD receiving intermittent hemodialysis tics, the high ratio of area under the serum concentration–time
(IHD).78 In CRRT, the effluent rate (Qf) and the extraction curve (AUC) to the bacterial MIC has been associated with
coefficient (sieving coefficient [Sc] or saturation coefficient antibacterial success. Besides bacterial killing characteristics,
[Sd]) are 2 primary parameters used to calculate dialytic drug the postantibiotic effect, the persistent suppression of bacterial
clearance (CLcrrt ¼ Qf  Sc or Sd). The extraction coefficient growth after concentrations fall below the MIC for a particular
approximates the percentage of unbound drug and the Qf is organism,91 can influence antibacterial effect and help to deter-
determined from dialysate rate (Qd) and ultrafiltration rate mine optimal antibiotic dosing and frequency of administra-
(Quf; Qf ¼ Qd þ Quf). Higher extraction coefficients and Qfs tion. Table 1 suggests several alternative antibiotic dosing
will result in greater drug clearance. Although several large strategies to maximize the efficacy of antibiotic therapy in cri-
randomized trials indicated that high Qfs do not reduce mortal- tically ill patients with AKI receiving various RRTs, based on
ity rate among critically ill patients,79-81 compared with low Qf, available data and consideration of antibiotic profile and extra-
these trials did not take the impact of high Qfs on increased corporeal antibiotic clearance.
antibiotic clearance into account.82,83 Because high Qfs yield
lower antibiotic concentrations, these studies may be con-
founded by reduced pharmacodynamic target attainment rates Extended Antibiotic Infusion
in patients receiving high Qfs. Optimal efficacy of time-dependent killing antibiotics is best
Clinicians employing prolonged intermittent RRT (PIRRT) correlated with the T > MIC for susceptible organisms. The
will find that <1% of drugs have validated dosing recommen- T > MIC of at least 40% to 60% of the dosing interval has
dations available to them.76,84 Recent reviews suggest that, known to be desirable to yield appropriate antibacterial effect
despite the ‘‘slower than IHD’’ blood and dialysate flow rates, of b-lactams,105,106 but maintaining a longer T > MIC has shown

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


Lewis and Mueller 5

Table 1. Antibiotic Dosing Strategies in Critically Ill Patients With AKI 25 mL/kg/h.93 The continuous infusion regimen generated a
to Improve Pharmacodynamic Target Attainment Rates. constant serum concentration above the MIC (Cmedian 19.1
Antibacterial
mg/L) for 100% of the treatment period, while the intermittent
Profile Potential Dosing Strategies bolus maintained serum concentration above the MIC of
8 mg/L for only 45% of the dosing interval. This 45% T >
Time dependent MIC is thought to be a minimally required T > MIC goal but
b-lactams (1) Continuous or prolonged infusions in might not be sufficient to cover intermediately resistant P
Carbapenems CRRT,83,92-96 but not IHD or PIRRT
aeruginosa, a common pathogen in critically ill patients
Vancomycin (2) More frequent administration of smaller doses
(3) Supplemental dose during PIRRT97,98 and post- (Figure 2). The benefit of continuous infusion has also been
IHD proposed for vancomycin which predominantly has a time-
(4) Weight-based dosing dependent killing activity and will be effectively removed
Concentration dependent by conventional CRRT.65,83,94,95,112,113 A retrospective eva-
Aminoglycosides (1) Loading dose to achieve target luation in patients receiving continuous venovenous hemofil-
Colistin pharmacodynamic goals (peaks) early92-94,99,100 tration or continuous venovenous hemodiafiltration reports
Daptomycin (2) Extended dosing interval for aminoglycosides99
that continuous infusion of vancomycin (16-35 mg/kg/d) after
Fluoroquinolones (3) Predialytic administration of aminoglycosides in
Linezolid IHD or PIRRT101-104 a loading dose of 15 mg/kg could be employed to achieve
Macrolides (4) Weight-based dosing adequate serum concentrations (20-30 mg/mL) on the first day
Metronidazole of therapy.83 However, drug accumulation was noticeable on
Vancomycin day 3 of therapy, and the proportion of patients with excessive
vancomycin trough concentrations (> 30 mg/mL) was signifi-
Abbreviations: AKI, acute kidney injury; CRRT, continuous renal replacement
therapy; IHD, intermittent hemodialysis; PIRRT, prolonged intermittent renal cantly higher in those with higher daily doses. Potential drug
replacement therapy. accumulation and higher risk of toxicity mandates routine
monitoring of vancomycin serum concentration and subse-
quent dosage adjustment for this strategy.85
to further maximize the time-dependent killing effect. McKin- Prolonged infusion is a compromise between a typical
non et al indicated that achievement of T > MIC of 100% for 30-minute antibiotic infusion and continuous infusion. Pro-
b-lactams resulted in much higher clinical cure than T > MIC longed infusion duration can vary but is often as long as half
of less than 100% in patients with sepsis (82% vs 33%).107 the dosing interval and does extend T > MIC. An advantage
Serendipitously, patients with AKI may be able to take an of this practice is that an intravenous line does not have to be
advantage of reduced renal clearance to prolong T > MIC. dedicated solely to continuous antibiotic infusion. Use of pro-
However, RRT drug removal, particularly CRRT, may inter- longed infusion antibiotics has not been published in patients
fere with this advantage and even cause the drug concentration with CRRT. With higher peaks and lower troughs, intermittent
to fall below the MIC for a susceptible organism unless a dos- infusion will produce the same AUC as prolonged infusion, but
ing change is made to account for CRRT clearance. In patients a higher proportion of T > MIC with prolonged infusion theo-
with AKI receiving continuous venovenous hemodiafiltration retically should result in better efficacy.
at an Qf of 1 to 2 L/h, the mean elimination of piperacillin/tazo- This strategy of extended infusions may be particularly
bactam was reported to be nearly equivalent to patients with valuable in a case with reduced susceptibility, high CRRT
normal renal function.108 Seyler and colleagues report that rec- Qf, increased antibiotic volume of distribution, and/or in
ommended b-lactam dosing for critically ill patients under- immunocompromised patients where prolonged serum concen-
going CRRT with the mean Qf of 45 mL/kg/h failed to tration above the MIC may be critical. Compared to shorter
achieve stated pharmacodynamic goals against Pseudomonas conventional infusion times, extended antibiotic infusions can
aeruginosa within 48 hours of antibiotic therapy in 34% to be difficult to employ because of limited vascular access in the
100% of modeled cases.20 Therefore, initiation of CRRT ICU and drug compatibility/stability issues. More frequent
should be followed by the consideration of increased antibiotic administration of smaller doses may be able to mimic the T
doses or alternative administration strategies to prolong T > > MIC of extended infusion without a drop below the organ-
MIC with b-lactams. ism’s MIC. Further studies are warranted to investigate the effi-
Extended antibiotic infusion time can increase T > MIC; cacy and safety of extended antibiotic infusion strategies in the
however, clinically improved outcomes of longer infusions patients with AKI.
continues to be debated.109,110 A few studies show the effec-
tiveness of antibiotic continuous infusion in maintaining steady
state target serum concentrations in critically ill patients receiv- Loading or Postdialysis Supplemental Dose
ing CRRT.92,93,111 A small randomized study involving 6 ICU Loading doses have been traditionally used to achieve early
patients compared meropenem 500 mg loading dose followed adequate antibiotic concentrations or initial high-peak concen-
by 2 g given as continuous infusion over 24 hours versus 1 g trations which are strongly related to clinical response to anti-
every 12 hours as intermittent bolus (10-20 minutes) in patients biotics with concentration-dependent activity including
with continuous venovenous hemodialysis (HD) with an Qf of aminoglycosides.114 The importance of a loading dose is

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


6 Journal of Intensive Care Medicine

maintenance of therapeutic serum concentrations.97 The con-


25
1g q12h Intermittent infusion ventional daptomycin dose of 6 mg/kg recommended for IHD
did not produce targeted AUC values when it was infused
Serum Concentration (mg/L)

20 2g q24h Continuous infusion


within 8 hours before an 8-hour PIRRT session (Qb and
Qd of 160 mL/min) due to increased PIRRT drug
15
clearance.97 Clinicians may use additional postdialytic antibio-
tic doses to accelerate the achievement of pharmacodynamic
10
goals or to prevent unwanted drop in antibiotic level caused
MIC of 8mg/L
by the extended duration of PIRRT.98
5

0 Altered Antibiotic Administration Time


0 2 4 6 8 10 12 14 16 18 20 22 24
Time (h)
Changing antibiotic administration time with relation to RRT
can be a strategy to meet pharmacodynamic goals while
Figure 2. Modeled meropenem steady state concentrations with
accounting for RRT drug clearance. Predialysis administration
continuous infusion versus intermittent infusion in a patient on con- of higher dosage has been suggested to be more efficacious
tinuous renal replacement therapy (CRRT).93 A hypothetical 70-kg than conventional postdialysis aminoglycoside administration
patient with anuria having AKI receiving CRRT with an effluent rate of because it can allow a higher peak concentration to maximize
35 mL/kg/h is modeled. The percentage of time of the serum con- antibacterial pharmacodynamic goals (peak/MIC) while subse-
centration above the minimum inhibitory concentration (MIC; T > quently rapid dialytic clearance can minimize toxicity from
MIC) at 8 mg/L for intermediately resistant Pseudomonas aeruginosa is prolonged drug exposure (Figure 3).101,102,118,119 A simulated
58% with meropenem 1 g every 12 hours intermittent infusion over 30
minutes and 100% with 2 g every 24 hours continuous infusion.
gentamicin 6 mg/kg dose given 1 hour prior to a 4-hour HD
treatment achieved predetermined pharmacodynamic targets
with a mean peak concentration of 31 mg/mL. However, a
reinforced by the growing findings that initial suboptimal anti- simulated post-HD infusion of 1.7 mg/kg dose, the maximum
biotic exposure can reduce the susceptibility of pathogens and dose recommended by the package insert, failed to meet any
facilitate the amplification of resistant strains in a large pharmacodynamic targets (Cmax 8.8 + 3.1 mg/L).103 Use of
inoculum.115,116 Considering the increased volume of distribu- a high predialytic antibiotic dose is also proposed for PIRRT,
tion and constant extracorporeal drug removal, patients receiv- where ‘‘when to dose’’ matters more than ‘‘how much to
ing CRRT may require a loading dose of all types of antibiotics. dose’’ due to unpredictable drug clearance with the prolonged
A subgroup analysis by Taccone et al indicated that patients duration of dialysis.77 Roberts et al showed that gentamicin
with severe sepsis treated with CRRT with the mean Qf of 6 mg/kg every 48 hours given 30 minutes to 1 hour before a
33 mL/kg/h required a loading dose of at least 25 mg/kg of ami- 10-hour PIRRT session can be an effective dosing strategy to
kacin, substantially higher than the usual loading dose,117 to maximize pharmacodynamic target attainment while minimiz-
attain adequate peak concentrations.99 However, while the ing sustained elevated serum concentrations responsible for
25mg/kg loading dose yielded target peak concentrations, the toxicity.104 However, caution should be followed when this
serum concentrations 24 hours later were persistently high. novel strategy is instituted. Delay in dialysis schedule or unex-
Consequently, if this regimen is to be used, the dosing interval pected dialysis discontinuation secondary to clotting or intoler-
should be longer than 24 hours for these CRRT Qfs.99 ance can put a patient at risk of antibiotic toxicity due to
Use of loading doses should not be limited to concentration- inadequate dialytic drug clearance. Thus, when the benefit of
dependent antibiotics. A loading dose can be effective to maximized pharmacokinetic/pharmacodynamic goal is critical
achieve early therapeutic concentration of all antibiotics and a subsequent full dialysis session is ensured, clinicians may
including those with time-dependent activity.94,95,100 A loading consider predialytic administration of a higher aminoglycoside
dose is needed when continuous infusion b-lactams are used dose.
because continuous infusion initially results in subtherapeutic
drug levels until the serum concentration slowly reaches the
equilibration with the tissues.92,93 Early attainment of thera- Weight-Based Dosing
peutic drug concentrations at the infection site should improve The ‘‘one-size-fit-all’’ dosing strategy (eg 500 mg regardless of
increased mortality associated with a delay in appropriate anti- body mass) ignores the variability in body size and body fluid
biotic therapy.1 Consequently, continuous infusion b-lactams composition of critically ill patients, possibly leading to an
should follow a loading dose.96 Other RRTs offer different inappropriately low serum concentration. For example, in the
antibiotic administration challenges. The PIRRT removes a case described at the beginning of this article, we present a
significant amount of antibiotics because of prolonged dialysis patient with *15 L fluid overloaded, sepsis, possibly hypoal-
duration. It may require a higher dose during the duration of buminemia, and in need of RRT. Most penicillin, cephalos-
PIRRT and less when PIRRT is off. Supplemental doses may porin, and carbapenem antibiotics have recommended dosing
be guided by therapeutic drug monitoring to ensure adequate in adults who are ‘‘flat dosed’’ with a dosing interval adjusted

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


Lewis and Mueller 7

may be warranted to ensure adequate local antibiotic concen-


30
trations in these patients.
Pre-dialytic 6mg/kg dose
Serum Concentration (mg/L)

25
Post-dialytic 1.7mg/kg dose
20
Rising Bacterial Resistance Rates
Finally, the increasing prevalence of antibiotic resistance high-
15
lights the significance of ‘‘appropriate’’ antibiotic therapy.
10
Antibiotic resistance in critically ill patients has been related
to increased mortality rates in several studies.9,127,128 In a retro-
5 spective cohort, a significantly higher risk of hospital mortality
was noted in patients with sepsis who were infected with resis-
0 tant gram-negative bacteria (63.4% vs 40.0%).9 Further logistic
0 2 4 6 8 10 12 14 16 18 20 22 24
regression analysis determined resistance to the initial antibio-
Time (h)
tics as an independent risk factor, associated with a doubling of
hospital mortality.9 A prospective study also demonstrated that
Figure 3. Modeled gentamicin predialytic high dose versus postdia- patients infected with high vancomycin MIC ( 2 mg/mL) of
lytic lower dose.103,120 A hypothetical 70-kg patient with anuria having
AKI receiving a 4-hour intermittent hemodialysis (IHD) session is
methicillin-resistant Staphylococcus aureus (MRSA) strains
modeled. Gentamicin predialytic 6 mg/kg dose yielded a high-peak had 20% lower clinical response to vancomycin therapy, com-
serum concentration (Cmax) of 26.8 mg/L which is higher than pro- pared to those infected with low MIC (MIC < 2 mg/mL) MRSA
posed pharmacodynamic target of Cmax  8 mg/L, while postdialytic strains, despite targeted vancomycin trough concentrations of
1.7 mg/kg dose yielded Cmax of 7.6 mg/L. 15 to 20 mg/mL.129 For a pathogen with an elevated MIC,
higher doses of antibiotic therapy will be required to achieve
for AKI. The use of a 500-mg carbapenem/cephalosporin dose targets.130 However, higher antibiotic doses may cause antibio-
in a patient with volume of distribution that is likely 50% to tic toxicity in this vulnerable patient population, as high vanco-
100% larger than that of the ‘‘normal volunteer’’ in whom this mycin trough levels (15-20 mg/mL) were found to be a
dose was derived will have a substantially lower chance to significant predictor of nephrotoxicity development in the lat-
attain pharmacodynamic targets. Use of mg/kg or mg/m2 dos- ter study.129 Alternative antibiotic agents may be considered
ing regimens, like our colleagues who treat pediatric patients in a case with high MIC strains and increased predisposition
usually use, is much more likely to meet these targets. to drug toxicity.131 Treatment of infection in the ICU becomes
more challenging with the emerging incidence of multidrug
resistant and even panantibiotic-resistant pathogens among Aci-
netobacter species, P aeruginosa, carbapenem-resistant Kleb-
Inadequate Concentration at the Target Sites siella species and E coli.132-134 The effectiveness of available
The emphasis of the Surviving Sepsis Guidelines on antibiotics antibiotics and efforts to develop novel antibiotics have been
that ‘‘penetrate in adequate concentrations into tissue presumed declining as well.134 All these circumstances mandate clinicians
to be the source of sepsis’’ is often overlooked because of our to aim toward higher pharmacodynamic targets and higher anti-
limited ability to measure drug concentrations at the actual sites biotic doses to ensure bacterial eradication and to reduce the risk
of infection which mostly occur in tissue. Serum concentrations of further development of antibiotic resistance.127,128 As dis-
are routinely monitored to predict therapeutic efficacy, with a cussed, clinicians should endeavor to maximize antibacterial
theoretical assumption of equilibration between free drug in killing effect by using aggressive antibiotic dosing and/or alter-
plasma and tissue. However, tissue penetration might be native dosing strategies.19,20,41,92,93,102,111,136
impaired in critically ill patients due to altered pathophysiology
and transporter activity.121-123 In a small prospective study,
interstitial concentrations of piperacillin in soft tissues were
Toxicity
7-fold lower than free plasma concentrations in patients with Although there are many compelling reasons to consider more
septic shock.123 Results from animal and human studies aggressive antibiotic dosing in critically ill patients with AKI,
demonstrate that plasma concentrations do not always repre- toxicity from antibiotic doses that are too high is a genuine con-
sent equivalent concentration in the sites of infection,124- cern. These dynamic yet vulnerable patients often have multi-
126
suggesting that poor tissue penetration may result in sub- ple organ dysfunction but often receive many toxic
therapeutic antibiotic concentrations at infection sites and con- medications. Nephrotoxic drugs account for 22% of the total
sequent treatment failure, which is not uncommon in patients medication orders in the adult ICU patients.137 With the pres-
with sepsis. Accordingly, modifying antibiotic doses based ence of numerous risk factors, these patients would be more
on the measured plasma concentration may not guarantee to prone to drug accumulation and toxicity due to AKI. In their
achieve the desired concentration at the sites of infection. 7.5-year retrospective study, Kane-Gill and colleagues identi-
Therefore, in the absence to measure adequacy of antibiotic fied that critically ill patients with AKI were 16 times more
concentration at the infection sites, higher antibiotic dosage prone to manifest drug-induced toxicity compared to those

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


8 Journal of Intensive Care Medicine

without AKI.138 Antibiotics were the most common drug class agent, viewing them as acute issues. However, short-term out-
causing toxicity in the ICU.138 comes during a hospital stay may not reflect the full spectrum
Despite the concern for their well-known nephrotoxicity, of antibiotic toxicity. This approach may underestimate poten-
aminoglycosides and vancomycin remain as mainstays of anti- tial long-term effect on morbidity and mortality of these
biotic therapy in the ICU. Another renowned nephrotoxin, patients.162 Although there is a dearth of studies on this issue,
colistin, has been reintroduced as a last treatment option a prospective observational study reports that after 5-year
for multidrug resistant gram-negative bacterial infections, follow-up, the mortality of survivors from severe AKI requir-
despite its exceedingly high rate of nephrotoxicity ing RRT was 47%.163 The causes of AKI in these patients
(~50%).139,140 Treatment with these nephrotoxic agents requires include sepsis (33%) and nephrotoxins (7%). Among those sur-
the clinician to make difficult choices when it is apparent that viving patients, only 57% of them had complete recovery of
antibiotic toxicity is likely to develop. A study with 201 criti- renal function at hospital discharge. Patients with partial renal
cally ill patients treated with aminoglycosides reported up to function recovery showed significantly higher mortality rate,
30% of incidence of nephrotoxicity.141 Moreover, fever, endo- compared to those with complete recovery after 5 years (P ¼
toxemia, and renal hypoperfusion associated with sepsis and .001).163 Therefore, short-term toxicity caused by antibiotics
AKI can aggravate preexisting renal impairment with the use may carry the prolonged risk of deteriorating organ function
of aminoglycosides.142 and increased probability of mortality as a long-term conse-
Although the guidelines suggest raising goal trough serum quence in critically ill patients.
concentrations for vancomycin, a recent meta-analysis suggests Antibiotic toxicity is also associated with substantial
that patients with higher trough serum concentrations (>15 mg/ increase in health care costs, although it may be understated
dL) had a 2- to 3-time greater risk of nephrotoxicity than those compared to the cost related to antibiotic treatment failure.
who had lower troughs (<15 mg/dL).143 High dose, longer Unfortunately, data quantifying both direct and indirect costs
duration of therapy, concomitant use of other nephrotoxins, and of antibiotic toxicity are scarce. In an economic evaluation of
renal impairment are common risk factors to precipitate aminoglycoside nephrotoxicity, the total hospital costs includ-
nephrotoxicity with these antibiotics.139,140,143-149 Ensuring ing pharmacokinetic and nephrologist consultations, ancillary
pharmacodynamic target attainment with high doses can laboratory tests, and additional hospital stay was estimated to
improve patient outcome, but such benefit has definite but be 2-fold higher in patients with nephrotoxicity compared to
unpredictable toxicity trade-offs.150-152 Thus, the risk–benefit those without nephrotoxicity.164
analysis should be performed when clinicians consider these
antibiotics, and close monitoring must occur during the
therapy. Conclusion
Beyond nephrotoxicity, other toxicities associated with
antibiotic accumulation can occur. For example, b-lactam anti- A multitude of factors influence the determination of ‘‘appro-
biotics have been associated with neurotoxicity. Carbapenem- priate antibiotic dosing’’ in critically ill patients with AKI.
induced seizures have been well documented in the literature Many of them motivate clinicians to consider higher antibiotic
in patients with renal insufficiency receiving high doses doses, but toxicity associated with higher doses may also cause
(eg imipenem > 4 g/day).153-157 Neurotoxicity associated a negative patient outcome. Balancing the concerns of ‘‘too
with cefepime in the ICU setting has been increasingly much’’ antibiotic with the compelling pharmacokinetic, phar-
described.158-161 In a prospective study, reduced renal function macodynamic, and antibacterial resistance evidence suggesting
(creatinine clearance < 30 mL/min) was a strong predictor for that higher doses are necessary should initiate intriguing con-
nonconvulsive epilepsy (eg altered mental status, myoclonus) versations among the interdisciplinary team caring for criti-
in patients receiving cefepime.160 Intriguingly, the doses used cally ill patients with AKI. For patients like the case
in these incidents were appropriately adjusted for renal described at the beginning of this article, it is likely best that
function and were not initially linked to the drug accumula- we try to rapidly identify the appropriate antibiotics and then
tion until the trough level confirmed toxicity. These authors use as much of those antibiotics as we can safely, particularly
postulated that other individual pathophysiological variables early in the course of the patient’s sepsis and AKI. Patient mon-
such as plasma albumin and hemoglobin concentrations may itoring needs to be as vigilant as possible to identify antibiotic
influence pharmacokinetic properties, increasing the serum toxicity as early as possible.
cefepime trough levels.160 The authors suggested that unno-
ticed toxicity may contribute to the increased mortality rate Declaration of Conflicting Interests
associated with cefepime therapy versus other b-lactam
The author(s) declared no potential conflicts of interest with respect to
therapy.160 Alternative administration techniques such as the research, authorship, and/or publication of this article.
continuous or prolonged infusion might yield lower peaks
and have an added benefit of reducing the toxicity risk of
b-lactams. Funding
Most studies suggest that antibiotic-induced toxicities are The author(s) received no financial support for the research, authorship,
transient and reversible upon discontinuation of the insulting and/or publication of this article.

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


Lewis and Mueller 9

References 18. Shorr AF, Micek ST, Welch EC, Doherty JA, Reichley RM, Kol-
1. Dellinger RP, Levy MM, Rhodes A, et al. Surviving Sepsis Cam- lef MH. Inappropriate antibiotic therapy in Gram-negative sepsis
paign: international guidelines for management of severe sepsis increases hospital length of stay. Crit Care Med. 2011;39(1):
and septic shock, 2012. Intensive Care Med. 2013;39(2):165-228. 46-51.
2. Uchino S, Kellum JA, Bellomo R, et al. Acute renal failure in cri- 19. Taccone FS, Laterre PF, Dugernier T, et al. Insufficient B-lactam
tically ill patients. JAMA. 2005;294(7):813-818. concentrations in the early phase of severe sepsis and septic
3. Silvester W, Bellomo R, Cole L. Epidemiology, management, and shock. Crit Care. 2010;14(4):R126.
outcome of severe acute renal failure of critical illness in Austra- 20. Seyler L, Cotton F, Taccone FS, et al. Recommended B-lactam
lia. Crit Care Med. 2001;29(10):1910-1915. regimens are inadequate in septic patients treated with continuous
4. Hoste EA, Clermont G, Kersten A, et al. RIFLE criteria for acute renal replacement therapy. Crit Care. 2011;15(3):R137.
kidney injury are associated with hospital mortality in critically ill 21. Udy AA, Varghese JM, Altukroni M, et al. Subtherapeutic initial
patients: a cohort analysis. Crit Care. 2006;10(3):R73. B-lactam concentrations in select critically ill patients. Chest.
5. Waikar SS, Liu KD, Chertow GM. The incidence and prognostic 2012;142(1):30-39.
significance of acute kidney injury. Curr Opin Nephrol Hyper- 22. Binder L, Schwörer H, Hoppe S, et al. Pharmacokinetics of mer-
tens. 2007;16(3):227-236. openem in critically ill patients with severe infections. Ther Drug
6. Bagshaw SM, Uchino S, Kellum JA, et al. Association between Monit. 2013;35(1):63-70.
renal replacement therapy in critically ill patients with severe 23. Stechmiller JK, Treloar D, Allen N. Gut dysfunction in critically
acute kidney injury and mortality. J Crit Care. 2013;28(6): ill patients: a review of the literature. Am J Crit Care. 1997;6(3):
1011-1018. 204-209.
7. Aronoff GR, Aronoff JR. Drug prescribing in kidney disease: 24. Boucher BA, Wood GC, Swanson JM. Pharmacokinetic changes
can’t we do better? Am J Kidney Dis. 2014;63(3):382-383. in critically illness. Crit Care Clin. 2006;22(2):255-271.
8. Farag A, Garg AX, Li L, Jain AK. Dosing errors in prescribed 25. Smith BS, Yogaratnam D, Levasseur-Franklin KE, Forni A, Fong
antibiotics for older persons with CKD: a retrospective time series J. Introduction to drug pharmacokinetics in the critically ill
analysis. Am J Kidney Dis. 2014;63(3):422-428. patient. Chest. 2012;141(5):1327-1336.
9. Micek ST, Welch EC, Khan J, et al. Resistance to empiric 26. Roberts DJ. Drug absorption, distribution, metabolism and excre-
antimicrobial treatment predicts outcome in severe sepsis tion considerations in critically ill adults. Expert Opin Drug
associated with gram-negative bacteremia. J Hosp Med. Metab Toxicol. 2013;9(9):1067-1084.
2011;6(7):405-410. 27. Nguyen NQ, Ng MP, Chapman M, Fraser RJ, Holloway RH. The
10. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo impact of admission diagnosis on gastric emptying in critically ill
J, Pinsky MR. Epidemiology of severe sepsis in the United States: patients. Crit Care. 2007;11(1):R16.
analysis of incidence, outcome, and associated costs of care. Crit 28. Mueller BA, Brierton DG, Abel SR, Bowman L. Effect of enteral
Care Med. 2001;29(7):1303-1310. feeding with Ensure on oral bioavailabilities of ofloxacin and cipro-
11. Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of floxacin. Antimicrob Agents Chemother. 1994;38(9):2101-2105.
sepsis in the United States from 1979 through 2000. N Engl J 29. Mimoz O, Binter V, Jacolot A, et al. Pharmacokinetics and abso-
Med. 2003;348(16):1546-1554. lute bioavailability of ciprofloxacin administered through a naso-
12. Angus DC, Pereira CA, Silva E. Epidemiology of severe sepsis gastric tube with continuous enteral feeding to critically ill
around the world. Endocr Metab Immune Disord Drug Targets. patients. Intensive Care Med. 1998;24(10):1047-1051.
2006;6(2):207-212. 30. Blum RA, D’Andrea DT, Florentino BM, et al. Increased gastric
13. Vincent JL, Rello J, Marshall J, et al. International study of the pH and the bioavailability of fluconazole and ketoconazole. Ann
prevalence and outcomes of infection in intensive care units(EPIC Intern Med. 1991;114(9):755-757.
II study). JAMA. 2009;302(21):2323-2329. 31. Piscitelli SC, Goss TF, Wilton J, D’Andrea DT, Goldstein H,
14. Quenot JP, Binquet C, Kara O, et al. The epidemiology of septic Schentag JJ. Effects of ranitidine and sucralfate on ketocona-
shock in French intensive care units: the prospective multicenter zole bioavailability. Antimicrob Agents Chemother. 1991;
cohort EPISS study. Crit Care. 2013;17(2):R65. 35(9):1765-1771.
15. Kollef MH, Sherman G, Ward S, Fraser VJ. Inadequate antimicro- 32. Chin TWF, Loeb M, Fong IW. Effects of an acidic beverage
bial treatment of infections: a risk factor for hospital mortality (Coca-Cola) on absorption of ketoconazole. Antimicrob Agents
among critically ill patients. Chest. 1999;115(2):462-474. Chemother. 1995;39(8):1671-1675.
16. Ibrahim EH, Sherman G, Ward S, Fraser VJ, Kollef MH. The 33. Dörffler-Melly J, de Jonge E, Pont AC, et al. Bioavailability of
influence of inadequate antimicrobial treatment of bloodstream subcutaneous low-molecular- weight heparin to patients on vaso-
infections on patient outcomes in the ICU setting. Chest. 2000; pressors. Lancet. 2002;359(9309):849-850.
118(1):146-155. 34. Haas CE, Nelsen JL, Raghavendran K, et al. Pharmacokinetics
17. Harbarth S, Garbino J, Pugin J, Romand JA, Lew D, Pittet D. and pharmacodynamics of enoxaparin in multiple trauma patients.
Inappropriate initial antimicrobial therapy and its effect on sur- J Trauma. 2005;59(6):1336-1343.
vival in a clinical trial of immunomodulating therapy for severe 35. Parrillo JE. Pathogenetic mechanisms of septic shock. N Engl J
sepsis. Am J Med. 2003;115(7):529-535. Med. 1993;328(20):1471-1477.

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


10 Journal of Intensive Care Medicine

36. Triginer C, Izquierdo I, Fernandez R, et al. Gentamicin volume of Syndrome Network. Fluid balance, diuretic use, and mortality in
distribution in critically ill septic patients. Intensive Care Med. acute kidney injury. Clin J Am Soc Nephrol. 2011;6(5):966-973.
1990;16(5):303-306. 53. Teixeira C, Garzotto F, Piccinni P, et al. Fluid balance and urine
37. Gous AG, Dance MD, Lipman J, Luyt DK, Mathivha R, Scribante volume are independent predictors of mortality in acute kidney
J. Changes in vancomycin pharmacokinetics in critically ill injury. Crit Care. 2013;17(1):R14.
infants. Anaesth Intensive Care. 1995;23(6):678-682. 54. SAFE Study Investigators. Finfer S, Bellomo R, McEvoy S, et al.
38. Tang GJ, Tang JJ, Lin BS, Kong CW, Lee TY. Factors affecting Effect of baseline serum albumin concentration on outcome of
gentamicin pharmacokinetics in septic patients. Acta Anaesthesiol resuscitation with albumin or saline in patients in intensive care
Scand. 1999;43(7):726-730. units: analysis of data from the Saline versus Albumin Fluid Eva-
39. Buijk SE, Mouton JW, Gyssens IC, Verbrugh HA, Bruining HA. luation (SAFE) study. BMJ. 2006;333(7577):1044.
Experience with a once- daily dosing program of aminoglycosides 55. Roberts JA, Federico P, Lipman J. The clinical relevance of
in critically ill patients. Intensive Care Med. 2002;28(7):936-942. plasma protein binding changes. Clin Pharmacokinet. 2013;
40. Del Mar Fernandez de Gatta Garcia M, Revilla N, Calvo MV, 52(1):1-8.
Dominquez-Gil A, Sanchez Navarro A. Pharmacokinetic/pharma- 56. Burkhardt O, Kumar V, Katterwe D, et al. Ertapenem in critically
codynamics analysis of vancomycin in ICU patients. Intensive ill patients with early-onset ventilator-associated pneumonia:
Care Med. 2007;33(2):279-285. pharmacokinetics with special consideration of free-drug concen-
41. Taccone FS, Hites M, Beumier M, Scolletta S, Jacobs F. Appro- tration. J Antimicrob Chemother. 2007;59(2):277-284.
priate antibiotic dosage levels in the treatment of severe sepsis 57. Eyler RF, Vilay AM, Nader AM, et al. Pharmacokinetics of erta-
and septic shock. Curr Infect Dis Rep. 2011;13(5):406-415. penem in critically ill patients receiving continuous venovenous
42. Vilay AM, Grio M, DePestel DD, et al. Daptomycin pharmacoki- hemodialysis or hemodiafiltration. Antimicrob Agents Che-
netics in critically ill patients receiving continuous venovenous mother. 2014;58(3):1320-1326.
hemodialysis. Crit Care Med. 2011;39(1):19-25. 58. Andreasen F. The effect of dialysis on the protein binding of drugs
43. Wenisch JM, Meyer B, Fuhrmann V, et al. Multiple-dose pharma- in the plasma of patients with acute renal failure. Acta Pharmacol
cokinetics of daptomycin during continuous venovenous haemo- Toxicol. 1974;34(4):284-294.
diafiltration. J Antimicrob Chemother. 2012;67(4):977-983. 59. Mussche MM, Belpaire FM, Bogaert MG. Plasma protein binding
44. Bauer SR, Salem C, Connor MJ Jr, et al. Pharmacokinetics and of phenylbutazone during recovery from acute renal failure. Eur J
pharmacodynamics of piperacillin-tazobactam in 42 patients Clin Pharmacol. 1975;9(1):69-71.
treated with concomitant CRRT. Clin J Am Soc Nephrol. 2012; 60. Vilay AM, Churchwell MD, Mueller BA. Clinical review: drug
7(3):452-457. metabolism and nonrenal clearance in acute kidney injury. Crit
45. Khadzhynov D, Slowinski T, Lieker I, et al. Plasma pharmacoki- Care. 2008;12(6):235.
netics of daptomycin in critically ill patients with renal failure and 61. Lalande L, Charpiat B, Leboucher G, Tod M. Consequences of
undergoing CVVHD. Int J Clin Pharmacol Ther. 2011;49(11): renal failure on non-renal clearance of drugs. Clin Pharmacoki-
656-665. net. 2014;53(6):521-532.
46. Dvorchik B, Arbeit RD, Chung J, Liu S, Knebel W, Kastrissios H. 62. Kirwan CJ, MacPhee IA, Lee T, Holt DW, Philips BJ. Acute kid-
Population pharmacokinetics of daptomycin. Antimicrob Agents ney injury reduces the hepatic metabolism of midazolam in criti-
Chemother. 2004;48(8):2799-2807. cally ill patients. Intensive Care Med. 2012;38(1):76-84.
47. Botha FJ, van der BIJL P, Seifart HI, Parkin DP. Fluctuation of 63. Sun H, Frassetto L, Benet LZ. Effects of renal failure on drug
the volume of distribution of amikacin and its effect on once- transport and metabolism. Pharmacol Ther. 2006;109(1-2):1-11.
daily dosage and clearance in a seriously ill patient. Intensive 64. Jones EM, McMullin CM, Hedges AJ, et al. The pharmacokinetics
Care Med. 1996;22(5):443-446. of intravenous ciprofloxacin 400 mg 12 hourly in patients with
48. Tormo C, Abad FJ, Ronchera-Oms CL, Parra V, Jimenez NV. severe sepsis: the effect of renal function and intra-abdominal dis-
Critically-ill patients receiving total parenteral nutrition show ease. J Antimicrob Chemother. 1997;40(1):121-124.
altered amikacin pharmacokinetics. Clinical Nutrition. 1995; 65. Macias WL, Mueller BA, Scarim SK. Vancomycin pharmacoki-
14(4):254-259. netics in acute renal failure: preservation of nonrenal clearance.
49. Ronchera-Oms CL, Tormo C, Ordovas JP, Abad J, Jimenez NV. Clin Pharmacol Ther. 1991;18(4):451-458.
Expanded gentamicin volume of distribution in critically ill adult 66. Mueller BA, Scarim SK, Macias WL. Comparison of imipenem
patients receiving total parenteral nutrition. J Clin Pharm Ther. pharmacokinetics in patients with acute or chronic renal failure
1995;20(5):253-258. treated with continuous hemofiltration. Am J Kidney Dis. 1993;
50. Payen D, de Pont AC, Sakr Y, et al. A positive fluid balance is 21(2):172-179.
associated with a worse outcome in patients with acute renal fail- 67. Tegeder I, Bremer F, Oelkers R, et al. Pharmacokinetics of
ure. Crit Care. 2008;12(3):R74. imipenem-cilastatin in critically ill patients undergoing continu-
51. Bouchard J, Soroko SB, Chertow GM, et al. Fluid accumulation, ous venovenous hemofiltration. Antimicrob Agents Chemother.
survival and recovery of kidney function in critically ill patients 1997;41(12):2540-2545.
with acute kidney injury. Kidney Int. 2009;76(4):422-427. 68. Golper TA, Noonan HM, Elzinga L, et al. Vancomycin pharma-
52. Grams ME, Estrella MM, Coresh J, Brower RG, Liu KD; National cokinetics, renal handling, and nonrenal clearances in normal
Heart, Lung, and Blood Institute Acute Respiratory Distress human patients. Clin Pharmacol Ther. 1988;43(5):565-570.

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


Lewis and Mueller 11

69. Brunet S, Leblanc M, Geadah D, Parent D, Courteau S, Cardinal J. patients with acute renal failure. Crit Care Med. 2004;32(12):
Diffusive and convective solute clearances during continuous 2437-2442.
renal replacement therapy at various dialysate and ultrafiltration 86. Arzuaga A, Maynar J, Gascón AR, et al. Influence of renal func-
flow rates. Am J Kidney Dis. 1999;34(3):886-892. tion on the pharmacokinetics of piperacillin/tazobactam in inten-
70. Churchwell MD, Mueller BA. Drug dosing during continuous sive care unit patients during continuous venovenous
renal replacement therapy. Semin Dial. 2009;22(2):185-188. hemofiltration. J Clin Pharmacol. 2005;45(2):168-176.
71. Pasko DA, Churchwell MD, Salama NN, Mueller BA. Longitudi- 87. Kielstein JT, Czock D, Schopke T, et al. Pharmacokinetics and
nal hemodiafilter performance in modeled continuous renal total elimination of meropenem and vancomycin in intensive
replacement therapy. Blood Purif. 2011;32(2):82-88. care unit patients undergoing extended daily dialysis. Crit Care
72. Choi G, Gomersall CD, Tian Q, Joynt GM, Freebairn R, Lipman Med. 2006;34(1):51-56.
J. Principles of antibacterial dosing in continuous renal replace- 88. Burkhardt O, Joukhadar C, Traunmuller F, Hadem J, Welte T,
ment therapy. Crit Care Med. 2009;37(7):2268-2282. Kielstein JT. Elimination of daptomycin in a patient with acute
73. Heintz BH, Matzke GR, Dager WE. Antimicrobial dosing con- renal failure undergoing extended daily dialysis. J Antimicrob
cepts and recommendations for critically ill adult patients receiv- Chemother. 2008;61(1):224-225.
ing continuous renal replacement therapy or intermittent 89. Wilson FP, Berns JS. Vancomycin levels are frequently subther-
hemodialysis. Pharmacotherapy. 2009;29(5):562-577. apeutic during continuous venovenous hemodialysis (CVVHD).
74. Eyler RF, Mueller BA. Antibiotic dosing in critically ill patients Clin Nephrol. 2012;77(4):329-331.
with acute kidney injury. Nat Rev Nephrol. 2011;7(4):226-235. 90. Zoller M, Maier B, Hornuss C, et al. Variability of linezolid con-
75. Kielstein JT, Burkhardt O. Dosing of antibiotics in critically ill centrations after standard dosing in critically ill patients: a pro-
patients undergoing renal replacement therapy. Curr Pharm Bio- spective observational study. Crit Care. 2014;18(4):R148.
technol. 2011;12(12):2015-2019. 91. Owens RC, Shorr AF. Rational dosing of antimicrobial agents:
76. Bogard KN, Peterson NT, Plumb TJ, Erwin MW, Fuller PD, pharmacokinetic and pharmacodynamics strategies. Am J
Olsen KM. Antibiotic dosing during sustained low-efficiency dia- Health-Sys Pharm. 2009;66(4):S23-S30.
lysis: special consideration in adult critically ill patients. Crit 92. Mariat C, Venet C, Jehl F, et al. Continuous infusion of ceftazi-
Care Med. 2011;39(3):560-570. dime in critically ill patients undergoing continuous venovenous
77. Scoville BA, Mueller BA. Medication dosing in critically ill haemodiafiltration: pharmacokinetic evaluation and dose recom-
patients with acute kidney injury with renal replacement therapy. mendation. Crit Care. 2006;10(1):R26.
Am J Kideny Dis. 2013;61(3):490-500. 93. Langgartner J, Vasold A, Glück T, Reng M, Kees F. Pharma-
78. Mueller BA, Smoyer WE. Challenges in developing evidence- cokinetics of meropenem during intermittent and continuous
based drug dosing guidelines for adults and children receiving intravenous application in patients treated by continuous
renal replacement therapy. Clin Pharmacol Ther. 2009;86(5): renal replacement therapy. Intensive Care Med. 2008;34(6):
479-482. 1091-1096.
79. Vesconi S, Cruz DN, Fumagalli R, et al. DOse REsponse Multi- 94. Saugel B, Nowack MC, Hapfelmeier A, et al. Continuous intra-
centre International collaborative Initiative (DO-RE-MI Study venous administration of vancomycin in medical intensive care
Group): delivered dose of renal replacement therapy and mortality unit patients. J Crit Care. 2013;28(1):9-13.
in critically ill patients with acute kidney injury. Crit Care. 2009; 95. Beumier M, Roberts JA, Kabtouri H, et al. A new regimen for
13(2):R57. continuous infusion of vancomycin during continuous renal
80. VA/NIH Acute Renal Failure Trial Network, Palevsky PM, Zhang replacement therapy. J Antimicrob Chemother. 2013;68(12):
JH, O’Connor TZ, et al. Intensity of renal support in critically ill 2859-2865.
patients with acute kidney injury. N Engl J Med. 2008;359(1): 96. Lodise TP, Lomaestro B, Drusano G. Piperacillin-tazobactam
7-20. for Pseudomonas aeruginosa infection: clinical implications of
81. RENAL Replacement Therapy Study Investigators, Bellomo R, an extended infusion dosing strategy. Clin Infect Dis. 2007;
Cass A, Cole L, et al. Intensity of continuous renal replacement 44(3):357-363.
therapy in critically ill patients. N Eng J Med. 2009;361(17): 97. Kielstein JT, Engbers C, Bode-Boeger SM, et al. Dosing of dap-
1627-1638. tomycin in intensive care unit patients with acute kidney injury
82. Kielstein JT, David S. Pro: renal replacement trauma or paracel- undergoing extended dialysis-a pharmacokinetic study. Nephrol
sus 2.0. Nephrol Dial Transplant. 2013;28(11):2728-2733. Dial Transplant. 2010;25(5):1537-1541.
83. Covajes C, Scolletta S, Penaccini L, et al. Continuous infusion of 98. Mueller BA, Scoville BA. Adding to the armamentarium: anti-
vancomycin in septic patients receiving continuous renal replace- biotic dosing in extended dialysis. Clin J Am Soc Nephrol.
ment therapy. Int J Antimicrob Agents. 2013;41(3):261-266. 2012;7(3):373-375.
84. Roberts JA, Mehta RL, Lipman J. Sustained low efficiency dialy- 99. Taccone FS, de Backer D, Laterre PF, et al. Pharmacokinetics of
sis allows rational renal replacement therapy, but dose it allow a loading dose of amikacin in septic patients undergoing contin-
rational drug dosing? Crit Care Med. 2011;39(3):602-603. uous renal replacement therapy. Int J Antimicrob Agents. 2011;
85. Fiaccadori E, Maggiore U, Rotelli C, et al. Removal of linezolid 37(6):5.
by conventional intermittent hemodialysis, sustained low- 100. Truong J, LEvkovich BJ, Padiglione AA. Simple approach to
efficiency dialysis, or continuous venovenous hemofiltration in improving vancomycin dosing in intensive care: a standardized

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


12 Journal of Intensive Care Medicine

loading dose results in earlier therapeutic levels. Intern Med J. 114. Moore RD, Lietman PS, Smith CR. Clinical response to amino-
2012;42(1):23-29. glycoside therapy: importance of the ratio of peak concentration
101. Kamel Mohamed OH, Wahba IM, Watnick S, et al. Administra- to minimal inhibitory concentration. J Infect Dis. 1987;155(1):
tion of tobramycin in the beginning of the hemodialysis session: 93-99.
a novel intradialytic dosing regimen. Clin J Am Soc Nephrol. 115. Martinez MN, Papich MG, Drusano GL. Dosing regimen mat-
2007;2(4):694-699. ters: the importance of early intervention and rapid attainment
102. Sowinski KM, Magner SJ, Lucksiri A, Scott MK, Hamburger of the pharmacokinetic/pharmacodynamic target. Antimicrob
RJ, Mueller BA. Influence of hemodialysis on gentamicin phar- Agents Chemother. 2012;56(6):2795-2805.
macokinetics, removal during hemodialysis, and recommended 116. Jumbe N, Louie A, Leary R, et al. Application of a mathematical
dosing. Clin J Am Soc Nephrol. 2008;3(2):355-361. model to prevent in vivo amplification of antibiotic-resistant
103. Veistein A, Venisse N, Badin J, Pinsard M, Robert R, Dupuis A. bacterial populations during therapy. J Clin Invest. 2003;
Gentamicin in hemodialyzed critical care patients: early dialysis 112(2):275-285.
after administration of a high dose should be considered. Antimi- 117. Trotman RL, William JC, Shoemaker DM, Salzer WL. Anti-
crob Agents Chemother. 2013;57(2):977-982. biotic dosing in critically ill adult patients receiving continuous
104. Roberts JA, Field J, Visser A, et al. Using population phar- renal replacement therapy. Clin Infect Dis. 2005;41(8):
macokinetics to determine gentamicin dosing during 1159-1166.
extended daily diafiltration in critically ill patients with 118. Matsuo H, Hayashi J, Ono K, et al. Administration of aminogly-
acute kidney injury. Antimicrob Agents Chemother. 2010; cosides to hemodialysis patients immediately before dialysis: a
54(9):3635-3640. new dosing modality. Antimicrob Agents Chemother. 1997;
105. Calbo E, Garau J. Application of pharmacokinetics and phar- 41(12):2597-2601.
macodynamics to antimicrobial therapy of community 119. Teigen MM, Duffull S, Dang L, Johnson DW. Dosing
acquired respiratory tract infections. Respiration. 2005;72(6): of gentamicin in patients with end-stage renal disease
561-571. receiving hemodialysis. J Clin Pharmacol. 2006;46(11):
106. Scaglione F, Paraboni L. Influence of pharmacokinetics/pharma- 1259-1267.
codynamics of antibacterials in their dosing regimen selection. 120. Gentamicin [package insert]. Lake Forest, IL: Hospira, Inc;
Expert Rev Anti Infect Ther. 2006;4(3):479-490. August 2005. http://www.hospira.com/Images/EN-1004_32-
107. McKinnon PS, Paladino JA, Schentag JJ. Evaluation of area 5467_1.pdf. Accessed March 12, 2014.
under the inhibitory curve (AUIC) and time above the minimum 121. Ryan DM, Cars O. A problem in the interpretation of beta-
inhibitory concentration (T > MIC) as predictors of outcome for lactam antibiotic levels in tissues. J Antimicrob Chemother.
cefepime and ceftazidime in serious bacterial infections. Int J 1983;12(3):281-284.
Antimicrob Agents. 2008;31:345-351. 122. Schentag JJ, Heller AS, Hardy BG, Wels PB. Antibiotic penetra-
108. Valtonen M, Tiula, Takkunen O, Backman JT, Neuvonen PJ. tion in liver infection: a case of tobramycin failure responsive to
Elimination of the piperacillin/tazobactam combination during moxalactam. Am J Gastroenterol. 1983;78(10):641-644.
continuous venovenous haemofiltration and haemodiafiltration 123. Joukhadar C, Frossard M, Mayer BX, et al. Impaired target site
in patients with acute renal failure. J Antimicrob Chemother. penetration of beta-lactams may account for therapeutic failure
2001;48(6):881-885. in patients with septic shock. Crit Care Med. 2001;29(2):
109. Falagas ME, Tansarli GS, Ikawa K, Vardakas KZ. Clinical out- 385-391.
comes with extended or continuous versus short-term intrave- 124. Zeitlinger MA, Erovic BM, Sauermann R, Georgopoulos A,
nous infusion of carbapenems and piperacillin/tazobactam: a Müller M, Joukhadar C. Plasma concentrations might lead
systematic review and meta-analysis. Clin Infec Dis. 2013; to overestimation of target site activity of piperacillin in
56(2):272-282. patients with sepsis. J Antimicrob Chemother. 2005;56(4):
110. Chant C, Leung A, Friedrich JO. Optimal dosing of antibio- 703-708.
tics in critically ill patients using continuous/extended infu- 125. Klekner A, Bagyi K, Bognar L, Gaspar A, Andrasi M, Szabo J.
sion: a systematic review and meta-analysis. Crit Care. Effectiveness of cephalosporins in the sputum of patients with
2013;17(6):R279. nosocomial bronchopneumonia. J Clin Microbiol. 2006;44(9):
111. Moriyama B, Henning SA, Neuhauser MM, Danner RL, Walsh TJ. 3418-3421.
Continuous-infusion beta-lactam antibiotics during continuous 126. Stolle LB, Plock N, Joukhadar C, et al. Pharmacokinetics of line-
venovenous hemofiltration for the treatment of resistant gram- zolid in bone tissue investigated by in vivo microdialysis. Scand
negative bacteria. Ann Pharmacother. 2009;43(7):1324-1337. J Infect Dis. 2008;40(1):24-29.
112. Joy MS, Matzke GR, Frye RF, Palevsky PM. Determinants of 127. Tam VH, Gamez EA, Weston JS, et al. Outcomes of bacter-
vancomycin clearance by continuous venovenous hemofiltration emia due to Pseudomonas aeruginosa with reduced suscept-
and continuous venovenous hemodialysis. Am J Kidney Dis. ibility to piperacillin-tazobactam: implications on the
1998;31(6):1019-1027. appropriateness of the resistance breakpoint. Clin Infect Dis.
113. DelDot ME, Lipman J, Tett SE. Vancomycin pharmacokinetics 2008;46(6):862-867.
in critically ill patients receiving continuous venovenous haemo- 128. Soriano A, Marco F, Martı́nez JA, et al. Influence of vancomycin
diafiltration. Br J Clin Pharmacol. 2004;58(3):259-268. minimum inhibitory concentration on the treatment of

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


Lewis and Mueller 13

methicillin-resistant Staphylococcus aureus bacteremia. Clin dosing schedules that maintain troughs between 15 and 20 milli-
Infect Dis. 2008;46(2):193-200. grams per liter. Antimicrob Agents Chemother. 2013;57(2):
129. Hidayat LK, Hsu DI, Quist R, Shriner KA, Wong-Beringer A. 734-744.
High-dose vancomycin therapy for methicillin-resistant Staphy- 144. Moore RD, Smith CR, Lipsky JJ, Mellits ED, Lietman PS. Risk
lococcus aureus infections: efficacy and toxicity. Arch Intern factors for nephrotoxicity in patients treated with aminoglyco-
Med. 2006;166(19):2138-2144. sides. Ann Intern Med. 1984;100(3):352-357.
130. Rybak MJ, Lomaestro BM, Rotschafer JC, et al. Vancomycin 145. Lodise TP, Lomaestro B, Graves J, Drusano GL. Larger vanco-
therapeutic guidelines: a summary of consensus recommenda- mycin doses (at least four grams per day) are associated with an
tions from the Infectious Diseases Society of America, the increased incidence of nephrotoxicity. Antimicrob Agents Che-
American Society of Health-System Pharmacists, and the Soci- mother. 2008;52(4):1330-1336.
ety of Infectious Diseases Pharmacist. Clin Infect Dis. 2009; 146. Lodise TP, Patel N, Lomaestro BM, Rodvold KA, Drusano
49(3):325-327. GL. Relationship between initial vancomycin concentration
131. Kullar R, McClellan I, Geriak M, Sakoulas G. Efficacy and time profile and nephrotoxicity. Clin Infect Dis. 2009;49(4):
safety of daptomycin in patients with renal impairment: a multi- 507-514.
center retrospective analysis. Pharmacotherapy. 2014;34(6): 147. Pritchard L, Baker C, Leggett J, Sehdev P, Brown A, Bayley KB.
582-589. Increasing vancomycin serum trough concentrations and inci-
132. National Nosocomial Infections Surveillance System. dence of nephrotoxicity. Am J Med. 2010;123(12):1143-1149.
National Nosocomial Infections Surveillance (NNIS) System 148. Bosso JA, Nappi J, Rudisill C, et al. Relationship between van-
Report, data summary from January 1992 through June comycin trough concentrations and nephrotoxicity: a prospec-
2004, issued October 2004. Am J Infect Control. 2004; tive multicenter trial. Antimicrob Agents Chemother. 2011;
32(6):470-485. 55(12):5475-5479.
133. Klevens RM, Edwards JR, Gaynes RP. The impact of 149. Cano EL, Haque NZ, Welch VL, et al. Incidence of Nephrotoxi-
antimicrobial-resistant, health care- associated infections on city and Association With Vancomycin Use in Intensive Care
mortality in the United States. Clin Infect Dis. 2008;47(7): Unit Patients With Pneumonia: Retrospective Analysis of the
927-930. IMPACT-HAP Database. Clin Ther. 2012;34(1):149-157.
134. Boucher HW, Talbot GH, Bradley JS, et al. Bad bugs, no drugs: 150. Bertino JS, Booker LA, Franck PA, Jenkins PL, Franck KR, Naf-
no ESKAPE! An update from the Infectious Diseases Society of ziger AN. Incidence of and significant risk factors for
America. Clin Infect Dis. 2009;48(1):1-12. aminoglycoside-associated nephrotoxicity in patients dosed by
135. Peters NK, Dixon DM, Holland SM, Fauci AS. The research using individualized pharmacokinetic monitoring. J Infect Dis.
agenda of the national institute of allergy and infectious diseases 1993;167(1):173-179.
for antimicrobial resistance. J Infect Dis. 2008;197(8): 151. Rybak MJ, Abate BJ, Kang SL, Ruffing MJ, Lerner SA, Drusano
1087-1093. GL. Prospective evaluation of the effect of an aminoglycoside
136. Barth RH, DeVincenzo N. Use of vancomycin in high-flux dosing regimen on rates of observed nephrotoxicity and ototoxi-
hemodialysis: experience with 130 courses of therapy. Kidney city. Antimicrob Agents Chemother. 1999;43(7):1549-1555.
Int. 1996;50(3):929-936. 152. Drusano GL, Ambrose PG, Bhavnani SM, Bertino JS, Nafziger
137. Taber SS, Mueller BA. Drug-associated renal dysfunction. Crit AN, Louie A. Back to the future: using aminoglycosides again
Care Clin. 2006;22(2)357-374. and how to dose them optimally. Clin Infect Dis. 2007;45(15):
138. Kane-Gill SL, Kirisci L, Verrico MM, Rothschild JM. Analysis 753-760.
of risk factors for adverse drug events in critically ill patients. 153. Calandra GB, Brown KR, Grad LC, Ahonkhai VI, Wang C, Aziz
Crit Care Med. 2012;40(3):823-828. MA. Review of adverse experiences and tolerability in the first
139. Hartzell JD, Neff R, Ake J, et al. Nephrotoxicity associated with 2516 patients treated with imipenem/cilastatin. Am J Med.
intravenous colistin (colistin sodium) treatment at a tertiary care 1985;78(6A):73-78.
medical center. Clin Infect Dis. 2009;48(12):1724-1728. 154. Hellinger WC, Brewer NS. Carbapenems and monobactams:
140. Kwon JA, Lee JE, Huh W, et al. Predictors of acute kidney injury imipenem, meropenem and aztreonam. Mayo Clin Proc. 1999;
associated with intravenous colistin treatment. Int J Antimicrob 74(4):420-434.
Agents. 2010;35(5):473-477. 155. Norrby SR. Carbapenems in serious infections: a risk benefit
141. Schentag JJ, Cerra FB, Plaut ME. Clinical and pharmacokinetic assessment. Drug Saf. 2000;22(3):191-194.
characteristics of aminoglycoside nephrotoxicity in 201 criti- 156. Norrby SR, Newell PA, Faukner KL, Lesky W. Safety profile of
cally ill patients. Antimicrob Agents Chemother. 1982;21(5): meropenem international clinical experience based on the first
721-726. 3125 patients treated with meropenem. J Antimicrob Chemother.
142. Zager RA. Endotoxemia, renal hypoperfusion, and fever: inter- 2005;36(suppl A):207-223.
active risk factors for aminoglycoside and sepsis associated 157. Nicolau DP. Carbapenems: a potent class of antibiotics. Expert
acute renal failure. Am J Kid Dis. 1992;20(3):223-230. Opin Pharmacother. 2008;9(1):23-37.
143. Van Hal, Paterson DL, Lodise TP. Systematic review and meta- 158. Martinez-Rodriguez JE, Barriga FJ, Santamaria J, et al.
analysis of vancomycin- induced nephrotoxicity associated with Nonconvulsive status epilepticus associated with

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


14 Journal of Intensive Care Medicine

cephalosporins in patients with renal failure. Am J Med. unit: a cause of severe, underappreciated encephalopathy. Crit
2001;111(2):115-119. Care. 2013;17(6):R264.
159. Grill MF, Maganti R. Cephalosporin-induced neurotoxicity: 162. Bagshaw SM. Short- and long-term survival after acute
clinical manifestations, potential pathogenic mechanisms, and kidney injury. Nephrol Dial Transplant. 2008;23(7):
the role of electroencephalographic monitoring. Ann Pharmac- 2126-2128.
other. 2008;42(12):1843-1850. 163. Schiffl H, Fischer R. Five-year outcomes of severe acute kidney
160. Chapuis TM, Giannoni E, Majcherczyk PA, et al. Prospective injury requiring renal replacement therapy. Nephrol Dial Trans-
monitoring of cefepime in intensive care unit adult patients. Crit plant. 2008;23(7):2235-2241.
Care. 2010;14(2):R51. 164. Eisenberg JM, Koffer H, Glick HA, et al. What is the cost of
161. Fugate JE, Kalimullah EA, Hocker SE, Clark SL, Wijdicks EF, nephrotoxicity associated with aminoglycosides? Ann Intern
Rabinstein AA. Cefepime neurotoxicity in the intensive care Med. 1987;107(6):900-909.

Downloaded from jic.sagepub.com at UNIV OF MICHIGAN on December 2, 2014


View publication stats

You might also like