Dose Atbc Ir 2016

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Analytic Review

Journal of Intensive Care Medicine


2016, Vol. 31(3) 164-176
Antibiotic Dosing in Patients With Acute ª The Author(s) 2014
Reprints and permission:
sagepub.com/journalsPermissions.nav
Kidney Injury: ‘‘Enough But Not Too Much’’ DOI: 10.1177/0885066614555490
jic.sagepub.com

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 The Surviving Sepsis Guidelines emphasize the significance


of ‘‘appropriate antimicrobial therapy,’’ which is defined as
A 68-year-old female (height 5 ft 2 in, weight 95 kg) was
‘‘initial empiric anti-infective therapy of one or more drugs that
admitted for complaint of increasing abdominal pain and have activity against all likely pathogens and that penetrate in
altered mental status. Her past medical history is extensive
adequate concentrations into tissue presumed to be the source
including recurrent urosepsis caused by b-lactamase-positive
of sepsis.’’1 However, even in the straightforward case men-
Escherichia coli. In the intensive care unit (ICU), she has not
tioned earlier, numerous barriers exist to achieve ‘‘adequate
yet received empiric antibiotic therapy but did get aggressive
concentrations’’ at the infection site. Recent evidence suggests
fluid resuscitation in the emergency department. The patient’s
that clinicians prescribe antibiotics with doses that are higher
oxygen saturation dropped rapidly and she developed respira-
than what is recommended for the patients’ glomerular filtra-
tory failure and septic shock, requiring mechanical ventilation
tion rate in elderly patients with chronic kidney disease stages
with oxygen and vasopressor support. The laboratory results IV and V,7,8 increasing the risk of antibiotic toxicity. However,
include serum creatinine (SCr) 2.2 mg/dL and blood urea nitro-
in critically ill patients with AKI, higher doses may be war-
gen (BUN) 45 mg/dL (baseline SCr 1.1 mg/dL, BUN 23 mg/dL
ranted. Appropriate antibiotic therapy requires a balance
1 month ago). The patient’s weight in the nursing home chart 1
between a need for doses high enough to account for pharma-
week ago was 80 kg. Susceptibility results are pending. The
cokinetic alterations in these patients, pathogen resistance that
ICU team needs to prescribe antibiotic therapy.
is common in the ICU,9 and extracorporeal drug clearance ver-
This complicated scenario is not unlike those faced regu-
sus concerns of antibiotic toxicity in these vulnerable patients
larly by clinicians. The Surviving Sepsis Guidelines provide
with impaired drug clearance (Figure 1). The purpose of this
very straightforward guidance on how to treat patients with article is to explore the challenges of providing appropriate
sepsis,1 yet the patient in the previous case comes with some
challenges that are not readily apparent. Much of the dosing
of her antibiotics is predicated on knowledge of her renal func-
tion, and it is apparent in this case that she has acute kidney 1
Department of Clinical, Social, and Administrative Sciences, University of
injury (AKI), which is a common complication of sepsis in the Michigan College of Pharmacy, Ann Arbor, MI, USA
ICU2 and is associated with high morbidity and mortality Received March 13, 2014, and in revised form August 4, 2014. Accepted
(60%-77%).3-5 About 70% of patients with AKI in the ICU for publication August 25, 2014.
require some type of renal replacement therapy (RRT),6 and
with the degree of fluid overload manifested by this patient, Corresponding Author:
Bruce A. Mueller, Department of Clinical, Social and Administrative Sciences,
it is likely that RRT will be necessary as diuretics and conser-
University of Michigan College of Pharmacy, 428 Church St, Ann Arbor, MI
vative management are unlikely to be able to remove 15 L of 48109, USA.
extra volume in a timely manner. Email: muellerb@med.umich.edu
Lewis and Mueller 165

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
other drugs, resulting in reduced bioavailability.28,29 Elevated
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
usually altered, but absorption of subcutaneous drugs may be
affected by impaired peripheral circulation by sepsis and use of
Figure 1. Enough but not too much. The clinician’s decision to
vasopressors and peripheral edema during intravenous fluid
choose higher versus lower antibiotic doses in critically ill patients
with acute kidney disease requires balancing often conflicting factors. resuscitation, reducing their bioavailabilities.33,34 Fortunately,
antibiotics are not administered by this route obviating this con-
cern, although many other drugs in the ICU are administered sub-
antibiotic doses to attain adequate concentrations at the site of cutaneously (heparins, insulin, etc).
infection and to suggest alternative strategies balancing neces-
sity and risk of antibiotic higher doses to maximize antibiotic
efficacy while minimizing toxicity. Distribution
The pathogenesis of sepsis involves endothelial abnormalities,
increase in capillary permeability, and the fluid accumulation in
Mortality Related to Inappropriate Antibiotic
interstitial spaces, resulting in increased antibiotic volume of
Dosing distribution.35 Expanded volume of distribution may be espe-
Mortality rates in critically ill patients with sepsis reported cially prominent with hydrophilic drugs such as aminoglycosides,
from various regions around the world range from 20% to b-lactams, glycopeptides, and daptomycin. Many researchers
60%.10-14 In the United States, severe sepsis and septic shock have demonstrated decreased serum levels of antibiotics in criti-
account for approximately 10% of all mortality, representing cally ill patients secondary to increased volume of distribution,
a substantial health care burden.10 Consequently, appropriate suggesting the need for higher doses than standard dosing recom-
antibiotic therapy is essential for both humanistic and eco- mendations to maintain suitable serum concentrations.22,36-44 The
nomic reasons. However, available data suggest that inap- increased volume of distribution is more pronounced in severe
propriate antibiotic therapy in patients with sepsis in the ICU sepsis and in the early stages of sepsis. Patients with hyperdy-
is prevalent and is the most important independent risk factor namic sepsis have a significantly larger volume of distribution
for hospital mortality.15-18 The primary reasons for inap- of gentamicin (0.48 L/kg), compared with those who have hypo-
propriate antibiotic therapy have been identified as antibiotic dynamic sepsis (0.32 L/kg) and nonseptic patients receiving post-
resistance and absence of efficacious therapy in most surgical prophylactic doses of gentamicin (0.29 L/kg).38 Taccone
studies.15 However, attention should be paid to the other evi- et al noted that the first conventional dose of broad-spectrum b-
dence showing that even in the case where pathogens are sus- lactams administered in the early stage of severe sepsis and septic
ceptible to the prescribed antibiotics, hospital mortality still shock failed to achieve adequate concentrations due to increased
remains high.9 It is not enough to select the appropriate anti- volume of distribution.19 Significantly elevated volume of distri-
biotic, but one must also prescribe doses sufficient to deliver suf- bution is also observed in critically ill patients with AKI receiving
ficient antibiotic concentrations at the site of infection. continuous RRT (CRRT) after the administration of the first dose
Conventional antibiotic dosing may not yield pharmacodynamic of daptomycin which possesses both hydrophilic and lipophilic
target attainment for the infecting pathogens in this patient pop- properties (0.23 L/kg42,45 vs 0.08-0.15 L/kg in healthy volun-
ulation.19-22 Many factors influence antibiotic concentrations at teers46). However, the initially elevated volume of distribution
the infection site, beginning with the numerous pharmacokinetic tends to decline over the treatment course36,45 possibly due to
changes seen in critically ill patients. physiologic changes or correction of fluid overload by RRTs.
166 Journal of Intensive Care Medicine 31(3)

Taken together, the increased volume of distribution of hydrophi- L/kg) and higher total clearance (43 mL/min vs 20 mL/min) in
lic antibiotics in the early stage of sepsis or severe sepsis necessi- patients with hypoalbuminemia and sepsis, compared with
tates a higher dose or a loading dose to rapidly achieve adequate healthy individuals.56,57 Pharmacokinetic changes in this magni-
concentrations to meet Surviving Sepsis Guidelines. However, tude should result in dosing increases to maintain therapeutic
increased volume of distribution will normalize after a few days ertapenem plasma concentrations. However, as renal function
of treatment, therefore, clinicians may need to move to more con- improves, gradual normalization of protein-binding capacity is
ventional dosing over the course of therapy to avoid toxicity. Not observed in patients with AKI.58,59
only can pathophysiologic changes related to sepsis influence vol-
ume of distribution in critically ill patients, but fluid therapy to
resuscitate patients with sepsis can alter it as well. Volume of dis-
Metabolism
tribution of amikacin has been shown to range widely in a patient Hepatic metabolism depends on primarily hepatic blood flow,
receiving aggressive fluid support (0.27 to 0.61 L/kg), compared enzymatic activity, and protein binding.60,61 Alteration in one
to that of healthy individuals (0.27 + 0.06 L/kg).47 Other sources or more of these physiologic processes will translate into
of fluid administration in these patients, parenteral nutrition and changes in hepatic drug metabolism. The effect of AKI on
intravenous drug administration, add even more volume in this hepatic blood flow is uncertain, but different stages of sep-
population, although their impact may not be readily appreciated. sis and fluid status can affect hepatic blood flow.24 Use
The volume of distribution of aminoglycosides is increased sub- of vasoconstrictors can decrease total hepatic blood flow,
stantially in critically ill patients receiving both total parenteral while inotropes like dobutamine and dopamine have been
nutrition and fluid therapy, compared with those receiving only shown to increase hepatic perfusion by enhancing cardiac
fluid therapy.48,49 Clinicians must recognize the influence of all output.24 Acute kidney injury may also affect Cytochrome
the prescribed volume via fluid, blood products, parenteral nutri- P450 (CYP) enzyme activity. Animal studies indicate that
tion, and drug infusions on antibiotic volume of distribution and effects of AKI on hepatic metabolism are drug specific, but one
may need to use higher antibiotic doses to compensate. The influ- human study demonstrated AKI reduces the activity of CYP 3A
ence of fluid overload on antibiotic concentrations and outcomes enzymes,62 which play a significant metabolic role for more than
in critically ill patients has not been demonstrated definitively. 50% of all drugs. Drugs metabolized by the liver may exert pro-
Several observational studies have highlighted that fluid overload longed therapeutic efficacy by sustaining elevated drug serum
is associated with an increased mortality rate in critically ill concentrations. On the other hand, delayed pharmacologic
patients with AKI.50-53 The Fluids And Catheters Treatment Trial response may be seen with prodrugs requiring activation via
(FACTT) study found that the risk of mortality was increased 1.6- hepatic enzymes. The mechanisms of altered hepatic metabo-
fold per L/d of accumulated fluid.52 In a post hoc analysis from lism in AKI is complicated and poorly understood,60 but clin-
The Program to Improve Care in Acute Renal Disease (PICARD) icians should recognize these potential changes and monitor
trial among nondialyzed patients, the percentage of fluid overload drug serum concentrations to make appropriate dosage modifi-
at the time of AKI diagnosis was twice as high in nonsurvivors cation and to avoid toxicity whenever feasible.
compared with survivors, and the adjusted odds ratio for mortality
related to fluid overload at AKI diagnosis was 3.14.51 Theoreti-
cally, fluid overload could dilute the serum concentrations of anti-
Elimination
biotics 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 overload diminished glomerular filtration and impaired tubular secretion
on mortality and the necessity of higher antibiotic doses to secondary to decreased drug transporter activity can cause
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. Hypo- accumulation and toxicity. This is particularly important with
albuminemia 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
free drug is responsible for pharmacologic effects, protein bind- decreased renal clearance by increasing other elimination path-
ing changes strongly influence efficacy and toxicity.55 Unbound ways may occur during AKI. For these drugs, dosage adjust-
drugs will distribute into tissues, thus increasing volume of dis- ment on a basis of SCr alone may potentially lead to
tribution. Further, unbound drugs will be cleared by kidneys and/ subtherapeutic concentrations. Jones et al found that unmodi-
or RRT, thereby increasing drug clearance. Consequently, fied ciprofloxacin dosage (400 mg intravenously twice daily)
protein-binding changes in critical illness can have a significant did not result in significantly different serum concentrations
effect on highly protein-bound antibiotics. For example, ertape- in patients with severe sepsis with renal impairment (creatinine
nem, a highly protein-bound (85%-95%) carbapenem, yielded clearance < 20 mL/min), compared to those with creatinine
markedly larger volume of distribution (0.21-0.4 L/kg vs 0.07 clearance > 20 mL/min.64 Enhanced transintestinal and biliary
Lewis and Mueller 167

elimination to compensate for reduced renal clearance may current antibiotic dosing recommendations in RRT often fail
account for these findings. to reach pharmacodynamic goals.20,44,67,85-90 These findings
Alteration in nonrenal clearances of several antibiotics such have prompted several clinicians to consider antibiotic doses
as imipenem and vancomycin are also noted in patients with higher than what are traditionally recommended to attain phar-
AKI. Overall, estimated nonrenal clearances of these antibiotics macodynamic target.20,44,67,85-90
decline in patients in AKI compared to that seen in patients with
normal renal function but tend to be higher than those observed
in patients with end-stage renal disease (ESRD).65-68 However, Pharmacodynamic Target Attainment
as AKI persists, vancomycin nonrenal clearance rates progres- Although pharmacokinetics predicts how patient physiology
sively decline and approach that of patients with ESRD.65 affects antibiotic concentrations, pharmacodynamics integrates
The addition of RRT can markedly increase drug clearance, pharmacokinetic characteristics with antibacterial effect. In order
adding another layer to the complexity of antibiotic dosing in cri- to optimize antibiotic therapy, pharmacokinetic parameters must
tically ill patients. Physicochemical properties of drugs such as be accounted for if pharmacodynamic targets are to be met. This
molecular weight, protein binding, and volume of distribution complex pharmacokinetic/pharmacodynamic relationship has
will all influence on drug clearance via RRT. Determining drug been explained by characterizing antibiotics into 2 different cate-
clearance can be complicated, given the heterogeneity in modal- gories, concentration-dependent activity or time-dependent activ-
ities of RRT.69,70 Duration and intensity of RRT can increase ity. For concentration-dependent antibiotics (aminoglycosides,
solute clearance,71 and different modes of fluid replacement daptomycin, and fluoroquinolones), therapeutic outcome will
(predilution or postdilution) will affect solute clearance.69 The be maximized when a high peak serum concentration (Cmax)
details of pharmacokinetic studies of antibiotics in different RRT relative to the organism’s minimum inhibitory concentration
settings are evaluated in several recent reviews.72-77 (MIC) is achieved. With time-dependent activity antibiotics (b-
With the benefits of hemodynamic stability and fluid control, lactams, carbapenems), the time of the serum concentration above
CRRT is often preferred in critically ill patients with AKI, but the MIC (T > MIC) is the most important determinant of their effi-
many antibiotic dosing recommendations in patients receiving cacy. For several antibiotics (vancomycin, linezolid) displaying
CRRT are based on in vitro and very small pharmacokinetic both characteristics, the high ratio of area under the serum con-
studies using obsolete techniques or are extrapolated from phar- centration–time curve (AUC) to the bacterial MIC has been asso-
macokinetic data in noncritically ill patients or patients with ciated with antibacterial success. Besides bacterial killing
ESRD receiving intermittent hemodialysis (IHD).78 In CRRT, characteristics, the postantibiotic effect, the persistent suppres-
the effluent rate (Qf) and the extraction coefficient (sieving coef- sion of bacterial growth after concentrations fall below the MIC
ficient [Sc] or saturation coefficient [Sd]) are 2 primary para- for a particular organism,91 can influence antibacterial effect and
meters used to calculate dialytic drug clearance (CLcrrt ¼ Qf help to determine optimal antibiotic dosing and frequency of
 Sc or Sd). The extraction coefficient approximates the per- administration. Table 1 suggests several alternative antibiotic
centage of unbound drug and the Qf is determined from dialysate dosing strategies to maximize the efficacy of antibiotic therapy
rate (Qd) and ultrafiltration rate (Quf; Qf ¼ Qd þ Quf). Higher in critically ill patients with AKI receiving various RRTs, based
extraction coefficients and Qfs will result in greater drug clear- on available data and consideration of antibiotic profile and extra-
ance. Although several large randomized trials indicated that corporeal antibiotic clearance.
high Qfs do not reduce mortality rate among critically ill
patients,79-81 compared with low Qf, these trials did not take the
impact of high Qfs on increased antibiotic clearance into
Extended Antibiotic Infusion
account.82,83 Because high Qfs yield lower antibiotic concentra- Optimal efficacy of time-dependent killing antibiotics is best
tions, these studies may be confounded by reduced pharmacody- correlated with the T > MIC for susceptible organisms. The
namic target attainment rates in patients receiving high Qfs. T > MIC of at least 40% to 60% of the dosing interval has
Clinicians employing prolonged intermittent RRT (PIRRT) known to be desirable to yield appropriate antibacterial effect
will find that <1% of drugs have validated dosing recommenda- of b-lactams,105,106 but maintaining a longer T > MIC has shown
tions available to them.76,84 Recent reviews suggest that, despite to further maximize the time-dependent killing effect. McKin-
the ‘‘slower than IHD’’ blood and dialysate flow rates, the non et al indicated that achievement of T > MIC of 100% for
extended duration of PIRRT (8-12 hours) contributes to substan- b-lactams resulted in much higher clinical cure than T > MIC
tial antibiotic clearance, increasing the risk of prolonged subther- of less than 100% in patients with sepsis (82% vs 33%).107
apeutic antibiotic concentrations.76,77 In addition, the extended Serendipitously, patients with AKI may be able to take an
duration of PIRRT may result in some doses administered while advantage of reduced renal clearance to prolong T > MIC.
dialysis is running, which will affect delivered drug dose. This However, RRT drug removal, particularly CRRT, may inter-
can further complicate efficacy of antibiotic therapy. fere with this advantage and even cause the drug concentration
Appropriate antibiotic dosing in critically ill patients with to fall below the MIC for a susceptible organism unless a dos-
RRT requires understanding of the influences of RRT variables ing change is made to account for CRRT clearance. In patients
and non-RRT clearance on antibiotic therapy to determine opti- with AKI receiving continuous venovenous hemodiafiltration
mal dosing regimens. As evidenced in numerous reports, at an Qf of 1 to 2 L/h, the mean elimination of piperacillin/
168 Journal of Intensive Care Medicine 31(3)

Table 1. Antibiotic Dosing Strategies in Critically Ill Patients With AKI hemofiltration or continuous venovenous hemodiafiltration
to Improve Pharmacodynamic Target Attainment Rates. reports that continuous infusion of vancomycin (16-35 mg/
Antibacterial
kg/d) after a loading dose of 15 mg/kg could be employed
Profile Potential Dosing Strategies to achieve adequate serum concentrations (20-30 mg/mL) on
the first day of therapy.83 However, drug accumulation was
Time dependent noticeable on day 3 of therapy, and the proportion of patients
b-lactams (1) Continuous or prolonged infusions in with excessive vancomycin trough concentrations (> 30 mg/
Carbapenems CRRT,83,92-96 but not IHD or PIRRT
mL) was significantly higher in those with higher daily doses.
Vancomycin (2) More frequent administration of smaller doses
(3) Supplemental dose during PIRRT97,98 and post- Potential drug accumulation and higher risk of toxicity man-
IHD dates routine monitoring of vancomycin serum concentration
(4) Weight-based dosing and subsequent dosage adjustment for this strategy.85
Concentration dependent Prolonged infusion is a compromise between a typical
Aminoglycosides (1) Loading dose to achieve target 30-minute antibiotic infusion and continuous infusion. Pro-
Colistin pharmacodynamic goals (peaks) early92-94,99,100 longed infusion duration can vary but is often as long as half
Daptomycin (2) Extended dosing interval for aminoglycosides99
the dosing interval and does extend T > MIC. An advantage
Fluoroquinolones (3) Predialytic administration of aminoglycosides in
Linezolid IHD or PIRRT101-104 of this practice is that an intravenous line does not have to be
Macrolides (4) Weight-based dosing dedicated solely to continuous antibiotic infusion. Use of pro-
Metronidazole longed infusion antibiotics has not been published in patients
Vancomycin with CRRT. With higher peaks and lower troughs, intermittent
infusion will produce the same AUC as prolonged infusion, but
Abbreviations: AKI, acute kidney injury; CRRT, continuous renal replacement
therapy; IHD, intermittent hemodialysis; PIRRT, prolonged intermittent renal a higher proportion of T > MIC with prolonged infusion theo-
replacement therapy. retically should result in better efficacy.
This strategy of extended infusions may be particularly valu-
tazobactam was reported to be nearly equivalent to patients able in a case with reduced susceptibility, high CRRT Qf,
with normal renal function.108 Seyler and colleagues report that increased antibiotic volume of distribution, and/or in immuno-
recommended b-lactam dosing for critically ill patients under- compromised patients where prolonged serum concentration
going CRRT with the mean Qf of 45 mL/kg/h failed to achieve above the MIC may be critical. Compared to shorter conven-
stated pharmacodynamic goals against Pseudomonas aerugi- tional infusion times, extended antibiotic infusions can be diffi-
nosa within 48 hours of antibiotic therapy in 34% to 100% of cult to employ because of limited vascular access in the ICU and
modeled cases.20 Therefore, initiation of CRRT should be fol- drug compatibility/stability issues. More frequent administration
lowed by the consideration of increased antibiotic doses or of smaller doses may be able to mimic the T > MIC of extended
alternative administration strategies to prolong T > MIC with infusion without a drop below the organism’s MIC. Further stud-
b-lactams. ies are warranted to investigate the efficacy and safety of
Extended antibiotic infusion time can increase T > MIC; extended antibiotic infusion strategies in the patients with AKI.
however, clinically improved outcomes of longer infusions
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
25 mL/kg/h.93 The continuous infusion regimen generated a reinforced by the growing findings that initial suboptimal anti-
constant serum concentration above the MIC (Cmedian 19.1 biotic exposure can reduce the susceptibility of pathogens and
mg/L) for 100% of the treatment period, while the intermittent facilitate the amplification of resistant strains in a large
bolus maintained serum concentration above the MIC of inoculum.115,116 Considering the increased volume of distribu-
8 mg/L for only 45% of the dosing interval. This 45% T > tion and constant extracorporeal drug removal, patients receiv-
MIC is thought to be a minimally required T > MIC goal but ing CRRT may require a loading dose of all types of antibiotics.
might not be sufficient to cover intermediately resistant P A subgroup analysis by Taccone et al indicated that patients
aeruginosa, a common pathogen in critically ill patients with severe sepsis treated with CRRT with the mean Qf of
(Figure 2). The benefit of continuous infusion has also been 33 mL/kg/h required a loading dose of at least 25 mg/kg of ami-
proposed for vancomycin which predominantly has a time- kacin, substantially higher than the usual loading dose,117 to
dependent killing activity and will be effectively removed attain adequate peak concentrations.99 However, while the
by conventional CRRT.65,83,94,95,112,113 A retrospective eva- 25mg/kg loading dose yielded target peak concentrations, the
luation in patients receiving continuous venovenous serum concentrations 24 hours later were persistently high.
Lewis and Mueller 169

accounting for RRT drug clearance. Predialysis administration


25
1g q12h Intermittent infusion of higher dosage has been suggested to be more efficacious than
conventional postdialysis aminoglycoside administration
Serum Concentration (mg/L)

20 2g q24h Continuous infusion


because it can allow a higher peak concentration to maximize
antibacterial pharmacodynamic goals (peak/MIC) while subse-
15
quently rapid dialytic clearance can minimize toxicity from pro-
longed drug exposure (Figure 3).101,102,118,119 A simulated
10
gentamicin 6 mg/kg dose given 1 hour prior to a 4-hour HD
MIC of 8mg/L
treatment achieved predetermined pharmacodynamic targets
5
with a mean peak concentration of 31 mg/mL. However, a simu-
lated post-HD infusion of 1.7 mg/kg dose, the maximum dose
0
0 2 4 6 8 10 12 14 16 18 20 22 24
recommended by the package insert, failed to meet any pharma-
Time (h)
codynamic targets (Cmax 8.8 + 3.1 mg/L).103 Use of a high pre-
dialytic antibiotic dose is also proposed for PIRRT, where
Figure 2. Modeled meropenem steady state concentrations with
‘‘when to dose’’ matters more than ‘‘how much to dose’’ due
continuous infusion versus intermittent infusion in a patient on con- to unpredictable drug clearance with the prolonged duration of
tinuous renal replacement therapy (CRRT).93 A hypothetical 70-kg dialysis.77 Roberts et al showed that gentamicin 6 mg/kg every
patient with anuria having AKI receiving CRRT with an effluent rate of 48 hours given 30 minutes to 1 hour before a 10-hour PIRRT ses-
35 mL/kg/h is modeled. The percentage of time of the serum con- sion can be an effective dosing strategy to maximize pharmaco-
centration above the minimum inhibitory concentration (MIC; T > dynamic target attainment while minimizing sustained elevated
MIC) at 8 mg/L for intermediately resistant Pseudomonas aeruginosa is serum concentrations responsible for toxicity.104 However, cau-
58% with meropenem 1 g every 12 hours intermittent infusion over 30
minutes and 100% with 2 g every 24 hours continuous infusion.
tion should be followed when this novel strategy is instituted.
Delay in dialysis schedule or unexpected dialysis discontinua-
Consequently, if this regimen is to be used, the dosing interval tion secondary to clotting or intolerance can put a patient at risk
should be longer than 24 hours for these CRRT Qfs.99 of antibiotic toxicity due to inadequate dialytic drug clearance.
Use of loading doses should not be limited to concentration- Thus, when the benefit of maximized pharmacokinetic/pharma-
dependent antibiotics. A loading dose can be effective to codynamic goal is critical and a subsequent full dialysis session
achieve early therapeutic concentration of all antibiotics is ensured, clinicians may consider predialytic administration of
including those with time-dependent activity.94,95,100 A loading a higher aminoglycoside dose.
dose is needed when continuous infusion b-lactams are used
because continuous infusion initially results in subtherapeutic Weight-Based Dosing
drug levels until the serum concentration slowly reaches the
The ‘‘one-size-fit-all’’ dosing strategy (eg 500 mg regardless of
equilibration with the tissues.92,93 Early attainment of thera-
body mass) ignores the variability in body size and body fluid
peutic drug concentrations at the infection site should improve
composition of critically ill patients, possibly leading to an
increased mortality associated with a delay in appropriate anti-
inappropriately low serum concentration. For example, in the
biotic therapy.1 Consequently, continuous infusion b-lactams
case described at the beginning of this article, we present a
should follow a loading dose.96 Other RRTs offer different
patient with *15 L fluid overloaded, sepsis, possibly hypo-
antibiotic administration challenges. The PIRRT removes a
albuminemia, and in need of RRT. Most penicillin, cephalo-
significant amount of antibiotics because of prolonged dialysis
sporin, and carbapenem antibiotics have recommended dosing
duration. It may require a higher dose during the duration of
in adults who are ‘‘flat dosed’’ with a dosing interval adjusted
PIRRT and less when PIRRT is off. Supplemental doses may
for AKI. The use of a 500-mg carbapenem/cephalosporin dose
be guided by therapeutic drug monitoring to ensure adequate
in a patient with volume of distribution that is likely 50% to
maintenance of therapeutic serum concentrations.97 The con-
100% larger than that of the ‘‘normal volunteer’’ in whom this
ventional daptomycin dose of 6 mg/kg recommended for IHD
dose was derived will have a substantially lower chance to
did not produce targeted AUC values when it was infused
attain pharmacodynamic targets. Use of mg/kg or mg/m2 dos-
within 8 hours before an 8-hour PIRRT session (Qb and
ing regimens, like our colleagues who treat pediatric patients
Qd of 160 mL/min) due to increased PIRRT drug
usually use, is much more likely to meet these targets.
clearance.97 Clinicians may use additional postdialytic antibio-
tic doses to accelerate the achievement of pharmacodynamic
goals or to prevent unwanted drop in antibiotic level caused Inadequate Concentration at the Target Sites
by the extended duration of PIRRT.98
The emphasis of the Surviving Sepsis Guidelines on antibiotics
that ‘‘penetrate in adequate concentrations into tissue presumed
Altered Antibiotic Administration Time to be the source of sepsis’’ is often overlooked because of our
Changing antibiotic administration time with relation to RRT limited ability to measure drug concentrations at the actual sites
can be a strategy to meet pharmacodynamic goals while of infection which mostly occur in tissue. Serum concentrations
170 Journal of Intensive Care Medicine 31(3)

20% lower clinical response to vancomycin therapy, compared


30
to those infected with low MIC (MIC < 2 mg/mL) MRSA strains,
Pre-dialytic 6mg/kg dose despite targeted vancomycin trough concentrations of 15 to 20
Serum Concentration (mg/L)

25
Post-dialytic 1.7mg/kg dose mg/mL.129 For a pathogen with an elevated MIC, higher doses
20 of antibiotic therapy will be required to achieve
targets.130 However, higher antibiotic doses may cause antibiotic
15
toxicity in this vulnerable patient population, as high vancomy-
10
cin trough levels (15-20 mg/mL) were found to be a significant
predictor of nephrotoxicity development in the latter
5 study.129 Alternative antibiotic agents may be considered in a
case with high MIC strains and increased predisposition to drug
0 toxicity.131 Treatment of infection in the ICU becomes more
0 2 4 6 8 10 12 14 16 18 20 22 24
challenging with the emerging incidence of multidrug resistant
Time (h)
and even panantibiotic-resistant pathogens among Acinetobacter
species, P aeruginosa, carbapenem-resistant Klebsiella species
Figure 3. Modeled gentamicin predialytic high dose versus postdia- and E coli.132-134 The effectiveness of available antibiotics and
lytic lower dose.103,120 A hypothetical 70-kg patient with anuria having
AKI receiving a 4-hour intermittent hemodialysis (IHD) session is
efforts to develop novel antibiotics have been declining as
modeled. Gentamicin predialytic 6 mg/kg dose yielded a high-peak well.135 All these circumstances mandate clinicians to aim
serum concentration (Cmax) of 26.8 mg/L which is higher than pro- toward higher pharmacodynamic targets and higher antibiotic
posed pharmacodynamic target of Cmax  8 mg/L, while postdialytic doses to ensure bacterial eradication and to reduce the risk of fur-
1.7 mg/kg dose yielded Cmax of 7.6 mg/L. ther development of antibiotic resistance.127,128 As discussed,
clinicians should endeavor to maximize antibacterial killing
are routinely monitored to predict therapeutic efficacy, with a effect by using aggressive antibiotic dosing and/or alternative
theoretical assumption of equilibration between free drug in dosing strategies.19,20,41,92,93,102,111,136
plasma and tissue. However, tissue penetration might be
impaired in critically ill patients due to altered pathophysiology Toxicity
and transporter activity.121-123 In a small prospective study,
interstitial concentrations of piperacillin in soft tissues were 7- Although there are many compelling reasons to consider more
fold lower than free plasma concentrations in patients with septic aggressive antibiotic dosing in critically ill patients with AKI,
shock.123 Results from animal and human studies demonstrate toxicity from antibiotic doses that are too high is a genuine
that plasma concentrations do not always represent equivalent concern. These dynamic yet vulnerable patients often have
concentration in the sites of infection,124-126 suggesting that poor multiple organ dysfunction but often receive many toxic
tissue penetration may result in subtherapeutic antibiotic concen- medications. Nephrotoxic drugs account for 22% of the total
trations at infection sites and consequent treatment failure, which medication orders in the adult ICU patients.137 With the pres-
is not uncommon in patients with sepsis. Accordingly, modify- ence of numerous risk factors, these patients would be more
ing antibiotic doses based on the measured plasma concentration prone to drug accumulation and toxicity due to AKI. In their
may not guarantee to achieve the desired concentration at the 7.5-year retrospective study, Kane-Gill and colleagues identi-
sites of infection. Therefore, in the absence to measure adequacy fied that critically ill patients with AKI were 16 times more
of antibiotic concentration at the infection sites, higher antibiotic prone to manifest drug-induced toxicity compared to those
dosage may be warranted to ensure adequate local antibiotic without AKI.138 Antibiotics were the most common drug class
concentrations in these patients. causing toxicity in the ICU.138
Despite the concern for their well-known nephrotoxicity, ami-
noglycosides and vancomycin remain as mainstays of antibiotic
Rising Bacterial Resistance Rates therapy in the ICU. Another renowned nephrotoxin, colistin, has
Finally, the increasing prevalence of antibiotic resistance high- been reintroduced as a last treatment option for multidrug resis-
lights the significance of ‘‘appropriate’’ antibiotic therapy. Anti- tant gram-negative bacterial infections, despite its exceedingly
biotic resistance in critically ill patients has been related to high rate of nephrotoxicity (~50%).139,140 Treatment with these
increased mortality rates in several studies.9,127,128 In a retro- nephrotoxic agents requires the clinician to make difficult choices
spective cohort, a significantly higher risk of hospital mortality when it is apparent that antibiotic toxicity is likely to develop. A
was noted in patients with sepsis who were infected with resis- study with 201 critically ill patients treated with aminoglycosides
tant gram-negative bacteria (63.4% vs 40.0%).9 Further logistic reported up to 30% of incidence of nephrotoxicity.141 Moreover,
regression analysis determined resistance to the initial antibiotics fever, endotoxemia, and renal hypoperfusion associated with sep-
as an independent risk factor, associated with a doubling of hos- sis and AKI can aggravate preexisting renal impairment with the
pital mortality.9 A prospective study also demonstrated that use of aminoglycosides.142
patients infected with high vancomycin MIC ( 2 mg/mL) of Although the guidelines suggest raising goal trough serum
methicillin-resistant Staphylococcus aureus (MRSA) strains had concentrations for vancomycin, a recent meta-analysis suggests
Lewis and Mueller 171

that patients with higher trough serum concentrations (>15 mg/ compared to the cost related to antibiotic treatment failure.
dL) had a 2- to 3-time greater risk of nephrotoxicity than those Unfortunately, data quantifying both direct and indirect costs
who had lower troughs (<15 mg/dL).143 High dose, longer dura- of antibiotic toxicity are scarce. In an economic evaluation of
tion of therapy, concomitant use of other nephrotoxins, and renal aminoglycoside nephrotoxicity, the total hospital costs includ-
impairment are common risk factors to precipitate nephrotoxi- ing pharmacokinetic and nephrologist consultations, ancillary
city with these antibiotics.139,140,143-149 Ensuring pharmacody- laboratory tests, and additional hospital stay was estimated to
namic target attainment with high doses can improve patient be 2-fold higher in patients with nephrotoxicity compared to
outcome, but such benefit has definite but unpredictable toxicity those without nephrotoxicity.164
trade-offs.150-152 Thus, the risk–benefit analysis should be per-
formed when clinicians consider these antibiotics, and close
monitoring must occur during the therapy. Conclusion
Beyond nephrotoxicity, other toxicities associated with anti- A multitude of factors influence the determination of ‘‘appro-
biotic accumulation can occur. For example, b-lactam antibio- priate antibiotic dosing’’ in critically ill patients with AKI.
tics have been associated with neurotoxicity. Carbapenem- Many of them motivate clinicians to consider higher antibiotic
induced seizures have been well documented in the literature doses, but toxicity associated with higher doses may also cause
in patients with renal insufficiency receiving high doses a negative patient outcome. Balancing the concerns of ‘‘too
(eg imipenem > 4 g/day).153-157 Neurotoxicity associated much’’ antibiotic with the compelling pharmacokinetic, phar-
with cefepime in the ICU setting has been increasingly macodynamic, and antibacterial resistance evidence suggesting
described.158-161 In a prospective study, reduced renal function that higher doses are necessary should initiate intriguing con-
(creatinine clearance < 30 mL/min) was a strong predictor for versations among the interdisciplinary team caring for criti-
nonconvulsive epilepsy (eg altered mental status, myoclonus) cally ill patients with AKI. For patients like the case
in patients receiving cefepime.160 Intriguingly, the doses used described at the beginning of this article, it is likely best that
in these incidents were appropriately adjusted for renal we try to rapidly identify the appropriate antibiotics and then
function and were not initially linked to the drug accumula- use as much of those antibiotics as we can safely, particularly
tion until the trough level confirmed toxicity. These authors early in the course of the patient’s sepsis and AKI. Patient mon-
postulated that other individual pathophysiological variables itoring needs to be as vigilant as possible to identify antibiotic
such as plasma albumin and hemoglobin concentrations may toxicity as early as possible.
influence pharmacokinetic properties, increasing the serum
cefepime trough levels.160 The authors suggested that unno- Declaration of Conflicting Interests
ticed toxicity may contribute to the increased mortality rate
The author(s) declared no potential conflicts of interest with respect to
associated with cefepime therapy versus other b-lactam the research, authorship, and/or publication of this article.
therapy.160 Alternative administration techniques such as
continuous or prolonged infusion might yield lower peaks
Funding
and have an added benefit of reducing the toxicity risk of
b-lactams. The author(s) received no financial support for the research, authorship,
and/or publication of this article.
Most studies suggest that antibiotic-induced toxicities are
transient and reversible upon discontinuation of the insulting
agent, viewing them as acute issues. However, short-term out- References
comes during a hospital stay may not reflect the full spectrum 1. Dellinger RP, Levy MM, Rhodes A, et al. Surviving Sepsis Cam-
of antibiotic toxicity. This approach may underestimate poten- paign: international guidelines for management of severe sepsis
tial long-term effect on morbidity and mortality of these and septic shock, 2012. Intensive Care Med. 2013;39(2):165-228.
patients.162 Although there is a dearth of studies on this issue, 2. Uchino S, Kellum JA, Bellomo R, et al. Acute renal failure in cri-
a prospective observational study reports that after 5-year tically ill patients. JAMA. 2005;294(7):813-818.
follow-up, the mortality of survivors from severe AKI requir- 3. Silvester W, Bellomo R, Cole L. Epidemiology, management, and
ing RRT was 47%.163 The causes of AKI in these patients outcome of severe acute renal failure of critical illness in Austra-
include sepsis (33%) and nephrotoxins (7%). Among those sur- lia. Crit Care Med. 2001;29(10):1910-1915.
viving patients, only 57% of them had complete recovery of 4. Hoste EA, Clermont G, Kersten A, et al. RIFLE criteria for acute
renal function at hospital discharge. Patients with partial renal kidney injury are associated with hospital mortality in critically ill
function recovery showed significantly higher mortality rate, patients: a cohort analysis. Crit Care. 2006;10(3):R73.
compared to those with complete recovery after 5 years (P ¼ 5. Waikar SS, Liu KD, Chertow GM. The incidence and prognostic
.001).163 Therefore, short-term toxicity caused by antibiotics significance of acute kidney injury. Curr Opin Nephrol Hyper-
may carry the prolonged risk of deteriorating organ function tens. 2007;16(3):227-236.
and increased probability of mortality as a long-term conse- 6. Bagshaw SM, Uchino S, Kellum JA, et al. Association between
quence in critically ill patients. renal replacement therapy in critically ill patients with severe
Antibiotic toxicity is also associated with substantial acute kidney injury and mortality. J Crit Care. 2013;28(6):
increase in health care costs, although it may be understated 1011-1018.
172 Journal of Intensive Care Medicine 31(3)

7. Aronoff GR, Aronoff JR. Drug prescribing in kidney disease: 25. Smith BS, Yogaratnam D, Levasseur-Franklin KE, Forni A, Fong
can’t we do better? Am J Kidney Dis. 2014;63(3):382-383. J. Introduction to drug pharmacokinetics in the critically ill
8. Farag A, Garg AX, Li L, Jain AK. Dosing errors in prescribed patient. Chest. 2012;141(5):1327-1336.
antibiotics for older persons with CKD: a retrospective time series 26. Roberts DJ. Drug absorption, distribution, metabolism and excre-
analysis. Am J Kidney Dis. 2014;63(3):422-428. tion considerations in critically ill adults. Expert Opin Drug
9. Micek ST, Welch EC, Khan J, et al. Resistance to empiric Metab Toxicol. 2013;9(9):1067-1084.
antimicrobial treatment predicts outcome in severe sepsis 27. Nguyen NQ, Ng MP, Chapman M, Fraser RJ, Holloway RH. The
associated with gram-negative bacteremia. J Hosp Med. impact of admission diagnosis on gastric emptying in critically ill
2011;6(7):405-410. patients. Crit Care. 2007;11(1):R16.
10. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo 28. Mueller BA, Brierton DG, Abel SR, Bowman L. Effect of enteral
J, Pinsky MR. Epidemiology of severe sepsis in the United States: feeding with Ensure on oral bioavailabilities of ofloxacin and cipro-
analysis of incidence, outcome, and associated costs of care. Crit floxacin. Antimicrob Agents Chemother. 1994;38(9):2101-2105.
Care Med. 2001;29(7):1303-1310. 29. Mimoz O, Binter V, Jacolot A, et al. Pharmacokinetics and abso-
11. Martin GS, Mannino DM, Eaton S, Moss M. The epidemiology of lute bioavailability of ciprofloxacin administered through a naso-
sepsis in the United States from 1979 through 2000. N Engl J gastric tube with continuous enteral feeding to critically ill
Med. 2003;348(16):1546-1554. patients. Intensive Care Med. 1998;24(10):1047-1051.
12. Angus DC, Pereira CA, Silva E. Epidemiology of severe sepsis 30. Blum RA, D’Andrea DT, Florentino BM, et al. Increased gastric
around the world. Endocr Metab Immune Disord Drug Targets. pH and the bioavailability of fluconazole and ketoconazole. Ann
2006;6(2):207-212. Intern Med. 1991;114(9):755-757.
13. Vincent JL, Rello J, Marshall J, et al. International study of the 31. Piscitelli SC, Goss TF, Wilton J, D’Andrea DT, Goldstein H,
prevalence and outcomes of infection in intensive care units(EPIC Schentag JJ. Effects of ranitidine and sucralfate on ketocona-
II study). JAMA. 2009;302(21):2323-2329. zole bioavailability. Antimicrob Agents Chemother. 1991;
14. Quenot JP, Binquet C, Kara O, et al. The epidemiology of septic 35(9):1765-1771.
shock in French intensive care units: the prospective multicenter 32. Chin TWF, Loeb M, Fong IW. Effects of an acidic beverage
cohort EPISS study. Crit Care. 2013;17(2):R65. (Coca-Cola) on absorption of ketoconazole. Antimicrob Agents
15. Kollef MH, Sherman G, Ward S, Fraser VJ. Inadequate antimicro- Chemother. 1995;39(8):1671-1675.
bial treatment of infections: a risk factor for hospital mortality 33. Dörffler-Melly J, de Jonge E, Pont AC, et al. Bioavailability of
among critically ill patients. Chest. 1999;115(2):462-474. subcutaneous low-molecular- weight heparin to patients on vaso-
16. Ibrahim EH, Sherman G, Ward S, Fraser VJ, Kollef MH. The pressors. Lancet. 2002;359(9309):849-850.
influence of inadequate antimicrobial treatment of bloodstream 34. Haas CE, Nelsen JL, Raghavendran K, et al. Pharmacokinetics
infections on patient outcomes in the ICU setting. Chest. 2000; and pharmacodynamics of enoxaparin in multiple trauma patients.
118(1):146-155. J Trauma. 2005;59(6):1336-1343.
17. Harbarth S, Garbino J, Pugin J, Romand JA, Lew D, Pittet D. 35. Parrillo JE. Pathogenetic mechanisms of septic shock. N Engl J
Inappropriate initial antimicrobial therapy and its effect on sur- Med. 1993;328(20):1471-1477.
vival in a clinical trial of immunomodulating therapy for severe 36. Triginer C, Izquierdo I, Fernandez R, et al. Gentamicin volume of
sepsis. Am J Med. 2003;115(7):529-535. distribution in critically ill septic patients. Intensive Care Med.
18. Shorr AF, Micek ST, Welch EC, Doherty JA, Reichley RM, Kollef 1990;16(5):303-306.
MH. Inappropriate antibiotic therapy in Gram-negative sepsis 37. Gous AG, Dance MD, Lipman J, Luyt DK, Mathivha R, Scribante
increases hospital length of stay. Crit Care Med. 2011;39(1):46-51. J. Changes in vancomycin pharmacokinetics in critically ill
19. Taccone FS, Laterre PF, Dugernier T, et al. Insufficient B-lactam infants. Anaesth Intensive Care. 1995;23(6):678-682.
concentrations in the early phase of severe sepsis and septic 38. Tang GJ, Tang JJ, Lin BS, Kong CW, Lee TY. Factors affecting
shock. Crit Care. 2010;14(4):R126. gentamicin pharmacokinetics in septic patients. Acta Anaesthesiol
20. Seyler L, Cotton F, Taccone FS, et al. Recommended B-lactam Scand. 1999;43(7):726-730.
regimens are inadequate in septic patients treated with continuous 39. Buijk SE, Mouton JW, Gyssens IC, Verbrugh HA, Bruining HA.
renal replacement therapy. Crit Care. 2011;15(3):R137. Experience with a once- daily dosing program of aminoglycosides
21. Udy AA, Varghese JM, Altukroni M, et al. Subtherapeutic initial in critically ill patients. Intensive Care Med. 2002;28(7):936-942.
B-lactam concentrations in select critically ill patients. Chest. 40. Del Mar Fernandez de Gatta Garcia M, Revilla N, Calvo MV,
2012;142(1):30-39. Dominquez-Gil A, Sanchez Navarro A. Pharmacokinetic/pharma-
22. Binder L, Schwörer H, Hoppe S, et al. Pharmacokinetics of mer- codynamics analysis of vancomycin in ICU patients. Intensive
openem in critically ill patients with severe infections. Ther Drug Care Med. 2007;33(2):279-285.
Monit. 2013;35(1):63-70. 41. Taccone FS, Hites M, Beumier M, Scolletta S, Jacobs F. Appro-
23. Stechmiller JK, Treloar D, Allen N. Gut dysfunction in critically priate antibiotic dosage levels in the treatment of severe sepsis
ill patients: a review of the literature. Am J Crit Care. 1997;6(3): and septic shock. Curr Infect Dis Rep. 2011;13(5):406-415.
204-209. 42. Vilay AM, Grio M, DePestel DD, et al. Daptomycin pharmacoki-
24. Boucher BA, Wood GC, Swanson JM. Pharmacokinetic changes netics in critically ill patients receiving continuous venovenous
in critically illness. Crit Care Clin. 2006;22(2):255-271. hemodialysis. Crit Care Med. 2011;39(1):19-25.
Lewis and Mueller 173

43. Wenisch JM, Meyer B, Fuhrmann V, et al. Multiple-dose pharma- 58. Andreasen F. The effect of dialysis on the protein binding of drugs
cokinetics of daptomycin during continuous venovenous haemo- in the plasma of patients with acute renal failure. Acta Pharmacol
diafiltration. J Antimicrob Chemother. 2012;67(4):977-983. Toxicol. 1974;34(4):284-294.
44. Bauer SR, Salem C, Connor MJ Jr, et al. Pharmacokinetics and 59. Mussche MM, Belpaire FM, Bogaert MG. Plasma protein binding
pharmacodynamics of piperacillin-tazobactam in 42 patients of phenylbutazone during recovery from acute renal failure. Eur J
treated with concomitant CRRT. Clin J Am Soc Nephrol. 2012; Clin Pharmacol. 1975;9(1):69-71.
7(3):452-457. 60. Vilay AM, Churchwell MD, Mueller BA. Clinical review: drug
45. Khadzhynov D, Slowinski T, Lieker I, et al. Plasma pharmacoki- metabolism and nonrenal clearance in acute kidney injury. Crit
netics of daptomycin in critically ill patients with renal failure and Care. 2008;12(6):235.
undergoing CVVHD. Int J Clin Pharmacol Ther. 2011;49(11): 61. Lalande L, Charpiat B, Leboucher G, Tod M. Consequences of
656-665. renal failure on non-renal clearance of drugs. Clin Pharmacoki-
46. Dvorchik B, Arbeit RD, Chung J, Liu S, Knebel W, Kastrissios H. net. 2014;53(6):521-532.
Population pharmacokinetics of daptomycin. Antimicrob Agents 62. Kirwan CJ, MacPhee IA, Lee T, Holt DW, Philips BJ. Acute kid-
Chemother. 2004;48(8):2799-2807. ney injury reduces the hepatic metabolism of midazolam in criti-
47. Botha FJ, van der BIJL P, Seifart HI, Parkin DP. Fluctuation of cally ill patients. Intensive Care Med. 2012;38(1):76-84.
the volume of distribution of amikacin and its effect on once- 63. Sun H, Frassetto L, Benet LZ. Effects of renal failure on drug
daily dosage and clearance in a seriously ill patient. Intensive transport and metabolism. Pharmacol Ther. 2006;109(1-2):1-11.
Care Med. 1996;22(5):443-446. 64. Jones EM, McMullin CM, Hedges AJ, et al. The pharmacokinetics
48. Tormo C, Abad FJ, Ronchera-Oms CL, Parra V, Jimenez NV. of intravenous ciprofloxacin 400 mg 12 hourly in patients with
Critically-ill patients receiving total parenteral nutrition show severe sepsis: the effect of renal function and intra-abdominal dis-
altered amikacin pharmacokinetics. Clinical Nutrition. 1995; ease. J Antimicrob Chemother. 1997;40(1):121-124.
14(4):254-259. 65. Macias WL, Mueller BA, Scarim SK. Vancomycin pharmacoki-
49. Ronchera-Oms CL, Tormo C, Ordovas JP, Abad J, Jimenez NV. netics in acute renal failure: preservation of nonrenal clearance.
Expanded gentamicin volume of distribution in critically ill adult Clin Pharmacol Ther. 1991;18(4):451-458.
patients receiving total parenteral nutrition. J Clin Pharm Ther. 66. Mueller BA, Scarim SK, Macias WL. Comparison of imipenem
1995;20(5):253-258. pharmacokinetics in patients with acute or chronic renal failure
50. Payen D, de Pont AC, Sakr Y, et al. A positive fluid balance is treated with continuous hemofiltration. Am J Kidney Dis. 1993;
associated with a worse outcome in patients with acute renal fail- 21(2):172-179.
ure. Crit Care. 2008;12(3):R74. 67. Tegeder I, Bremer F, Oelkers R, et al. Pharmacokinetics of
51. Bouchard J, Soroko SB, Chertow GM, et al. Fluid accumula- imipenem-cilastatin in critically ill patients undergoing continu-
tion, survival and recovery of kidney function in critically ill ous venovenous hemofiltration. Antimicrob Agents Chemother.
patients with acute kidney injury. Kidney Int. 2009;76(4): 1997;41(12):2540-2545.
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 Syn- human patients. Clin Pharmacol Ther. 1988;43(5):565-570.
drome Network. Fluid balance, diuretic use, and mortality in acute 69. Brunet S, Leblanc M, Geadah D, Parent D, Courteau S, Cardinal J.
kidney injury. Clin J Am Soc Nephrol. 2011;6(5):966-973. Diffusive and convective solute clearances during continuous
53. Teixeira C, Garzotto F, Piccinni P, et al. Fluid balance and urine renal replacement therapy at various dialysate and ultrafiltration
volume are independent predictors of mortality in acute kidney flow rates. Am J Kidney Dis. 1999;34(3):886-892.
injury. Crit Care. 2013;17(1):R14. 70. Churchwell MD, Mueller BA. Drug dosing during continuous
54. SAFE Study Investigators. Finfer S, Bellomo R, McEvoy S, et al. renal replacement therapy. Semin Dial. 2009;22(2):185-188.
Effect of baseline serum albumin concentration on outcome of 71. Pasko DA, Churchwell MD, Salama NN, Mueller BA. Longitudi-
resuscitation with albumin or saline in patients in intensive care nal hemodiafilter performance in modeled continuous renal
units: analysis of data from the Saline versus Albumin Fluid Eva- replacement therapy. Blood Purif. 2011;32(2):82-88.
luation (SAFE) study. BMJ. 2006;333(7577):1044. 72. Choi G, Gomersall CD, Tian Q, Joynt GM, Freebairn R, Lipman
55. Roberts JA, Federico P, Lipman J. The clinical relevance of J. Principles of antibacterial dosing in continuous renal replace-
plasma protein binding changes. Clin Pharmacokinet. 2013; ment therapy. Crit Care Med. 2009;37(7):2268-2282.
52(1):1-8. 73. Heintz BH, Matzke GR, Dager WE. Antimicrobial dosing con-
56. Burkhardt O, Kumar V, Katterwe D, et al. Ertapenem in critically cepts and recommendations for critically ill adult patients receiv-
ill patients with early-onset ventilator-associated pneumonia: ing continuous renal replacement therapy or intermittent
pharmacokinetics with special consideration of free-drug concen- hemodialysis. Pharmacotherapy. 2009;29(5):562-577.
tration. J Antimicrob Chemother. 2007;59(2):277-284. 74. Eyler RF, Mueller BA. Antibiotic dosing in critically ill patients
57. Eyler RF, Vilay AM, Nader AM, et al. Pharmacokinetics of erta- with acute kidney injury. Nat Rev Nephrol. 2011;7(4):226-235.
penem in critically ill patients receiving continuous venovenous 75. Kielstein JT, Burkhardt O. Dosing of antibiotics in critically ill
hemodialysis or hemodiafiltration. Antimicrob Agents Che- patients undergoing renal replacement therapy. Curr Pharm Bio-
mother. 2014;58(3):1320-1326. technol. 2011;12(12):2015-2019.
174 Journal of Intensive Care Medicine 31(3)

76. Bogard KN, Peterson NT, Plumb TJ, Erwin MW, Fuller PD, 92. Mariat C, Venet C, Jehl F, et al. Continuous infusion of ceftazi-
Olsen KM. Antibiotic dosing during sustained low-efficiency dia- dime in critically ill patients undergoing continuous venovenous
lysis: special consideration in adult critically ill patients. Crit haemodiafiltration: pharmacokinetic evaluation and dose recom-
Care Med. 2011;39(3):560-570. mendation. Crit Care. 2006;10(1):R26.
77. Scoville BA, Mueller BA. Medication dosing in critically ill 93. Langgartner J, Vasold A, Glück T, Reng M, Kees F. Pharma-
patients with acute kidney injury with renal replacement therapy. cokinetics of meropenem during intermittent and continuous
Am J Kideny Dis. 2013;61(3):490-500. intravenous application in patients treated by continuous
78. Mueller BA, Smoyer WE. Challenges in developing evidence- renal replacement therapy. Intensive Care Med. 2008;34(6):
based drug dosing guidelines for adults and children receiving renal 1091-1096.
replacement therapy. Clin Pharmacol Ther. 2009;86(5):479-482. 94. Saugel B, Nowack MC, Hapfelmeier A, et al. Continuous intra-
79. Vesconi S, Cruz DN, Fumagalli R, et al. DOse REsponse Multi- venous administration of vancomycin in medical intensive care
centre International collaborative Initiative (DO-RE-MI Study unit patients. J Crit Care. 2013;28(1):9-13.
Group): delivered dose of renal replacement therapy and mortality 95. Beumier M, Roberts JA, Kabtouri H, et al. A new regimen for
in critically ill patients with acute kidney injury. Crit Care. 2009; continuous infusion of vancomycin during continuous renal
13(2):R57. replacement therapy. J Antimicrob Chemother. 2013;68(12):
80. VA/NIH Acute Renal Failure Trial Network, Palevsky PM, Zhang 2859-2865.
JH, O’Connor TZ, et al. Intensity of renal support in critically ill 96. Lodise TP, Lomaestro B, Drusano G. Piperacillin-tazobactam
patients with acute kidney injury. N Engl J Med. 2008;359(1): for Pseudomonas aeruginosa infection: clinical implications of
7-20. an extended infusion dosing strategy. Clin Infect Dis. 2007;
81. RENAL Replacement Therapy Study Investigators, Bellomo R, 44(3):357-363.
Cass A, Cole L, et al. Intensity of continuous renal replacement 97. Kielstein JT, Engbers C, Bode-Boeger SM, et al. Dosing of dap-
therapy in critically ill patients. N Eng J Med. 2009;361(17): tomycin in intensive care unit patients with acute kidney injury
1627-1638. undergoing extended dialysis-a pharmacokinetic study. Nephrol
82. Kielstein JT, David S. Pro: renal replacement trauma or paracel- Dial Transplant. 2010;25(5):1537-1541.
sus 2.0. Nephrol Dial Transplant. 2013;28(11):2728-2733. 98. Mueller BA, Scoville BA. Adding to the armamentarium: anti-
83. Covajes C, Scolletta S, Penaccini L, et al. Continuous infusion of biotic dosing in extended dialysis. Clin J Am Soc Nephrol.
vancomycin in septic patients receiving continuous renal replace- 2012;7(3):373-375.
ment therapy. Int J Antimicrob Agents. 2013;41(3):261-266. 99. Taccone FS, de Backer D, Laterre PF, et al. Pharmacokinetics of
84. Roberts JA, Mehta RL, Lipman J. Sustained low efficiency dialy- a loading dose of amikacin in septic patients undergoing contin-
sis allows rational renal replacement therapy, but dose it allow uous renal replacement therapy. Int J Antimicrob Agents. 2011;
rational drug dosing? Crit Care Med. 2011;39(3):602-603. 37(6):5.
85. Fiaccadori E, Maggiore U, Rotelli C, et al. Removal of linezolid by 100. Truong J, LEvkovich BJ, Padiglione AA. Simple approach to
conventional intermittent hemodialysis, sustained low-efficiency improving vancomycin dosing in intensive care: a standardized
dialysis, or continuous venovenous hemofiltration in patients with loading dose results in earlier therapeutic levels. Intern Med J.
acute renal failure. Crit Care Med. 2004;32(12):2437-2442. 2012;42(1):23-29.
86. Arzuaga A, Maynar J, Gascón AR, et al. Influence of renal func- 101. Kamel Mohamed OH, Wahba IM, Watnick S, et al. Administra-
tion on the pharmacokinetics of piperacillin/tazobactam in inten- tion of tobramycin in the beginning of the hemodialysis session:
sive care unit patients during continuous venovenous a novel intradialytic dosing regimen. Clin J Am Soc Nephrol.
hemofiltration. J Clin Pharmacol. 2005;45(2):168-176. 2007;2(4):694-699.
87. Kielstein JT, Czock D, Schopke T, et al. Pharmacokinetics and 102. Sowinski KM, Magner SJ, Lucksiri A, Scott MK, Hamburger
total elimination of meropenem and vancomycin in intensive care RJ, Mueller BA. Influence of hemodialysis on gentamicin phar-
unit patients undergoing extended daily dialysis. Crit Care Med. macokinetics, removal during hemodialysis, and recommended
2006;34(1):51-56. dosing. Clin J Am Soc Nephrol. 2008;3(2):355-361.
88. Burkhardt O, Joukhadar C, Traunmuller F, Hadem J, Welte T, 103. Veistein A, Venisse N, Badin J, Pinsard M, Robert R, Dupuis A.
Kielstein JT. Elimination of daptomycin in a patient with acute Gentamicin in hemodialyzed critical care patients: early dialysis
renal failure undergoing extended daily dialysis. J Antimicrob after administration of a high dose should be considered. Antimi-
Chemother. 2008;61(1):224-225. crob Agents Chemother. 2013;57(2):977-982.
89. Wilson FP, Berns JS. Vancomycin levels are frequently subther- 104. Roberts JA, Field J, Visser A, et al. Using population phar-
apeutic during continuous venovenous hemodialysis (CVVHD). macokinetics to determine gentamicin dosing during
Clin Nephrol. 2012;77(4):329-331. extended daily diafiltration in critically ill patients with
90. Zoller M, Maier B, Hornuss C, et al. Variability of linezolid con- acute kidney injury. Antimicrob Agents Chemother. 2010;
centrations after standard dosing in critically ill patients: a pro- 54(9):3635-3640.
spective observational study. Crit Care. 2014;18(4):R148. 105. Calbo E, Garau J. Application of pharmacokinetics and phar-
91. Owens RC, Shorr AF. Rational dosing of antimicrobial agents: macodynamics to antimicrobial therapy of community
pharmacokinetic and pharmacodynamics strategies. Am J acquired respiratory tract infections. Respiration. 2005;72(6):
Health-Sys Pharm. 2009;66(4):S23-S30. 561-571.
Lewis and Mueller 175

106. Scaglione F, Paraboni L. Influence of pharmacokinetics/pharma- 120. Gentamicin [package insert]. Lake Forest, IL: Hospira, Inc;
codynamics of antibacterials in their dosing regimen selection. August 2005. http://www.hospira.com/Images/EN-1004_32-
Expert Rev Anti Infect Ther. 2006;4(3):479-490. 5467_1.pdf. Accessed March 12, 2014.
107. McKinnon PS, Paladino JA, Schentag JJ. Evaluation of area 121. Ryan DM, Cars O. A problem in the interpretation of beta-
under the inhibitory curve (AUIC) and time above the minimum lactam antibiotic levels in tissues. J Antimicrob Chemother.
inhibitory concentration (T > MIC) as predictors of outcome for 1983;12(3):281-284.
cefepime and ceftazidime in serious bacterial infections. Int J 122. Schentag JJ, Heller AS, Hardy BG, Wels PB. Antibiotic penetra-
Antimicrob Agents. 2008;31:345-351. tion in liver infection: a case of tobramycin failure responsive to
108. Valtonen M, Tiula, Takkunen O, Backman JT, Neuvonen PJ. moxalactam. Am J Gastroenterol. 1983;78(10):641-644.
Elimination of the piperacillin/tazobactam combination during 123. Joukhadar C, Frossard M, Mayer BX, et al. Impaired target site
continuous venovenous haemofiltration and haemodiafiltration penetration of beta-lactams may account for therapeutic failure
in patients with acute renal failure. J Antimicrob Chemother. in patients with septic shock. Crit Care Med. 2001;29(2):
2001;48(6):881-885. 385-391.
109. Falagas ME, Tansarli GS, Ikawa K, Vardakas KZ. Clinical out- 124. Zeitlinger MA, Erovic BM, Sauermann R, Georgopoulos A,
comes with extended or continuous versus short-term intrave- Müller M, Joukhadar C. Plasma concentrations might lead
nous infusion of carbapenems and piperacillin/tazobactam: a to overestimation of target site activity of piperacillin in
systematic review and meta-analysis. Clin Infec Dis. 2013; patients with sepsis. J Antimicrob Chemother. 2005;56(4):
56(2):272-282. 703-708.
110. Chant C, Leung A, Friedrich JO. Optimal dosing of antibio- 125. Klekner A, Bagyi K, Bognar L, Gaspar A, Andrasi M, Szabo J.
tics in critically ill patients using continuous/extended infu- Effectiveness of cephalosporins in the sputum of patients with
sion: a systematic review and meta-analysis. Crit Care. nosocomial bronchopneumonia. J Clin Microbiol. 2006;44(9):
2013;17(6):R279. 3418-3421.
111. Moriyama B, Henning SA, Neuhauser MM, Danner RL, Walsh TJ. 126. Stolle LB, Plock N, Joukhadar C, et al. Pharmacokinetics of line-
Continuous-infusion beta-lactam antibiotics during continuous zolid in bone tissue investigated by in vivo microdialysis. Scand
venovenous hemofiltration for the treatment of resistant gram- J Infect Dis. 2008;40(1):24-29.
negative bacteria. Ann Pharmacother. 2009;43(7):1324-1337. 127. Tam VH, Gamez EA, Weston JS, et al. Outcomes of bacter-
112. Joy MS, Matzke GR, Frye RF, Palevsky PM. Determinants of emia due to Pseudomonas aeruginosa with reduced suscept-
vancomycin clearance by continuous venovenous hemofiltration ibility to piperacillin-tazobactam: implications on the
and continuous venovenous hemodialysis. Am J Kidney Dis. appropriateness of the resistance breakpoint. Clin Infect Dis.
1998;31(6):1019-1027. 2008;46(6):862-867.
113. DelDot ME, Lipman J, Tett SE. Vancomycin pharmacokinetics 128. Soriano A, Marco F, Martı́nez JA, et al. Influence of vancomycin
in critically ill patients receiving continuous venovenous haemo- minimum inhibitory concentration on the treatment of
diafiltration. Br J Clin Pharmacol. 2004;58(3):259-268. methicillin-resistant Staphylococcus aureus bacteremia. Clin
114. Moore RD, Lietman PS, Smith CR. Clinical response to amino- Infect Dis. 2008;46(2):193-200.
glycoside therapy: importance of the ratio of peak concentration 129. Hidayat LK, Hsu DI, Quist R, Shriner KA, Wong-Beringer A.
to minimal inhibitory concentration. J Infect Dis. 1987;155(1): High-dose vancomycin therapy for methicillin-resistant Staphy-
93-99. lococcus aureus infections: efficacy and toxicity. Arch Intern
115. Martinez MN, Papich MG, Drusano GL. Dosing regimen mat- Med. 2006;166(19):2138-2144.
ters: the importance of early intervention and rapid attainment 130. Rybak MJ, Lomaestro BM, Rotschafer JC, et al. Vancomycin
of the pharmacokinetic/pharmacodynamic target. Antimicrob therapeutic guidelines: a summary of consensus recommenda-
Agents Chemother. 2012;56(6):2795-2805. tions from the Infectious Diseases Society of America, the
116. Jumbe N, Louie A, Leary R, et al. Application of a mathematical American Society of Health-System Pharmacists, and the Soci-
model to prevent in vivo amplification of antibiotic-resistant ety of Infectious Diseases Pharmacist. Clin Infect Dis. 2009;
bacterial populations during therapy. J Clin Invest. 2003; 49(3):325-327.
112(2):275-285. 131. Kullar R, McClellan I, Geriak M, Sakoulas G. Efficacy and
117. Trotman RL, William JC, Shoemaker DM, Salzer WL. Antibiotic safety of daptomycin in patients with renal impairment: a multi-
dosing in critically ill adult patients receiving continuous renal center retrospective analysis. Pharmacotherapy. 2014;34(6):
replacement therapy. Clin Infect Dis. 2005;41(8):1159-1166. 582-589.
118. Matsuo H, Hayashi J, Ono K, et al. Administration of aminogly- 132. National Nosocomial Infections Surveillance System. National
cosides to hemodialysis patients immediately before dialysis: a Nosocomial Infections Surveillance (NNIS) System Report, data
new dosing modality. Antimicrob Agents Chemother. 1997; summary from January 1992 through June 2004, issued October
41(12):2597-2601. 2004. Am J Infect Control. 2004;32(6):470-485.
119. Teigen MM, Duffull S, Dang L, Johnson DW. Dosing 133. Klevens RM, Edwards JR, Gaynes RP. The impact of
of gentamicin in patients with end-stage renal disease antimicrobial-resistant, health care- associated infections on
receiving hemodialysis. J Clin Pharmacol. 2006;46(11): mortality in the United States. Clin Infect Dis. 2008;47(7):
1259-1267. 927-930.
176 Journal of Intensive Care Medicine 31(3)

134. Boucher HW, Talbot GH, Bradley JS, et al. Bad bugs, no drugs: Unit Patients With Pneumonia: Retrospective Analysis of the
no ESKAPE! An update from the Infectious Diseases Society of IMPACT-HAP Database. Clin Ther. 2012;34(1):149-157.
America. Clin Infect Dis. 2009;48(1):1-12. 150. Bertino JS, Booker LA, Franck PA, Jenkins PL, Franck KR, Naf-
135. Peters NK, Dixon DM, Holland SM, Fauci AS. The research ziger AN. Incidence of and significant risk factors for
agenda of the national institute of allergy and infectious diseases aminoglycoside-associated nephrotoxicity in patients dosed by
for antimicrobial resistance. J Infect Dis. 2008;197(8): using individualized pharmacokinetic monitoring. J Infect Dis.
1087-1093. 1993;167(1):173-179.
136. Barth RH, DeVincenzo N. Use of vancomycin in high-flux 151. Rybak MJ, Abate BJ, Kang SL, Ruffing MJ, Lerner SA, Drusano
hemodialysis: experience with 130 courses of therapy. Kidney GL. Prospective evaluation of the effect of an aminoglycoside
Int. 1996;50(3):929-936. dosing regimen on rates of observed nephrotoxicity and ototoxi-
137. Taber SS, Mueller BA. Drug-associated renal dysfunction. Crit city. Antimicrob Agents Chemother. 1999;43(7):1549-1555.
Care Clin. 2006;22(2)357-374. 152. Drusano GL, Ambrose PG, Bhavnani SM, Bertino JS, Nafziger
138. Kane-Gill SL, Kirisci L, Verrico MM, Rothschild JM. Analysis AN, Louie A. Back to the future: using aminoglycosides again
of risk factors for adverse drug events in critically ill patients. and how to dose them optimally. Clin Infect Dis. 2007;45(15):
Crit Care Med. 2012;40(3):823-828. 753-760.
139. Hartzell JD, Neff R, Ake J, et al. Nephrotoxicity associated with 153. Calandra GB, Brown KR, Grad LC, Ahonkhai VI, Wang C, Aziz
intravenous colistin (colistin sodium) treatment at a tertiary care MA. Review of adverse experiences and tolerability in the first
medical center. Clin Infect Dis. 2009;48(12):1724-1728. 2516 patients treated with imipenem/cilastatin. Am J Med.
140. Kwon JA, Lee JE, Huh W, et al. Predictors of acute kidney injury 1985;78(6A):73-78.
associated with intravenous colistin treatment. Int J Antimicrob 154. Hellinger WC, Brewer NS. Carbapenems and monobactams:
Agents. 2010;35(5):473-477. imipenem, meropenem and aztreonam. Mayo Clin Proc. 1999;
141. Schentag JJ, Cerra FB, Plaut ME. Clinical and pharmacokinetic 74(4):420-434.
characteristics of aminoglycoside nephrotoxicity in 201 criti- 155. Norrby SR. Carbapenems in serious infections: a risk benefit
cally ill patients. Antimicrob Agents Chemother. 1982;21(5): assessment. Drug Saf. 2000;22(3):191-194.
721-726. 156. Norrby SR, Newell PA, Faukner KL, Lesky W. Safety profile of
142. Zager RA. Endotoxemia, renal hypoperfusion, and fever: inter- meropenem international clinical experience based on the first
active risk factors for aminoglycoside and sepsis associated 3125 patients treated with meropenem. J Antimicrob Chemother.
acute renal failure. Am J Kid Dis. 1992;20(3):223-230. 2005;36(suppl A):207-223.
143. Van Hal, Paterson DL, Lodise TP. Systematic review and meta- 157. Nicolau DP. Carbapenems: a potent class of antibiotics. Expert
analysis of vancomycin- induced nephrotoxicity associated with Opin Pharmacother. 2008;9(1):23-37.
dosing schedules that maintain troughs between 15 and 20 milli- 158. Martinez-Rodriguez JE, Barriga FJ, Santamaria J, et al.
grams per liter. Antimicrob Agents Chemother. 2013;57(2): Nonconvulsive status epilepticus associated with cephalos-
734-744. porins in patients with renal failure. Am J Med. 2001;
144. Moore RD, Smith CR, Lipsky JJ, Mellits ED, Lietman PS. Risk 111(2):115-119.
factors for nephrotoxicity in patients treated with aminoglyco- 159. Grill MF, Maganti R. Cephalosporin-induced neurotoxicity:
sides. Ann Intern Med. 1984;100(3):352-357. clinical manifestations, potential pathogenic mechanisms, and
145. Lodise TP, Lomaestro B, Graves J, Drusano GL. Larger vanco- the role of electroencephalographic monitoring. Ann Pharmac-
mycin doses (at least four grams per day) are associated with an other. 2008;42(12):1843-1850.
increased incidence of nephrotoxicity. Antimicrob Agents Che- 160. Chapuis TM, Giannoni E, Majcherczyk PA, et al. Prospective
mother. 2008;52(4):1330-1336. monitoring of cefepime in intensive care unit adult patients. Crit
146. Lodise TP, Patel N, Lomaestro BM, Rodvold KA, Drusano Care. 2010;14(2):R51.
GL. Relationship between initial vancomycin concentration 161. Fugate JE, Kalimullah EA, Hocker SE, Clark SL, Wijdicks EF,
time profile and nephrotoxicity. Clin Infect Dis. 2009;49(4): Rabinstein AA. Cefepime neurotoxicity in the intensive care
507-514. unit: a cause of severe, underappreciated encephalopathy. Crit
147. Pritchard L, Baker C, Leggett J, Sehdev P, Brown A, Bayley KB. Care. 2013;17(6):R264.
Increasing vancomycin serum trough concentrations and inci- 162. Bagshaw SM. Short- and long-term survival after acute
dence of nephrotoxicity. Am J Med. 2010;123(12):1143-1149. kidney injury. Nephrol Dial Transplant. 2008;23(7):2126-2128.
148. Bosso JA, Nappi J, Rudisill C, et al. Relationship between van- 163. Schiffl H, Fischer R. Five-year outcomes of severe acute kidney
comycin trough concentrations and nephrotoxicity: a prospec- injury requiring renal replacement therapy. Nephrol Dial Trans-
tive multicenter trial. Antimicrob Agents Chemother. 2011; plant. 2008;23(7):2235-2241.
55(12):5475-5479. 164. Eisenberg JM, Koffer H, Glick HA, et al. What is the cost of
149. Cano EL, Haque NZ, Welch VL, et al. Incidence of Nephrotoxi- nephrotoxicity associated with aminoglycosides? Ann Intern
city and Association With Vancomycin Use in Intensive Care Med. 1987;107(6):900-909.

You might also like