You are on page 1of 29

ASHP REPORT

Therapeutic monitoring of vancomycin for serious


­methicillin-resistant Staphylococcus aureus infections:
A revised consensus guideline and review by the American
Society of Health-System Pharmacists, the Infectious Diseases
Society of America, the Pediatric Infectious Diseases Society,
and the Society of Infectious Diseases Pharmacists

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


An audio interview that
supplements the informa-
tion in this article is avail-
T  he first consensus guideline for ther-
apeutic monitoring of vancomycin
in adult patients was published in 2009.
Staphylococcus aureus (MRSA) infec-
tions. It should be noted, however, that
when the recommendations were orig-
able on AJHP’s website at
www.ajhpvoices.org.
A  committee representing 3 organiza- inally published, there were important
tions (the American Society for Health- issues not addressed and gaps in know-
Am J Health-Syst Pharm. 2020;77:835-864 System Pharmacists [ASHP], Infectious ledge that could not be covered ade-
Michael J. Rybak, PharmD, MPH, PhD, FCCP, Diseases Society of America [IDSA], quately because of insufficient data.
FIDP, FIDSA, Anti-Infective Research Laboratory,
Department of Pharmacy Practice, Eugene and Society for Infectious Diseases In fact, adequate data were not avail-
Applebaum College of Pharmacy & Health Sciences, Pharmacists [SIDP]) searched and re- able to make recommendations in the
Wayne State University, Detroit, MI, School of
Medicine, Wayne State University, Detroit, MI, and viewed all relevant peer-reviewed data original guideline for specific dosing
Detroit Receiving Hospital, Detroit, MI
Jennifer Le, PharmD, MAS, FIDSA, FCCP,
on vancomycin as it related to in vitro and monitoring for pediatric patients
FCSHP, BCPS-AQ ID, Skaggs School of Pharmacy and in vivo pharmacokinetic and phar- outside of the neonatal age group; spe-
and Pharmaceutical Sciences, University of
California San Diego, La Jolla, CA macodynamic (PK/PD) characteristics, cific recommendations for vancomycin
Thomas P. Lodise, PharmD, PhD, Albany College including information on clinical effi- dosage adjustment and monitoring
of Pharmacy and Health Sciences, Albany, NY, and
Stratton VA Medical Center, Albany, NY cacy, toxicity, and vancomycin resistance in the morbidly obese patient popu-
Donald P. Levine, MD, FACP, FIDSA, School of in relation to serum drug concentration lation and patients with renal failure,
Medicine, Wayne State University, Detroit, MI, and
Detroit Receiving Hospital, Detroit, MI and monitoring. The data were summar- including specific dialysis dosage ad-
John S. Bradley, MD, JSB, FIDSA, FAAP, ized, and specific dosing and monitoring justments; recommendations for the
FPIDS, Department of Pediatrics, Division of
Infectious Diseases, University of California at San recommendations were made. The pri- use of prolonged or continuous in-
Diego, La Jolla, CA, and Rady Children’s Hospital
San Diego, San Diego, CA mary recommendations consisted of fusion (CI) vancomycin therapy; and
Catherine Liu, MD, FIDSA, Division of Allergy and eliminating routine monitoring of serum safety data on the use of dosages that
Infectious Diseases, University of Washington, Seattle,
WA, and Vaccine and Infectious Disease Division, Fred peak concentrations, emphasizing a ratio exceed 3  g per day. In addition, there
Hutchinson Cancer Research Center, Seattle, WA
of area under the curve over 24 hours were minimal to no data on the safety
Bruce A. Mueller, PharmD, FCCP, FASN, FNKF,
University of Michigan College of Pharmacy, Ann to minimum inhibitory concentration and efficacy of targeted trough concen-
Arbor, MI
(AUC/MIC) of ≥400 as the primary PK/ trations of 15 to 20 mg/L.
Manjunath P. Pai, PharmD, FCCP, University of
Michigan College of Pharmacy, Ann Arbor, MI PD predictor of vancomycin activity, and This consensus revision evaluates
Annie Wong-Beringer, PharmD, FCCP, FIDSA, promoting serum trough concentrations
University of Southern California School of
the current scientific data and contro-
Pharmacy, Los Angeles, CA of 15 to 20  mg/L as a surrogate marker versies associated with vancomycin
John C. Rotschafer, PharmD, FCCP, University
of Minnesota College of Pharmacy, Minneapolis, MN
for the optimal vancomycin AUC/MIC dosing and serum concentration moni-
Keith A. Rodvold, PharmD, FCCP, FIDSA, if the MIC was ≤1 mg/L in patients with toring for serious MRSA infections (in-
University of Illinois College of Pharmacy, Chicago, IL
normal renal function. The guideline also cluding but not limited to bacteremia,
Holly D. Maples, PharmD, University of Arkansas
for Medical Sciences College of Pharmacy & recommended, albeit with limited data sepsis, infective endocarditis, pneu-
Arkansas Children’s Hospital, Little Rock, AR
support, that actual body weight be used monia, osteomyelitis, and meningitis)
Benjamin M. Lomaestro, PharmD, Albany
Medical Center Hospital, Albany, NY to determine the vancomycin dosage and and provides new recommendations
Address correspondence to Dr. Rybak loading doses for severe infections in pa- based on recent available evidence.
(m.rybak@wayne.edu).
tients who were seriously ill.1 Due to a lack of data to guide appro-
Keywords: nephrotoxicity, pharmacokinetics Since those recommendations were priate targets, the development of this
and pharmacodynamics, target attainment,
vancomycin, vancomycin consensus guideline generated, a number of publications guideline excluded evaluation of van-
© American Society of Health-System have evaluated the impact of the 2009 comycin for methicillin-susceptible
Pharmacists 2020. All rights reserved. guidelines on clinical efficacy and tox- S.  aureus (MSSA) strains, coagulase-
For permissions, please e-mail: journals.
permissions@oup.com. icity in patients receiving vancomycin negative staphylococci, and other
DOI 10.1093/ajhp/zxaa036 for the treatment of methicillin-resistant pathogens; thus, the extrapolation of

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  835


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

guideline recommendations to these


Table 1. Grading System for Recommendations Based on Quality of
pathogens should be viewed with ex-
Evidencea
treme caution. Furthermore, serious
invasive MRSA infections exclude Category and Grade Definition
nonbacteremic skin and skin structure
Strength of
and urinary tract infections. Since this
recommendation
guideline focuses on optimization of
vancomycin dosing and monitoring, A Good evidence to support a recommendation for or
against use
recommendations on the appropriate-
ness of vancomycin use, combination B Moderate evidence to support a recommendation for
or against use
or alternative antibiotic therapy, and
multiple medical interventions that C Poor evidence to support a recommendation

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


may be necessary for successful treat- Quality of evidence
ment of invasive MRSA infections are
I Evidence from 1 or more properly randomized con-
beyond the scope of this guideline and trolled trials
will not be presented.
II Evidence from 1 or more well-designed clinical trials,
without randomization; from cohort or case-
controlled analytic studies (preferably from more
Methods
than 1 center); from multiple time-series; or from
These are the consensus state- dramatic results from uncontrolled experiments
ments and guideline of ASHP, IDSA, the III Evidence from opinions of respected authorities,
Pediatric Infectious Diseases Society based on clinical experience, descriptive studies, or
(PIDS), and SIDP. Guideline panel com- reports of expert committees
position consisted of physicians, phar- Adapted from the Canadian Task Force on the Periodic Health Examination.2
a

macists, and a clinical pharmacologist


with expertise in clinical practice and/or
research with vancomycin. Committee Potential limitations of this review MICBMD ratio approximates or exceeds
members were assigned key topics re- included the fact that there are few 400. Furthermore, in vitro data sug-
garding vancomycin dosing and moni- published randomized clinical trials gest that an AUC of <400 potentiates
toring. A  draft document addressing of vancomycin dosing and monitoring the emergence of MRSA resistance and
these specific areas was reviewed by all available in the literature. Most pub- vancomycin-intermediate S.  aureus
committee members and made avail- lished studies evaluating vancomycin strains.8,9 There are also mounting clin-
able for public comments for 30  days dosing, dosage adjustment, and moni- ical data, albeit mostly retrospective in
through ASHP, IDSA, PIDS, and SIDP. toring were retrospective PK or PD nature, in support of this PK/PD target
The committee then met to review and clinical assessments or retrospective for vancomycin.10-18 A summary of these
revise the document based on the sub- observational studies in patients with investigations and their findings can be
mitted comments, suggestions, and MRSA infections. found in eTable 1.10-17,19-23
recommendations. After careful discus- PK/PD efficacy targets.  To
sion and consideration, the document optimize the dosing of any antimicro- Clinical PK/PD Data: Adults
was revised and circulated among the bial agent, a firm understanding of the While an AUC/MICBMD ratio of ≥400
committee and supporting organiza- drug’s exposure-effect and exposure- is currently considered the optimal
tions prior to final approval and publi- toxicity links are required. While a va- PK/PD “efficacy” target, it is important
cation. A search of PubMed and Embase riety of PD indices for vancomycin have to recognize that this target has been
was conducted using the following been suggested, an AUC/MIC ratio of largely derived from retrospective,
search terms: vancomycin, pharmacoki- ≥400 (with the MIC determined by broth single-center, observational studies of
netics, pharmacodynamics, efficacy, re- microdilution [BMD]) is the current ac- patients with MRSA bloodstream infec-
sistance, toxicity, obesity, and pediatrics. cepted critical PK/PD index in light tions.11-17 It is also important to recog-
All relevant and available peer-reviewed of our limited experience and studies nize that most of the landmark clinical
studies in the English-language litera- evaluating AUC/MIC values of <400.1,3-7 studies that established the contem-
ture published from 1958 through 2019 In vitro and in vivo assessments of PK/ porary PK/PD efficacy target relied on
were considered. Studies were rated by PD models applicable to human MRSA simple vancomycin clearance (CL) for-
their quality of evidence, and the subse- infection have found that bactericidal mulas based on daily vancomycin dose
quent recommendations were graded activity (ie, a 1- to 2-log reduction in bac- and estimated renal function to deter-
using the classification schemata de- terial inoculum in the animal model) is mine AUC values.10,11,13 Current evalu-
scribed in Table 1. achieved when the vancomycin AUC/ ation of these data demonstrates that

836    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

these CL formulas provide imprecise (bioMérieux USA, Hazelwood, MO) data for a MIC of 2 mg/L are limited, sug-
estimates of the AUC.24-26 This finding method, to arrive at an AUC/MICEtest gesting the need for more studies to ascer-
is not surprising, as there is consid- value.12,14,15 The MICEtest value tends to tain the optimal AUC/MIC target for this
erable interpatient variability in van- be 1.5- to 2-fold higher than the MICBMD MIC value or consideration for the use of
comycin exposure profiles in clinical value; therefore, it is likely that the alternative antibiotics. The currently avail-
practice, and it is not possible to gen- AUC threshold needed for response able data also highlight the critical need
erate valid estimates of exposure vari- from these 3 studies,12,14,15 if calculated for large-scale, multicenter, randomized,
ables in a given individual based on CL using the MICBMD, would align with the vancomycin dose–optimized clinical out-
formulas that are derived from glomer- studies by Lodise et  al16 and Casapao comes trials. As data from future prospec-
ular filtration rate estimation equations et al.17 tive, multicenter clinical studies emerge, it
alone.10,11,13 In most cases, the formula- In an effort to surmount the is important that clinicians recognize that
based approach will overestimate van- limitations­associated with previous­ our current understanding of the PK/PD

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


comycin CL by approximately 40% to single-center, retrospective vancomy­ target associated with maximal effect and
50%.16 cin­exposure-response clinical ana- toxicity is subject to change, and this may
While it has been cumbersome to lyses, a multicenter, observational ultimately alter the current way we dose
estimate AUC in the clinical setting prospective study was performed to vancomycin to optimize effect and mini-
in the past, Neely and colleagues24 re- evaluate the relationship between the mize toxicity.
cently demonstrated that Bayesian soft- prespecified day 2 AUC/MIC ratios (ie,
ware programs (refer to Therapeutic AUC/MICEtest of ≥320 and AUC/MICBMD Toxicodynamics: AKI
Monitoring section) can be used to gen- of ≥650) and outcomes in adult pa- A major concern with vancomycin
erate accurate and reliable estimates of tients (n = 265) with MRSA bacteremia. use is the occurrence of AKI. While
the daily AUC values with trough-only In the multivariate analyses, treatment multiple definitions of vancomycin-
PK sampling. However, the accuracy failure rates were not significantly dif- associated AKI have been employed in
of AUC estimation is higher with peak ferent between the prespecified day 2 the literature, most studies defined it
and trough measurements compared AUC/MIC groups. Post hoc global out- as an increase in the serum creatinine
to trough-only PK sampling.24 Using comes analyses suggested that patients (SCr) level of ≥0.5 mg/dL, or a 50% in-
this validated Bayesian method to es- in the 2 lowest AUC exposure quintiles crease from baseline in consecutive
timate the daily AUC in a single-center, (ie, those with an AUC of ≤515 mg·h/L) daily readings, or a decrease in calcu-
retrospective study of patients with experienced the best global outcome lated creatinine CL (CLcr) of 50% from
MRSA bloodstream infections, Lodise (defined as absence of both treatment baseline on 2 consecutive days in the
and colleagues16 found that outcomes failure and acute kidney injury [AKI]). absence of an alternative explanation.1
were maximized when day 1 and day While global outcomes were similar Recently, it has been proposed that a
2 AUC/MICBMD ratios exceeded 521 in the 2 lowest AUC-exposure quin- more sensitive threshold (ie, an increase
and 650, respectively. Employing the tiles, only 20% of the study population in SCr of ≥0.3 mg/dL over a 48-hour pe-
same Bayesian approach to estimate (n  =  54) had an AUC of ≤400 mg·h/L, riod) may be considered as an indicator
daily AUC values, Casapao and col- and it is unclear if efficacy outcomes of vancomycin-associated AKI. This
leagues17 also noted that the risk of are maintained at an AUC less than threshold was adopted from the Acute
vancomycin treatment failure among this threshold of 400 mg·h/L.23 Notably, Kidney Injury Network (AKIN) and
patients with MRSA infective endo- the higher AUC value cited above (515 the Kidney Disease: Improving Global
carditis was greatest among those mg·h/L) provides a new index that in- Outcomes (KDIGO) criteria.27-29 The
with an AUC/MICBMD ratio of ≤600 and corporates both efficacy and AKI that incidence of vancomycin-associated
that this exposure-failure relationship is still within the recommended AUC AKI has varied across published
persisted after adjusting for factors range of 400 to 600 mg·h/L (assuming a studies. In a meta-analysis by van Hal
such as intensive care unit (ICU) ad- MIC of 1 mg/L). and colleagues,29 the prevalence of
mission, presence of heteroresistant Collectively, recent studies highlight vancomycin-associated AKI varied
vancomycin-intermediate S.  aureus, the importance of generating valid esti- from 5% to 43%. Similarly, a recent
and other comorbidities. In contrast to mates of the AUC values through Bayesian meta-analysis of 13 studies by Sinha
the studies by Lodise et al and Casapao modeling techniques when conducting Ray et  al30 reported that the relative
et  al, several small-scale, retrospec- vancomycin exposure-outcomes analyses risk of AKI with vancomycin was 2.45
tive clinical evaluations of vancomycin in patients. Current vancomycin exposure- (95% confidence interval, 1.69-3.55),
exposure-response reported lower effectiveness data originated largely from with an attributable risk of 59%. Most
Bayesian-derived thresholds for AUC/ studies of MRSA bacteremia, with some episodes of AKI developed between 4
MIC since the AUC was measured at studies for pneumonia and infective en- and 17  days after initiation of therapy.
steady-state conditions and indexed docarditis and none for osteomyelitis Many patients, especially those who are
to the MIC, as determined by the Etest and meningitis. Furthermore, outcomes critically ill, do not fully recover renal

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  837


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

function after AKI,31 and even mild AKI increased along the day 2 AUC con- (i.v.) contrast dye, and vasopressors
can significantly decrease long-term tinuum in a stepwise manner and that has been shown to increase the risk of
survival rates, increase morbidity, pro- patients with day 2 AUC values of ≥793 nephrotoxicity. Recently, piperacillin/
long hospitalizations, and escalate mg·h/L were at the greatest risk for AKI.23 tazobactam and flucloxacillin have
healthcare costs.22,32 Given the understanding about po- been reported to increase the risk
With any drug, an understanding of tential toxic concentrations, there are for AKI in patients receiving vanco-
its toxicodynamic profile is required for also data to suggest that AUC-guided mycin.39-44 It is unclear if the threshold
optimal dosing. Several studies, largely vancomycin dosing may reduce the oc- for vancomycin-induced AKI varies
retrospective in nature, have attempted currence of vancomycin-associated AKI. according to these covariates, but clin-
to quantify the relationship between In a retrospective, quasi-experimental icians should be mindful of the poten-
vancomycin exposure and probability study of 1,280 hospitalized patients, tial for additional risk when prescribing
of AKI.33,34 Although data are limited, Finch et al20 compared the incidence of vancomycin to patients when these

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


the collective literature suggests that nephrotoxicity in patients monitored by conditions are present.34,40-50
the risk of AKI increases as a function individualized AUC vs trough concen- Based on the current best avail-
of the trough concentration, espe- tration. AUC-guided dosing was found able evidence, daily vancomycin AUC
cially when maintained above 15 to to be independently associated with a values (assuming a MIC of 1  mg/L)
20  mg/L.29 Similarly, there are recent significant decrease in AKI (odds ratio should be maintained between 400 and
data to suggest that the risk of AKI in- [OR], 0.52; 95% CI, 0.34-0.80; P = 0.003).20 600 mg·h/L to minimize the likelihood
creases along the vancomycin AUC Median Bayesian-estimated AUC was of nephrotoxicity and maximize effi-
continuum, especially when the daily significantly lower with AUC-guided cacy for suspected or definitive serious
AUC exceeds 650 to 1,300 mg·h/L.24,33-35 dosing vs trough monitoring (474 [SD, invasive MRSA infections. Once culture
Furthermore, animal studies corrob- 360-611] mg·h/L vs. 705 [SD, 540-883] results or the clinical presentation rule
orate the finding that increased AUC mg·h/L; P  <  0.001). In the prospective out invasive MRSA infection, the em-
rather than trough concentration is a study by Neely et al,22 252 patients were piric use of vancomycin at guideline-
strong predictor of AKI.36,37 monitored via troughs of 10 to 20 mg/L recommended exposures should be
Suzuki et  al33 evaluated the mean in year 1 vs Bayesian-estimated AUC de-escalated, either by a decrease in
vancomycin AUC in relation to AKI. values of ≥400 mg·h/L in years 2 and 3 vancomycin exposure or initiation of
Most patients who developed AKI of the investigation. Nephrotoxicity oc- alternative antibiotics. Extrapolation
had AUC values between 600 and 800 curred in 8% of subjects in year 1 and of guideline recommendations to non-
mg·h/L, compared with 400 to 600 in 0% and 2% of subjects in years 2 and invasive MRSA and other pathogens
mg·h/L in those without AKI (P = 0.014). 3, respectively (P  =  0.01). The median should be viewed with extreme caution.
Furthermore, Lodise and colleagues34 trough concentration and AUC values
showed that the probability of AKI in- associated with AKI were 15.7 mg/L and Therapeutic Monitoring
creased 2.5-fold among patients with 625 mg·h/L, as compared with values Therapeutic monitoring has cen-
AUC values above 1,300 mg·h/L com- of 8.7 mg/L and 423 mg·h/L in subjects tered on maintaining trough con-
pared with patients with lower values without AKI (P = 0.02).22 centrations between 15 and 20  mg/L
(30.8% vs 13.1%, P = 0.02). Although AUC Collectively, the published clinical for serious infections due to MRSA.
values above 1,300 mg·h/L were associ- exposure-response analyses suggest Previous expert guidelines recom-
ated with a substantial increase in AKI, that a daily AUC of ≥400 is the driver mended monitoring trough concen-
an AUC exposure-response relationship of effectiveness and that the risk of AKI trations as a surrogate marker for the
appeared to exist, and the probability of is related to AUC and trough values. AUC/MIC ratio based on the historical
a nephrotoxic event increased as a func- More importantly, these data provide difficulty in estimating the AUC in clin-
tion of the daily AUC and patient’s body the foundation for the current under- ical practice.1,5 In the past, calculation
weight.38 A study by Zasowski et al21 also standing of the therapeutic window of AUC in clinical practice involved col-
reported a similar relationship between for vancomycin. When evaluating the lection of multiple vancomycin serum
Bayesian-estimated vancomycin AUC toxicodynamics of vancomycin, it is im- concentrations during the same dosing
thresholds and AKI in 323 patients; AUC portant to recognize other factors that interval, with subsequent use of PK
values of ≥1,218 mg·h/L for 0 to 48 hours, may complicate or exacerbate the risk software that was not readily available
≥677 for 0 to 24 hours, and ≥683 for 24 of AKI. Host-related factors associated at all institutions. As such, the guide-
to 48 hours or troughs of ≥18.2  mg/L with nephrotoxicity include increased line viewed trough-directed dosing as a
were associated with a 3- to 4-fold in- weight, pre-existing renal dysfunc- more practical alternative to AUC/MIC-
creased risk of nephrotoxicity. Similarly, tion, and critical illness. Concurrent guided dosing in clinical practice.
the aforementioned multicenter, pro- administration of nephrotoxic agents Although the recommendation
spective study of patients with MRSA such as aminoglycosides, loop di­­ to maintain trough values between
bloodstream infections found that AKI uretics, amphotericin B, intravenous 15 and 20  mg/L for serious infections

838    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

due to MRSA has been well integrated First, the range of vancomycin MIC based on the way the drug behaved
into practice, the clinical benefits values among contemporary MRSA in a population of prior patients
of maintaining higher vancomycin isolates is narrow, and the BMD MIC90 (the Bayesian prior) and the revised
trough values have not been well docu- in most institutions is 1 mg/L or less.58-62 probability distribution of a specific
mented.38,51-55 From a PK/PD perspec- Second, measurement of MIC values is patient’s PK parameter values using
tive, it is not surprising that there are imprecise, with dilution of ±1 log2 and exact dosing and drug concentration
limited clinical data to support the variation of 10% to 20% considered ac- data (the Bayesian conditional poste-
range of 15 to 20 mg/L. Recent studies ceptable; therefore, the variability of rior). In short, Bayesian dose optimi-
have demonstrated that trough values reported MIC values encountered in zation software uses a well-developed
may not be an optimal surrogate for routine clinical practice is likely to re- vancomycin population PK model as
AUC values.26,56,57 While trough attain- flect measurement error.63 Third, there the Bayesian prior, together with the
ment ensures achievement of a min- is a high degree of variability between individual patient’s observed drug

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


imum cumulative exposure, a wide commercially available MIC testing concentrations in the data file, to cal-
range of concentration-time profiles methods relative to the BMD method culate a Bayesian posterior parameter
can result in an identical trough value. (see Vancomycin Susceptibility Testing value distribution for that patient.
Patel et  al26 reported a wide range of section). Last, MIC results are typically The dose optimization software then
AUC values from several different not available within the first 72 hours calculates the optimal dosing reg-
dosing regimens yielding similar trough of index culture collection, yet cur- imen based on the specific patient’s
values. The therapeutic discordance rent data indicate that the vancomycin profile.65-67
between trough and AUC values is not AUC/MIC ratio needs to be optimized An advantage of the Bayesian ap-
surprising, as the AUC is the integrated early in the course of infection. proach is that vancomycin concentra-
quantity of cumulative drug exposure Daily AUC values (assuming a tions can be collected within the first 24
(ie, the serum drug concentration–time MICBMD of 1  mg/L) should be main- to 48 hours rather than at steady-state
curve over a defined interval). In con- tained between 400 and 600 mg·h/L conditions (after the third or fourth
trast, the trough represents a single ex- to maximize efficacy and minimize dose), and this information can be used
posure point at the end of the dosing the likelihood of AKI. In the past, AUC to inform subsequent dosing (adap-
interval. In clinical practice, monitoring monitoring required the collection of tive feedback control). As part of their
of trough concentrations will translate multiple concentrations over the same output, Bayesian dosing programs pro-
into achievement of one specific min- dosing interval. With these data, a clini- vide innovative treatment schemes,
imum daily AUC value, whereas the cian would calculate the AUC using the such as front-loading doses with subse-
24-hour AUC (AUC24) largely represents linear-trapezoid rule. This approach quent transition to a lower maintenance
the average concentration during that required precise collection of vanco- dosing regimen, to rapidly achieve
time period [AUC24 (mg·h/L) = average mycin concentrations, making it largely target concentrations within the first 24
concentration (mg/L) x 24 (hours)]. For impractical outside of a research set- to 48 hours among critically ill patients.
troughs of 15 to 20 mg/L, this typically ting. However, this is no longer the case. The Bayesian approach also provides
equates to a daily AUC in excess of 400 It is now possible to accurately estimate the ability to integrate covariates, such
mg·h/L. However, there is considerable the AUC with limited PK sampling. as CLcr, in the structural PK models (the
variability in the upper range of AUC One such approach involves the use Bayesian prior density file) that account
values associated with a given trough of Bayesian software programs to esti- for the pathophysiological changes that
value. Although trough-only moni- mate the vancomycin AUC value with readily occur in critically ill patients.
toring is practical, the potential limita- minimal PK sampling (ie, 1 or 2 van- Incorporation of covariates that account
tions surrounding the practice suggest comycin concentrations) and provide for these “dynamic” changes serves
that trough monitoring may be insuffi- AUC-guided dosing recommendations as a way to identify dosing schemes
cient to guide vancomycin dosing in all in real time. An alternative approach that optimize effect and predict future
patients. involves use of 2 concentrations (peak dosing in a patient who has an evolving
Although the AUC/MIC ratio is con- and trough) and simple analytic PK PK profile.67
sidered the PK/PD driver of efficacy for equations to estimate AUC values.57,64 Bayesian dose-optimizing software
vancomycin, clinicians trying to opti- Bayesian-derived AUC moni- programs are now readily available and
mize vancomycin treatment for patients toring. Bayesian-guided dosing is can be used in real time to identify the
with serious MRSA infections may be based in part on Bayes’ Theorem, as it optimal vancomycin dosage that readily
best advised to use AUC-guided dosing quantifies the sequential relationship achieves the AUC target (assuming
and assume a MICBMD of 1 mg/L (unless between the estimated probability a MICBMD of 1  mg/L).66,68 Bayesian
it is known, through BMD, to be greater distribution of an individual patient’s programs offer numerous advantages
or less than 1 mg/L). The MIC value is PK parameter values (eg, volume [Vd] over the traditional first-order equa-
of less importance for several reasons. or CL) prior to administering the drug tion software programs. Using richly

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  839


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

sampled vancomycin PK data from proportional changes in observed the dosing interval (ie, 12 vs 24 hours)
3 studies comprising 47 adults with AUC24.6,71-73 The major limitation of this than steady-state conditions. Given
varying renal function, Neely and col- approach is that it is not adaptive like the importance of early, appropriate
leagues24 demonstrated that Bayesian the Bayesian approach, as it can only therapy,74 targeted AUC exposures
software programs, embedded with a provide a snapshot of the AUC for the should be achieved early during the
PK model based on richly sampled van- sampling period. As such, this AUC cal- course of therapy, preferably within
comycin data as the Bayesian prior, can culation will not be correct if a physio- the first 24 to 48 hours. If monitoring
be used to generate accurate and reli- logic change such as renal dysfunction is initiated after the first dose, the con-
able estimates of the daily AUC values occurs during or after the sampling tribution of the loading dose to the
with trough-only PK sampling. Of note, period. Furthermore, it is extremely dif- actual AUC may vary depending on
there was limited inclusion of special ficult to estimate the vancomycin AUC24 the magnitude of the loading dose vs
populations in this study, and it is un- with the equation-based method in pa- maintenance doses. The decision to

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


clear if this trough-only Bayesian AUC tients who receive multiple dosing re- delay therapeutic monitoring beyond
estimation approach can be applied gimens within a 24-hour period. If the 48 hours should be based on severity of
to obese patients, critically ill patients, vancomycin dosing interval is more infection and clinical judgment.
pediatric patients, and patients with frequent than once a day, the AUC24 will
unstable renal function. A  random- be a function of the number of identical Summary and
ized controlled study of 65 subjects by doses administered during that interval recommendations:
Al-Sulaiti et al69 showed that estimating (eg, AUC must be multiplied by 2 for a
AUC using both peak and trough con- 12-hour dosing interval to calculate the 1. In patients with suspected or definitive
centrations (vs trough-only estimates) true AUC24). It is also highly preferred serious MRSA infections, an individu-
may improve vancomycin-associated that concentrations are collected near alized target of the AUC/MICBMD ratio
therapeutic cure. Until more data are steady-state conditions. of 400 to 600 (assuming a vancomycin
available, it is preferred to estimate the Despite its drawbacks, this es- MICBMD of 1 mg/L) should be advo-
Bayesian AUC using 2 vancomycin con- timate of AUC is a clear step above cated to achieve clinical efficacy while
centrations (peak and trough). trough-only or peak-only concentra- improving patient safety (A-II). Doses
First-order PK analytic equa- tion interpretation and is familiar to of 15 to 20 mg/kg (based on actual
tions.   Alternatively, the AUC can be most clinicians. Several large medical body weight) administered every 8 to
accurately estimated based on the col- centers within the United States have 12 hours as an intermittent infusion
lection of 2 timed steady-state serum already adopted this approach of ac- are recommended for most patients
vancomycin concentrations and the quiring 2 postdose serum concentra- with normal renal function when as-
use of first-order PK equations.57 The tion estimates of the AUC to perform suming a MICBMD of 1 mg/L (A-II). In
equations used to compute AUC from 2 routine vancomycin dosing and moni- patients with normal renal function,
samples are based in part on an original toring and have demonstrated a con- these doses may not achieve the thera-
approach proposed by Begg, Barclay, siderable improvement in safety over peutic AUC/MIC target when the MIC
and Duffull70 for aminoglycosides the current trough-only concentration is 2 mg/L.
and modified by Pai and Rodvold.57 It monitoring method.37,64 2. Given the narrow vancomycin AUC
is preferred that a near steady-state, PK sampling time.  Timing of range for therapeutic effect and
postdistributional peak (1 to 2 hours achievement of targeted AUC values minimal AKI risk, the most accurate
after end of infusion) and trough con- (assuming a MICBMD of 1  mg/L) re- and optimal way to manage van-
centrations within the same dosing mains unclear. The early AUC/MIC comycin dosing should be through
interval (if possible) are used when target ratios derived in animal models AUC-guided dosing and monitoring
estimating the AUC with the equation- were based on the AUC value from 0 to (A-II). We recommend to accomplish
based methods. 24 hours.3,4 More recent clinical assess- this in one of two ways.
The major advantage of this ap- ments that identified a link between a. One approach relies on the collec-
proach is that it is simpler and re- AUC/MIC ratio and outcomes also as- tion of 2 concentrations (obtained
lies on fewer assumptions than the sessed the AUC values achieved early near steady-state, postdistributional
Bayesian approach. The first-order PK in the course of therapy.1,3,5-7,10,13,20-22 The peak concentration [Cmax] at 1 to
equations used to estimate the AUC 2009 vancomycin guideline stated that 2 hours after infusion and trough
are also familiar to most clinicians, the trough should be assessed prior concentration [Cmin] at the end of
facilitating ease of use in practice. to steady-state conditions (ie, prior to the dosing interval), preferably but
Once the AUC24 is estimated, the clini- the fourth dose).1,5 In fact, steady-state not required during the same dosing
cian simply revises the total daily dose conditions are difficult to determine interval (if possible) and utilizing
to achieve the desired AUC24, as alter- in clinical practice, and the timing of first-order PK equations to estimate
ations of total daily dose will provide the fourth dose is more dependent on the AUC (A-II).

840    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

b. The preferred approach to monitor MICBMD of 1 mg/L unless it is known variability in MIC results between the
AUC involves the use of Bayesian to be greater or less than 1 mg/L susceptibility testing methods.
software programs, embedded with by BMD). Independent of MRSA The challenge is that, according
a PK model based on richly sampled infection, vancomycin monitoring to the Clinical Laboratory Standards
vancomycin data as the Bayesian is also recommended for all pa- Institute (CLSI), acceptable variability
prior, to optimize the delivery of tients at high risk for nephrotoxicity for MIC measurement methods is within
vancomycin based on the collection (eg, critically ill patients receiving ±1 doubling dilution (essential agree-
of 1 or 2 vancomycin concentrations, concurrent nephrotoxins), patients ment), such that current susceptibility
with at least 1 trough. It is preferred with unstable (ie, deteriorating or testing methods are unable, with high
to obtain 2 PK samples (ie, at 1 to 2 significantly improving) renal func- reproducibility, to distinguish MICs
hours post infusion and at end of the tion, and those receiving prolonged of 1  mg/L from MICs of 0.5  mg/L or
dosing interval) to estimate the AUC courses of therapy (more than 3 to 2  mg/L. Most institutions routinely

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


with the Bayesian approach (A-II). 5 days). We suggest the frequency perform MIC testing using automa­
A trough concentration alone may of monitoring be based on clinical ted systems: BD Phoenix (BD, Franklin
be sufficient to estimate the AUC judgment; frequent or daily moni- Lakes, NJ), MicroScan WalkAway
with the Bayesian approach in some toring may be prudent for hemo- (Beckman Coulter, Brea, CA), or Vitek
patients, but more data are needed dynamically unstable patients (eg, 2 (bioMérieux), and in some cases the
across different patient populations those with end-stage renal disease), Etest methodology (bioMérieux). In a
to confirm the viability of using with once-weekly monitoring for study of 161 MRSA blood isolates, when
trough-only data (B-II). hemodynamically stable patients using the essential agreement defini-
3. When transitioning to AUC/MIC (B-II). tion of ±1 log2 dilution error, Vitek 2 and
monitoring, clinicians should con- MicroScan WalkAway demonstrated a
servatively target AUC values for 96.3% agreement with BMD, whereas
patients with suspected or docu- Vancomycin Susceptibility BD Phoenix demonstrated an 88.8%
mented serious infections due to Testing agreement.77 The Etest method had the
MRSA assuming a vancomycin With the MIC being a component lowest agreement with BMD, at 76.4%
MICBMD of 1 mg/L or less at most of the vancomycin AUC/MIC targeted (results were consistently higher by 1
institutions. Given the importance of surrogate for efficacy, it is important to 2 dilutions). The Etest will likely pro-
early, appropriate therapy, vanco- to be aware of local and national van- duce a higher value (0.5 to 2 dilutions
mycin targeted exposure should be comycin susceptibility patterns for higher) than BMD. In another study,
achieved early during the course of MRSA. Although in some centers there 92% of the strains were demonstrated
therapy, preferably within the first 24 has been a steady increase in the av- to have a vancomycin MIC of 1 mg/L by
to 48 hours (A-II). As such, the use of erage vancomycin MIC over several BMD; corresponding figures were 70%
Bayesian-derived AUC monitoring decades, recent national and inter- for MicroScan WalkAway and Etest and
may be prudent in these cases since it national studies that have evaluated 41% for Vitek 1.78
does not require steady-state serum MRSA susceptibility to glycopeptides, Rybak et al79 compared MicroScan
vancomycin concentrations to allow lipopeptides, and beta-lactams have WalkAway, Vitek 2, BD Phoenix, and
for early assessment of AUC target demonstrated that vancomycin MICs Etest to BMD methods among 200
attainment. have remained constant over time, with MRSA strains. In contrast to previous
4. Trough-only monitoring, with a a MIC of ≤1 mg/L demonstrated for studies, these investigators used an
target of 15 to 20 mg/L, is no longer more than 90% of isolates.58-62 A  meta- absolute agreement definition of ±0
recommended based on efficacy analysis of 29,234 MRSA strains from log2 dilution error to better charac-
and nephrotoxicity data in patients 55 studies revealed that the MIC deter- terize the precision. Using this def-
with serious infections due to MRSA minations performed by BMD, Etest, inition, results with BD Phoenix
(A-II). There is insufficient evidence and automated systems were predomi- and MicroScan WalkAway had the
to provide recommendations on nately 1 mg/L and that there was no ev- highest agreement with BMD (66.2%
whether trough-only or AUC-guided idence of a MIC creep phenomenon.75 and 61.8%, respectively), followed
vancomycin monitoring should be Furthermore, a global surveillance pro- by Vitek 2 (54.3%). As noted above,
used among patients with noninva- gram reported that 95% of 57,319 MRSA Etest tended to produce results that
sive MRSA or other infections. isolates had MICs of 1  mg/L, with no were 1 to 2 dilutions higher (agree-
5. Vancomycin monitoring is re- signs of MIC creep over 20  years.76 ment with BMD was 36.7%). However,
commended for patients receiving While there does not seem to be a large when compared to BMD, Etest identi-
vancomycin for serious MRSA number of organisms with a vanco- fied a MIC of 2 mg/L 80% of the time.
infections to achieve sustained mycin MIC of ≥2 mg/L when reference When compared to BMD, Micro­
targeted AUC values (assuming a methods are used, there is considerable Scan WalkAway (prompt method)

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  841


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

overcalled MIC values of 1  mg/L by the lack of precision and variability vancomycin CI (n = 61) and II (n = 58)
74.1%, and BD Phoenix and Vitek 2 in MIC results depending on method in 119 patients. Most patients had
undercalled MIC values of 2 mg/L by used (B-II). pneumonia or bacteremia, mostly due
76% and 20%, respectively. to MRSA. Mean serum steady-state and
The high variability of MIC results trough concentrations attained were
among the 4 systems compared to Continuous Infusion vs 24  mg/L and 15  mg/L, respectively,
BMD clearly poses a challenge to the Intermittent Infusion for both the CI and II groups. AUC24
clinician making treatment decisions Administration of vancomycin by values were comparable between the
based on MIC and poses questions as continuous infusion (CI) has been CI and II groups, with significantly less
to the most relevant MIC method.79 evaluated as an alternative to intermit- variability in the CI group (P  =  0.026).
This variability between MIC values tent infusion (II) with potential advan- Clinical failure rates were similar in
and testing methods routinely per- tages of earlier target attainment, less the CI and II groups on day 10 (21% vs

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


formed at most institutions further variability in serum concentrations, 26%) and at end of treatment (21% vs
supports the use of AUC (assuming ease of drug level monitoring (less de- 19%), although the mean AUC24 was
a MICBMD of 1  mg/L) to guide vanco- pendence on sampling time or multiple shown to be lower in the CI group than
mycin empiric dosing. For nonserious concentrations to calculate AUC), and in the II group (596 [SD,  159] mg·h/L
infections, this variability may be in- lower the potential risk of AKI. vs 685 [SD,  260] mg·h/L, P  <  0.05).
consequential. In a critically ill patient Comparative studies.  Published Nephrotoxicity occurred in 18% of pa-
infected by MRSA, who may require studies that compared intermittent to tients overall, with similar rates in the
prompt achievement of the target continuous administration primarily CI and II groups (16% vs 19%). However,
AUC/MIC, it is imperative to verify focused on 2 distinct populations, adult dialysis was required more often in
the MIC by a standardized method critically ill patients in the ICU with sus- those who received CI vs II (6 of 10 pa-
(preferably BMD, as Etest may result pected or documented infections and tients vs 3 of 11 patients). Risk factors
in a higher MIC than BMD) as soon those receiving outpatient antimicro- for nephrotoxicity such as diabetes and
as possible to avoid a delay in effec- bial therapy (OPAT) for bone and joint concomitant diuretic, aminoglycoside,
tive therapy. An AUC/MICBMD of 400 infections.80-89 Most studies compared and iodine use were similar between
to 600 is approximately equivalent CI to II for the risk of AKI and attain- groups. It is notable that the study only
to an AUC/MICEtest of 200 to 400, re- ment of target serum concentrations; had 23% power to detect a difference in
flecting values that are 1 to 2 dilutions only 4 studies included other outcome clinical outcomes between groups.1
higher than those yielded by Etest. endpoints such as treatment failure and Another study compared mor-
Furthermore, there are no data to sup- mortality.80,84,87,89 Measures of vanco- tality among critically ill burn patients
port decreasing the dose to achieve mycin drug exposure reported in clin- receiving CI (n  =  90) or II (n  =  81).84
the targeted AUC/MIC of 400 to 600 if ical trials include trough and average Mortality rates in the hospital and on
the MIC is less than 1 mg/L. steady-state concentrations and AUC24. days 14 and 28 were numerically higher
One challenge when comparing clin- for those receiving CI, but the differ-
Summary and ical outcomes between CI and II is the ences did not reach statistical signifi-
recommendations: lack of consistent reporting of exposure cance (10% vs 6.2%, 18.9% vs 11%, and
parameters between groups treated 32% vs 21%, respectively). However,
6. Based on current national vancomycin using the 2 dosing strategies. For CI, when mortality was compared by treat-
susceptibility surveillance data, the most commonly reported drug ex- ment indications, those who received
under most circumstances of empiric posure parameter was the steady-state CI for non–gram-positive sepsis had
dosing, the vancomycin MIC should concentration, while for II it was the significantly higher mortality (70% vs
be assumed to be 1 mg/L. When the trough concentration. For future inves- 16.7%, P = 0.001); nearly half of this sub-
MICBMD is >1 mg/L, the probability of tigations it would be beneficial to report group had gram-negative bacteremia or
achieving an AUC/MIC target of   ≥400 AUC and/or average steady-state con- candidemia. It is possible that the dif-
is low with conventional dosing; centration for both CI and II groups to ference in outcome may be attributed to
higher doses may risk unnecessary enable direct comparison of drug expo- differences in the management of those
toxicity, and the decision to change sure between groups and correlate with infections and not directly related to van-
therapy should be based on clinical efficacy and safety endpoints. comycin administration. Nephrotoxicity
judgment. In addition, when the Critically ill patients.  A total of occurred numerically less frequently in
MICBMD is <1 mg/L, we do not recom- 7 studies compared CI vs II of vanco- the CI group than in the II group (per-
mend decreasing the dose to achieve mycin in critically ill patients.81-87 Only centage of patients with increase in Scr
the AUC/MIC target. It is important to one study, by Wysocki et  al,80 evalu- of 0.5  mg/dL at end of therapy, 6.7% vs
note the limitations in automated sus- ated both efficacy and safety in a pro- 14.8%). While higher mean vancomycin
ceptibility testing methods, including spective randomized trial comparing concentrations were noted in the CI

842    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

group relative to the II group (20 [SD, 3.8] 1-mg/L increase in serum concentra- of 80 patients, a trend towards less fre-
mg/L vs 14.8 [SD, 4.4] mg/L, P < 0.001), tion was associated with an 11% increase quent occurrence of nephrotoxicity was
which would be expected when com- in the risk of nephrotoxicity, with lower observed in the CI group vs the II group
paring steady-state and trough concen- odds in those receiving II. However, lo- (10% vs 25%, P = 0.139), and when neph-
trations, AUC24 was not reported, thereby gistic regression analysis indicated the rotoxicity did occur it had a later onset
precluding comparison of drug exposure contrary in that II was associated with in the CI group (P  =  0.036).88 Patients
between the CI and II groups. an 8-fold higher odds of nephrotoxicity were matched by age, comorbid condi-
Five other studies compared serum (95% confidence interval, 2.87-23.41). tions, gender, baseline Scr, and receipt
drug concentrations achieved and the The lack of information provided on of concurrent nephrotoxins; those who
risk of nephrotoxicity between CI and confounding variables such as receipt of had an Scr of ≥1.5  mg/dL at baseline,
II in critically ill patients.81-83,85,86 As ex- concomitant nephrotoxins and relative developed nephrotoxicity as inpatients
pected, the ranges of measured vanco- AUCs between treatment groups pre- prior to OPAT, or experienced hypo-

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


mycin concentrations from the studies clude drawing a definitive conclusion tension resulting in renal dysfunction
were significantly higher in the CI groups regarding the safety of CI, especially in were excluded. In another retrospective
than in the II groups (steady-state con- light of the disparate results of bivariate study,90 the same investigators identified
centrations of 20-25  mg/L vs troughs and logistic regression analyses. a steady-state average concentration of
of 10-15  mg/L, respectively). Another Patients receiving OPAT. To date 28 mg/L as the threshold breakpoint for
study showed that a higher percentage there have been 2 studies comparing the development of nephrotoxicity using
of patients attained a vancomycin con- the efficacy of vancomycin adminis- CART (classification and regression
centration of >20  mg/L at least once tration by CI vs II in patients whose tree) analysis: Nephrotoxicity occurred
during the treatment course with CI therapy was initiated in the hospital and in 71.4% (5 of 7) and 11.6% (11 of 95) pa-
vs II administration (63.2% vs 44.9%, continued as OPAT. Duration of therapy tients with steady-state concentrations
P  =  0.065).82 One study reported lower ranged from 30  days to 14 weeks.87,89 of ≥28 mg/L and <28 mg/L, respectively.
mean AUC24 with CI vs II (529 [SD, 98] Most patients were treated for bone and In one prospective study of an elderly
mg·h/L vs 612 [SD, 213] mg·h/L, P value joint and skin structure–related infec- cohort (mean age, 70  years) receiving
not stated), and increased steady-state tions. In a small prospective study, rates high-dose vancomycin therapy by CI,
concentration compared with trough of osteomyelitis cure, defined as re- with targeting of a steady-state concen-
(25 ± 4 vs 17 ± 4.7 mg/L, respectively, maining asymptomatic 12 months after tration of 30 to 40  mg/L for a median
P = 0.42) with CI vs. II.83 The discordance completion of therapy, did not differ duration of 6 weeks, nephrotoxicity oc-
observed in the relationship of trough significantly between groups (94% vs curred in 32% of patients. Additionally,
concentration and AUC24 underscores 78%, P = 0.3), but only 27 patients were 4 patients in that study developed
the importance of measuring AUC24 to evaluable.87 Another study retrospec- leukopenia.91
compare relative drug exposure with CI tively evaluated the efficacy of vanco- Dosing and other consider-
vs II in future studies. mycin in patients with MRSA infections; ations for use of CI. Most published
In general, the rate of nephrotoxicity most had bone and joint and skin struc- studies of critically ill patients receiving
was reported to be similar or numeri- ture–related infections, while 10% had vancomycin CI employed a loading
cally lower with CI vs II administration bloodstream infections or endocar- dose of 15 to 20 mg/kg followed by daily
(range, 4%-16% vs 11%-19%); the same ditis.89 Rates of clinical failure were sim- maintenance infusions at doses of 30 to
trend but higher rates were reported in ilar in the CI and II groups (19% [25 of 40  mg/kg (up to 60  mg/kg) to achieve
studies that applied the AKIN criteria for 133 patients] vs 25% [9 of 36], P = 0.41) a target steady-state concentration of
nephrotoxicity (26%-28% vs 35%-37%).81- after excluding 29% of study patients 20 to 25  mg/L. By simply multiplying
83,85,86
In addition, Saugel et  al85 noted who had subtherapeutic serum van- the steady-state concentration by 24, a
significantly less frequent need for renal comycin concentrations for more than target steady-state concentration of 20
replacement therapy (RRT) during van- 1 week. However, it is not clear how to 25 mg/L would equate to an AUC24/
comycin treatment for patients in the CI frequent serum concentrations were MIC of 480 to 600 (assuming a MIC of
group than for those in the II group (7% monitored, if in-hospital treatment du- 1 mg/L). Of note, the PK/PD target for
[7 of 94 patients] vs 23% [12 of 52 pa- ration before OPAT differed between CI has not been validated. All of the PK/
tients] required RRT; P  =  0.007). Of in- groups, and whether treatment success PD data supporting an AUC24/MIC ratio
terest, in the largest retrospective study rates differed by type of infection. of >400 as the best correlate for clinical
comparing CI and II, conducted in 1,430 In studies that evaluated the safety of outcomes were derived from patients
ICU patients, Hanrahan et al86 reported a CI vancomycin as OPAT, treatment dura- who received II vancomycin dosing.
higher rate of nephrotoxicity in those re- tion ranged from 4 to 14 weeks, with a re- Rapid attainment of target serum
ceiving CI vs II (25% [161 of 653 patients] ported average mean steady-state serum concentrations has been cited as a po-
vs 20% [77 of 390 patients]; P  =  0.001); concentration of 13 to 30  mg/L.87,88 In tential advantage of CI over II when
bivariate analysis indicated that every a retrospective matched cohort study treating acute infections, particularly

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  843


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

in ICU patients early during the course 25 mg/L throughout the entire dosing events. An infusion rate of 10  mg/min
of infection. In 2 comparative studies, interval) by a factor of 24. Attaining or less is associated with fewer infusion-
target steady-state concentrations of 20 the desired drug exposure may be related events. Loading doses of 25 to
to 25 mg/L were achieved more rapidly more readily accomplished given the 35  mg/kg will require infusion times of
with use of CI vs II: in a mean time of ease of sampling time and dosage at least 2 to 3 hours.99 After administra-
36 (SD, 31)  hours vs 51 (SD, 39)  hours adjustment by changing the rate of tion of the loading dose, the initiation of
(P  =  0.03) in one study and 16 (SD, infusion, which is a highly desirable the maintenance dose should occur at
8) hours vs 50 (SD, 21) hours (P < 0.001) feature in critically ill patients (B-II). the next dosing interval (eg, an interval
in the other.81,83 Importantly, less var- 8. The risk of developing nephrotoxicity of every 6 hours indicates initiating the
iability in the steady-state concentra- with CI appears to be similar or lower maintenance dose 6 hours after the start
tion and fewer blood samples (a single than that with intermittent dosing of the loading dose).
steady-state concentration vs both when targeting a steady-state concen- In most studies that have employed

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


peak and trough concentrations) are tration of 15 to 25 mg/L and a trough loading doses, vancomycin dosing was
required to calculate AUC24 among pa- concentration of 10 to 20 mg/L (B-II). based on actual body weight. While
tients receiving CI vs II. Timing of blood Definitive studies are needed to com- this practice is commonplace, dosing
sampling for trough determinations pare drug exposure based on meas- by actual body weight assumes there
is critical during II, whereas steady- ured AUC24 and factors that predispose is a linear relationship between key
state concentration can be measured to development of nephrotoxicity, such population PK parameters (ie, Vd and
any time after steady state has been as receipt of concomitant nephro- clearance) and the body size descriptor
reached during CI. In addition, vanco- toxins, diuretics, and/or vasopressor employed. While a wide variety of ac-
mycin administration by CI in patients therapy in patients receiving CI vs II of tual weight–based estimates of Vd (for
receiving OPAT has the theoretical ad- vancomycin. example, 0.4 to 1  L/kg) have been re-
vantage of a need for less frequent ac- 9. Incompatibility of vancomycin ported in the literature,6 mounting
cess to the i.v. catheter and thus less with other drugs commonly data suggest that it is not entirely ac-
complications resulting from thrombus coadministered in the ICU requires curate to describe vancomycin Vd as
formation or infections. On the other the use of independent lines or mul- being proportional to body weight,
hand, incompatibility of vancomycin tiple catheters when vancomycin is particularly among obese patients
with certain drugs (particularly at high being considered for CI (A-III). (refer to Dosing in Obesity section).
concentrations), that are commonly As noted in several recent articles dis-
administered in the critical care setting cussing vancomycin PK in obesity, as
is a notable challenge of vancomycin Loading Doses weight increases the coefficient used
CI.92,93 The use of proper concentration, Loading doses of vancomycin have to calculate Vd decreases.48,110,111 At this
alternative agents, independent lines, been evaluated in several studies during point, dosing should be based on ac-
or multiple catheters may be warranted the past decade.94-109 Providing loading tual body weight, with doses capped
if vancomycin is to be administered by doses of 20 to 35  mg/kg based on ac- at 3,000 mg (refer to Dosing in Obesity
CI. tual body weight rapidly achieves tar- section.112 More intensive therapeutic
geted ranges of serum vancomycin monitoring should also be performed
Summary and concentrations and decreases the risk of in obese patients.
recommendations: subtherapeutic concentrations during
the first days of therapy. Loading doses are Summary and
7. The pharmacokinetics of CI suggest recommended in patients who are criti- recommendations:
that such regimens may be a reason- cally ill or in the ICU,95-102 require dialysis
able alternative to conventional II or renal replacement therapy,102-106 or are 10. In order to achieve rapid attain-
dosing when the AUC target cannot receiving vancomycin CI therapy.94-98,105,108 ment of targeted concentrations in
be achieved (B-II). Based on cur- While this approach is not currently sup- critically ill patients with suspected
rently available data, a loading dose ported by evidence from large random- or documented serious MRSA in-
of 15 to 20 mg/kg, followed by daily ized clinical trials, vancomycin loading fections, a loading dose of 20 to
maintenance CI of 30 to 40 mg/kg doses can be considered in the treatment 35 mg/kg can be considered for
(up to 60 mg/kg) to achieve a target of serious MRSA infections. Vancomycin intermittent-infusion administration
steady-state concentration of 20 to should be administered in a dilute solu- of vancomycin (B-II).1
25 mg/L may be considered for crit- tion (eg, concentrations of no more than 11. Loading doses should be based on
ically ill patients (B-II). AUC24 can 5  mg/mL) and infused over a period of actual body weight and not exceed
be simply calculated by multiplying not less than 60 minutes or at a rate of 3,000 mg (refer to Dosing in Obesity
the steady-state concentration (ie, 10 to 15 mg/min (≥1 hour per 1,000 mg) section). More intensive and early
the desired therapeutic range of 20 to to minimize infusion-related adverse therapeutic monitoring should

844    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

also be performed in obese patients tendency estimates approaching 75  L approaches to estimating vancomycin
(B-II). are observed in obese adults.112,120,121 CL, such as that defined by Rodvold
The nonlinear relationship between and colleagues,125 can be used to esti-
vancomycin Vd and body weight can be mate the total daily maintenance dose.
Dosing in Obesity resolved with piecewise functions of al- The population model–estimated van-
The original vancomycin dosing ternate weight descriptors, allometric comycin CL multiplied by the target
strategies predate our current defin- scaling, use of lower mg/kg doses with AUC estimates the initial daily main-
itions of obesity and understanding increasing body size, or capping the tenance dose.112,120,122 For example,
of drug pharmacokinetics in obesity. dose at a threshold.118,122 The underlying studies report an average vancomycin
Obesity is defined as a body mass index rationale for a loading dose is rapid at- CL of approximately 6  L/h in obese
(BMI) of ≥30  kg/m2 and is currently tainment of therapeutic concentra- patients that equates to achieving an
divided into 3 tiers: class  I  obesity tions. Therefore, using actual body AUC of approximately 500 mg·h/L with

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


(30.0-34.9 kg/m2), class II obesity (35.0- weight–based loading doses of 20 to a daily dose of 3,000  mg. Empiric van-
39.9  kg/m2), and class  III, or morbid, 25 mg/kg (doses lower than previously comycin maintenance dosages above
obesity (≥40  kg/m2).113 The prevalence recommended), with consideration of 4,500 mg/day are not expected in obese
of obesity increased from approxi- capping doses at 3,000 mg, is the most adults, because vancomycin CL rarely
mately 10% in the 1950s to 39.8% in practical strategy in obese patients exceeds 9 L/h.112,120,121
2015-2016, and the average US adult with serious infections.112 For example, Population PK models of vanco-
weighs approximately 83 kg, compared this strategy would result in calculated mycin cannot account for more than
to the historical standard of 70 kg.114,115 loading doses of 1,500 to 2,500  mg in 50% of the interindividual variabili­
This shift in the distribution of body patients weighing 80 to 99  kg, 2,000 ty, which supports therapeutic drug
­
size is relevant to the calculation of to 3,000  mg in those weighing 100 to monitoring (TDM) in this popula-
vancomycin doses based on patient 119  kg, and 2,500 to 3,000  mg in pa- tion.117,118,120,122 A  reliable estimate of
body weight. Obesity may be associated tients with a weight of ≥120  kg (doses vancomycin Vd is necessary for AUC
with an increased risk of vancomycin- rounded to the nearest 250  mg). The estimation when AUC is based solely
induced nephrotoxicity, in part due to decision of whether or not to employ a on a trough concentration measure-
supratherapeutic exposure resulting loading dose, as well as the magnitude ment.24,121,126,127 This bias is addressed
from maintenance doses calculated of this dose, should be driven by the and precision is improved by meas-
using actual body weight.45,116 severity of infection and the urgency urement of both a peak (collected
The selection of vancomycin to achieve a therapeutic concentration at least 1 hour after the end of infu-
loading dose is dependent on the es- rather than by body size alone. sion) and a trough concentration to
timated Vd. Pharmacokinetic studies Empiric maintenance dosing of estimate AUC accurately in obese
have repeatedly demonstrated that the vancomycin is reliant on estimated CL. patients.126 Once a reliable PK es-
vancomycin Vd increases with actual Vancomycin CL is predicted by kidney timate of vancomycin elimination
body weight; however, this PK param- function, which is most commonly is determined by using these 2 con-
eter does not increase with actual body estimated as CLcr with the Cockcroft- centration measurements, subsequent
weight in a proportionate manner and Gault equation using patient age, sex, vancomycin AUC estimation is achiev-
is not reliably predictable in obese indi- Scr, and body size.123 There is consider- able with trough-only measurements
viduals.111,117-121 Blouin and colleagues111 able controversy regarding the optimal by Bayesian methods in physiologically
demonstrated a statistically significant body size metric for this calculation in stable patients.57 For critically ill obese
difference in weight-indexed Vd be- obese patients.124 The Cockcroft-Gault patients with unstable physiology, ad-
tween obese and nonobese patients. equation predates the global standard- ditional work to design adaptive feed-
Similarly, using data from 704 patients, ization of Scr measurement traceable back models to tailor doses is needed.
Ducharme and colleagues118 found that to isotopic-dilution mass spectrometry
mean weight-indexed vancomycin Vd (IDMS) standards advocated to reduce Summary and
decreased with increasing body size. intralaboratory and interlaboratory recommendations:
The average weight-indexed Vd in a measurement variability.124 A  recent
study by Bauer and colleagues119 was population PK study by Crass and col- 12. A vancomycin loading dose of 20
much lower in 24 morbidly obese pa- leagues112 of obese patients (n  =  346) to 25 mg/kg using actual body
tients (0.32 L/kg) than in 24 patients of with BMI values of 30.1 to 85.7  kg/m2 weight, with a maximum dose of
normal weight (0.68  L/kg, P  <  0.001). and body weights of 70 to 294  kg pro- 3,000 mg, may be considered in
Recent studies in obese adults corrob- vided an equation to estimate vanco- obese adult patients with serious
orate these findings and suggest that mycin CL based on age, sex, Scr (IDMS infections (B-II). Initial mainte-
lower Vd estimates of approximately traceable), and allometrically scaled nance doses of vancomycin can be
0.5 L/kg or weight-independent central body weight. This model or similar computed using a population PK

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  845


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

estimate of vancomycin clearance these patients. These considerations and published a wide variety of vanco-
and the target AUC in obese patients. include the amount of time between mycin dosing protocols in an attempt to
Empiric maintenance doses for most vancomycin dose administration and compensate for the increase in vanco-
obese patients usually do not exceed the scheduled time of the next dialysis mycin dialytic CL caused by increases
4,500 mg/day, depending on their session,104 whether the dose is given in dialyzer permeability.
renal function (B-II). Early and fre- during dialysis or after hemodialysis An added complication of appro-
quent monitoring of AUC exposure is has ended, and the dialyzer’s per- priate vancomycin dosing in patients
recommended for dose adjustment, meability if the dose is administered receiving hemodialysis is the prevailing
especially when empiric doses exceed intradialytically.131 Dialysis frequency practice of administering the drug during
4,000 mg/day (A-II). Measurement also plays a role in dosing decisions. the final hours of the hemodialysis pro-
of peak and trough concentrations is For non–critically ill patients receiving cess, thus resulting in some of the infused
recommended to improve the accu- hemodialysis, 2 or 3  days is the most drug being removed immediately by the

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


racy of vancomycin AUC estimation common interdialytic period. Some hemodialyzer. This practice started back
and maintenance dose optimi- critically ill patients with severe catab- when low-permeability dialyzers were
zation in obese patients, aligning olism and AKI may require more than used and little vancomycin was elimin-
with recommendations 2 and 5 for thrice-weekly hemodialysis for optimal ated by hemodialysis. The practice has
nonobese adults. metabolic control, and their main- persisted at most dialysis units because
tenance vancomycin doses should most dialysis units treat 3 shifts of patients
be based on serum concentration per day, and holding a dialysis chair for
Renal Disease and Renal monitoring.132 60 to 90 additional minutes while vanco-
Replacement Therapies Vancomycin dosing in patients mycin infuses into a patient is not cost-ef-
Intermittent  hemodialysis. with acute or chronic kidney failure fective. Indeed, it is more cost-effective
Despite the common use of vanco- has transformed over time due to to infuse “extra” vancomycin during the
mycin in patients receiving hemodial- the changes in dialysis technology hemodialysis session to compensate for
ysis, there are few published outcome and techniques.133 Older (pre-1990s) intradialytic loss than it is to keep a dial-
studies that provide guidance on the hemodialyzers were not very perme- ysis unit open later to allow vancomycin
optimal PK/PD targets in this popula- able to large molecules. Vancomycin infusions. Intradialytically infused vanco-
tion. Previously published drug dosing (with a molecular weight of 1,450  Da) mycin results in reduced delivery of drug
recommendations generally targeted a was not considered “dialyzable” be- to the patient, similar to a first-pass phe-
predialysis serum concentration, even cause it poorly crossed the hemodi- nomenon. The extent of intradialytic drug
though other PD targets may be more alysis membranes of the era. Indeed, removal is variable and depends on pa-
appropriate. Predialysis vancomycin even today’s vancomycin package in- tient and dialysis system factors, the most
concentration to MRSA MIC ratios of sert, based on PK studies conducted important of which is dialyzer membrane
>18.6 have been associated with im- in the 1980s, states that “vancomycin permeability.135,137-139 Approximately 20%
proved bacteremic patient outcomes, is poorly removed by dialysis.”  134
As to 40% of an intradialytically adminis-
suggesting that serum concentration hemodialysis membrane technology tered vancomycin dose is removed by
monitoring is essential throughout the has improved, dialyzers have become the simultaneous hemodialysis, with the
course of therapy.128 Dosing to achieve far more permeable. Vancomycin is highly permeable dialyzers tending to the
predialysis vancomycin concentrations cleared substantially by contemporary higher end of this range.137,140,141
of 10 to 20 mg/L, as has been done clin- high-permeability hemodialyzers135,136; Maintenance dosing strategies that
ically,129 results in mean AUC24 values consequently, vancomycin dosing do not provide a dose with every he-
ranging from 250 to 450 mg·h/L, with strategies have changed substantially modialysis session (eg, a maintenance
some values below the AUC/MIC goals as well. For example, in spite of the dose is given with every second or
recommended in other populations.130 package insert statement “In anuria, third hemodialysis session) have been
Outcome studies validating the AUC24h a dose of 1000  mg every 7 to 10  days studied,102,142,143 but none have been
goal of 400 to 600 mg·h/L used in other has been recommended” and the found to meet vancomycin exposure
patient populations have not been con- statement that “vancomycin is poorly goals in the last day of the dosing in-
ducted in the hemodialysis population. removed by dialysis,” 134 far more fre- terval without giving massive doses
Nonetheless, the maintenance doses quent doses are needed to maintain that result in very high peak concen-
recommended in this section aim to therapeutic serum concentrations in trations. Consequently, maintenance
reach this AUC24 target (ie, 400-600 patients receiving hemodialysis. The vancomycin doses are recommended
mg·h/L), as recommended throughout extent of vancomycin removal by dial- to be administered with each hemo-
this document. ysis is dependent on the permeability dialysis session to ensure therapeutic
Many dialysis-related factors affect of the hemodialyzer used131; conse- serum concentrations throughout
the degree of vancomycin exposure in quently, investigators have developed the dosing interval. In the typical

846    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

thrice-weekly hemodialysis schedule, other patient populations have not been


Timing and Dialyzer Vancomycin
25% larger doses are needed for the conducted in the hemodialysis popula-
Permeability Dose, mg/kga
3-day interdialytic period (eg, Friday to tion. While determination of AUC/MIC
Monday) to maintain sufficient vanco- attainment is recommended, limited Intradialytic
mycin exposure on the third day.130,144 serum concentration monitoring is pos-
Low permeability Loading: 30
Dosing that is weight based ap- sible in patients receiving hemodialysis Maint.: 7.5-10b
pears to be superior to standard dosing in the outpatient setting for 2 reasons. The
High permeability Loading: 35
schemes that do not account for patient first reason is that frequent phlebotomy Maint.: 10-15b
size. Further, doses should be based on must be avoided in order to preserve
actual body weight rather than a calcu- future hemodialysis vascular access
a
From references 104, 129, 130, 137, 138, 140,
and 147.
lated body weight (see Dosing in Obesity needs; the second is that it is imprac- b
Thrice-weekly dose administration.
section for considerations on how to tical to obtain blood samples aside from

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


dose morbidly obese patients). Because the predialysis sample that is obtained 14. Since efficacy data are unavailable
vancomycin is water soluble, vanco- from the blood catheter inserted for use for AUC values of <400 mg·h/L,
mycin dosing in fluid overloaded patients in the dialysis process. Patients leave the monitoring based on predialysis
should also be based on actual body dialysis unit after hemodialysis and do serum concentrations and extrapo-
weight at the time of dosing rather than not return until the next dialysis session lating these values to estimate AUC
on some calculated adjusted weight.102-105 days later. Consequently, since data are is most practical. Maintaining
Serum concentration monitoring unavailable for an optimal AUC target in predialysis concentrations be-
is a valuable tool to guide vancomycin these patients, and no data are available tween 15 and 20 mg/L is likely
dosing in patients receiving dialysis, to demonstrate efficacy below an AUC to achieve the AUC of 400 to 600
provided that serum concentrations are threshold value of 400, the goal should mg·h/L in the previous 24 hours
obtained and interpreted correctly. For be to attain the AUC target of 400 to 600 (C-III). Predialysis serum con-
example, blood sampling for assess- mg·h/L used in other patient popula- centration monitoring should be
ment of vancomycin concentrations tions. It is most practical to continue performed not less than weekly and
should not occur during or for at least monitoring based on predialysis con- should drive subsequent dosing, as
2 hours after a hemodialysis treatment. centrations and extrapolate these values opposed to a strict weight-based
These samples will not be reflective of to estimate AUC. Maintaining predialysis recommendation, although these
the true vancomycin body load because concentrations between 15 and 20 mg/L recommended doses provide a
of the dialytic removal of vancomycin. is likely to attain the AUC target of 400 useful starting point until serum
Vancomycin serum concentrations will to 600 mg·h/L in the previous 24 hours, concentrations have been deter-
be low immediately following a dialysis with higher AUC/MIC values occurring mined (B-II).
treatment but will rebound substan- on days prior.
tially as drug redistributes from the tis- Hybrid hemodialysis ther-
sues back to the blood over the next few apies.  Contemporary renal replace-
Summary and
hours.131,142,145 Dosing decisions based ment therapies used to treat kidney
recommendations:
on serum concentrations obtained disease have expanded well beyond
during or soon after hemodialysis will thrice-weekly, 3- to 4-hour hemodial-
13. The following tabulation outlines ysis sessions. In the outpatient setting,
be inherently incorrect and could re-
recommended vancomycin loading shorter, more frequent home hemodi-
sult in administration of doses higher
and maintenance doses for pa- alysis treatments are used in a growing
than necessary.145 Serum concentration
tients receiving hemodialysis, with number of patients. In the inpatient set-
monitoring performed with blood sam-
accounting for permeability of the ting, various types of “hybrid” hemodi-
ples obtained prior to the hemodialysis
dialyzer and whether the dose is ad- alysis therapies are employed. These
treatment is recommended to guide
ministered intradialytically or after hybrid treatments go by many names,
dosing, although other serum concen-
dialysis ends (B-II). including prolonged intermittent renal
tration monitoring techniques have
been suggested.146 replacement therapy (PIRRT) and
Dosing to achieve predialysis vanco- Timing and Dialyzer Vancomycin slow-low efficiency dialysis (SLED).
mycin concentrations of 10 to 20 mg/L, Permeability Dose, mg/kga Essentially these hybrid therapies use
as has been conducted clinically,129 re- standard hemodialysis machines that
After dialysis ends
sults in mean AUC24 values ranging from run at slower blood and dialysate flow
250 to 450 mg·h/L, often below the AUC/ Low permeability Loading: 25 rates and for longer durations (usually
Maint.: 7.5b 6 to 12 hours per day). Even hemodi-
MIC goals recommended in other popu-
lations.130 Outcome studies validating the High permeability Loading: 25 alysis itself differs in the inpatient and
AUC target of 400 to 600 mg·h/L used in Maint.: 10b outpatient settings, as patients with AKI

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  847


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

are often hemodynamically unstable recognizing that these hybrid dialysis to 10 mg/kg every 12 hours (B-II).
and lack sufficient vascular access for therapies efficiently remove vanco- Maintenance dose and dosing in-
robust blood flow through the dialysis mycin (B-III). Initial doses should not terval should be based on serum con-
vascular access. All these hybrid dial- be delayed to wait for a dialysis treat- centration monitoring, which should
ysis therapies clear vancomycin to a ment to end. Maintenance doses of be conducted within the first 24
different extent than standard intermit- 15 mg/kg should be given after hybrid hours to ensure AUC/MIC targets are
tent hemodialysis.148,149 The timing of hemodialysis ends or during the final met.154 In fluid overloaded patients,
the vancomycin dose in relation to the 60 to 90 minutes of dialysis, as is done doses may be reduced as patients
hybrid hemodialysis session is essen- with standard hemodialysis (B-III).130 become euvolemic and drug Vd de-
tial in determining a dosing regimen. Concentration monitoring should creases. The use of CI of vancomycin
If hybrid hemodialysis is started soon guide further maintenance doses. in patients receiving CRRT appears
after the dose is administered, much of to be growing,84,105 and this method

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


the dose will be removed, whereas the Continuous renal replacement could be used in place of intermittent
same vancomycin dose given after the therapies.  The use of continuous vancomycin dosing, especially when
dialysis session ends will yield a much renal replacement therapy (CRRT) mo- high CRRT ultrafiltrate/dialysate
larger AUC24 and much higher average dalities like continuous venovenous flow rates are employed (B-II).
serum concentrations. As is the case hemofiltration (CVVH), continuous
with any hemodialysis therapy, serum venovenous hemodialysis (CVVHD), and
concentrations obtained during or continuous venovenous hemodiafiltration Pediatric Patients
within 2 hours from the end of hemo- (CVVHDF) has grown in popularity in In 2011, prior to the availability of
dialysis will be artificially low because critically ill patients with AKI because of alternative agents for MRSA in pediat-
dialysis will have efficiently removed their superior ability to provide fluid and rics, vancomycin was recommended
vancomycin from the blood, and van- solute balance. Provided these therapies as the drug of choice for invasive MRSA
comycin located in the tissues will not operate in an uninterrupted fashion, infections in children.5 Although there
have had time to redistribute back into vancomycin CL is relatively constant are limited prospective, comparative
the bloodstream. Calculation of main- over the dosing interval, although CL data on the value of vancomycin thera-
tenance doses based on an intra- or may decline as the hemodiafilter clogs peutic monitoring in adults with respect
postdialytic vancomycin serum con- over time.151 Vancomycin is removed to improving outcomes and decreasing
centration may result in doses that are by CRRT and its CL is related closely to toxicity, virtually no prospectively col-
too high. Caution is recommended in the rate of ultrafiltrate/dialysate flow,105 lected data on outcomes of MRSA infec-
basing any maintenance dosing on with hemodiafilter type being of lesser tion in newborns, infants and children
these serum concentration values. importance, because contemporary exist. Further, for newborns (particu-
Little has been published on the hemodiafilters are all very permeable to larly premature infants) compared with
patient outcomes achieved when van- the drug. older infants, immature renal elimina-
comycin is used in patients receiving In patients on CRRT, serum concen- tion mechanisms and a relative increase
hybrid dialysis. Authors of one small tration attainment goals often are not met in Vd by body weight further complicate
case series of 27 courses of vancomycin with conventional dosing.84,152 Although dosing guidelines during the first sev-
given to patients receiving a hybrid he- outcomes studies specific to patients re- eral weeks of life. Additional complexity
modialysis therapy reported that pre- ceiving CRRT have not been conducted, for dosing strategies during early child-
scribers have tried a wide variety of it seems prudent to apply the same van- hood is based on a continual maturation
dosing schemes.150 By these authors’ comycin AUC/MIC target (ie, 400-600) of glomerular filtration, which is directly
criteria, 89% of the prescribed vanco- in these critically ill patients as is recom- related to vancomycin CL. The glomer-
mycin doses in their institution were mended throughout this document. ular filtration rate increases through the
too low. Given the absence of outcome first years of life to rates in school-aged
data in patients receiving these ther- Summary and children that are greater than those in
apies, it seems prudent to use the same recommendations: adults, with subsequent decline during
vancomycin AUC goal recommended the teen years to adult normal rates.
throughout this document (400 to 600 16. Loading doses of 20 to 25 mg/kg by Such a diversity of PK parameter values
mg·h/L assuming a MIC of 1 mg/L). actual body weight should be used based on developmental pharmacology
in patients receiving CRRT at con- from neonates to adolescents pro-
Summary and ventional, KDIGO-recommended vides a challenge to develop general-
recommendations: effluent rates of 20 to 25 mL/kg/h ized vancomycin dosing. However, this
(B-II).153 Initial maintenance has improved with the application of
15. Loading doses of 20 to 25 mg/kg dosing for CRRT with effluent rates population-based PK models using al-
actual body weight should be used, of 20 to 25 mL/kg/h should be 7.5 lometric scaling and renal maturation

848    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

covariates. In a population-based PK and outcomes data to support the of 1  mg/L or less, p ­ resumably as a re-
study by Colin and colleagues155 that higher end of the AUC target range in sult of greater vancomycin CL than is
evaluated vancomycin PK throughout pediatrics, it is prudent to aim for an seen in adults.1,161-164 For children in-
the entire age continuum from infancy AUC/MIC of 400 in pediatrics to limit fected by MRSA pathogens with a MIC
to geriatric years using pooled data from the development of exposure-related of >1 mg/L, it is unlikely that the target
14 studies, age, weight, and kidney func- AKI. Furthermore, in pediatrics, an exposure can be reliably achieved with
tion were important factors in estimating AUC/MIC target of 400 is more readily previously investigated dosages of van-
clearance. Careful monitoring in the achievable than it is in adults and cor- comycin in children.
pediatric population is prudent, espe- relates to trough concentrations of 7 Le and colleagues164 utilized
cially with the evident dynamic changes to 10 mg/L rather than concentrations population-based PK modeling to an-
in renal function in this population. As of 15 to 20  mg/L as are reported in alyze 1,660 vancomycin serum con-
with adults, comorbidities and concur- adults. This wide variability in trough centrations obtained at 2 institutions

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


rent medications can influence vanco- concentrations between these popula- from 2003 to 2011 among 702 children
mycin tissue distribution, elimination, tions with regard to achieving an AUC/ older than 3 months of age with varying
and toxicity. MIC of 400 corroborates the need for comorbidities. They demonstrated that
Limitation of outcomes data. an AUC-guided approach to dosing 4 important factors (age, weight, renal
Recent retrospective studies on bac- and monitoring. It is possible that in function as assessed by SCr, and MIC)
teremic S.  aureus infections (both otherwise healthy children with fewer contributed to vancomycin exposure.
MRSA and MSSA strains) in children comorbidities than are typically seen Monte Carlo simulations were created
treated with vancomycin suggest that in adults, a lower target may yield out- using population-based PK modeling
trough concentrations of >15  mg/L comes equivalent to an AUC of 400 to with Bayesian estimation and MICs of
were not associated with improved 600 mg·hr/L. The decision to retain or clinical isolates as determined by Etest,
outcomes, yet an increase in AKI increase AUC target exposure should with 85% of clinical isolates demon-
was observed.156-158 Furthermore, an- be based on clinical judgment in the strated to have a MICEtest of 1  mg/L or
other retrospective pediatric study management of these patients. less. To achieve an AUC/MICEtest of ≥400
evaluating outcomes of MRSA bac- With use of currently recommended in 90% of subjects, a dosage of 80 mg/kg/
teremia as a function of an AUC/ vancomycin dosages of 45 to 60  mg/ day was necessary, particularly in those
MICBMD of ≥400 did not show im- kg/day, widespread treatment failures less than 12  years of age with normal
proved outcomes.159 Similarly, van- in children have not been reported in renal function. At a dosage of 80  mg/
comycin trough concentrations of the literature, which may be reflective kg/day, the median AUC and median
<10 mg/L, as compared with concen- of a younger host with a more robust trough concentration were 675 mg·h/L
trations of >10 mg/L, were not associ- systemic and immunologic response and 16 mg/L, respectively. As expected,
ated with increased 30-day mortality to infection, a different management subjects 12 years of age or older achieved
and recurrent bacteremia in chil- approach (surgical and antibiotic) to similar exposure at lower dosages of 60
dren, although the lower concentra- invasive MRSA infection, lack of associ- to 70  mg/kg/day. At a dosage of 60 to
tions were associated with prolonged ated comorbidities, or publication bias. 70  mg/kg/day (divided doses admin-
bacteremia.160 Prospective comparative clinical trials istered every 6 hours), an AUC of 400
In the absence of prospective, com- involving children with documented mg·h/L correlated to a mean trough of 8
parative outcomes data in children re- infections treated with different vanco- to 9 mg/L.164 The clinical applicability of
garding unique AUC/MIC exposures mycin dosages or exposures have not this PK model for vancomyin CL estima-
necessary for clinical and microbio- been published. tion to determine AUC exposure was val-
logic success in treating serious MRSA Empiric maintenance regimen. idated in a small study by Ploessl et al.165
infections in different neonatal and Published retrospective PK/PD data Other studies corroborated Le and
pediatric populations to validate the in children suggest that current van- colleagues’ findings regarding the need
observations reported in adults (see comycin dosing of 45 to 60 mg/kg/day to use higher dosages, ranging from 60
Clinical PK/PD Data: Adults section), (in divided doses administered every to 80 mg/kg/day, depending on age and
dosing in children should be designed 6 to 8 hours) may be insufficient to renal function.162,164,166,167 Using the liter-
to achieve an AUC of 400 mg·h/L and achieve currently recommended tar- ature for vancomyin CL published in or
potentially up to 600 mg·h/L (assuming gets for adults of an AUC of 400 to 600 before 2000 and Bayesian estimation
a MIC of 1 mg/L). This PD target range, mg·h/L (assuming a MIC of 1  mg/L).1 for one 25-kg base subject, Frymoyer
specifically a range closer to an AUC/ In fact, higher dosages, ranging from et  al163 evaluated the relationship be-
MIC of 400 rather than 600, has been 60 to 80  mg/kg/day and given in di- tween AUC and trough concentrations,
widely used by investigators to model vided doses every 6 hours, may be showing that a dosage of 60 mg/kg/day
pediatric dosing and therapeutic moni- needed to achieve these targets for achieved trough concentrations of 7 to
toring. With inadequate PK studies MRSA strains with a vancomycin MIC 10  mg/L and an AUC/MIC of ≥400 in

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  849


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

90% of children for MRSA pathogens every 8 hours) in renally impaired chil- greater than 15 to 20 mg/L and AKI in
with a MIC of 1  mg/L. However, their dren achieved AUC exposure similar to pediatric patients. In addition, they
finding may not be extrapolatable to the that achieved with a dosage of 60 mg/kg/ showed that children who received con-
entire pediatric population given the day in children with normal renal func- current nephrotoxic drugs (particularly
variable ages and renal function. In a tion. Notably, they showed that in 87% furosemide) and stayed in the pediatric
second study, these investigators dem- of children with initial renal impairment, ICU were also more likely to experi-
onstrated that a dosage of 60 mg/kg/day vancomycin CL improved (with a lag in ence AKI. Four studies published later
achieved AUC/MICBMD values between the recovery of renal function as assessed corroborated these findings, indicating
386 and 583 (assuming a MICBMD of by SCr) within the first 5 days of therapy, that the interplay of multiple factors
1 mg/L) in children 2 to 12 years of age, indicating some degree of renal function in addition to vancomycin exposure
indicating that some younger children recovery, a finding that provides support contributed to AKI.173-176 Interestingly,
may require higher doses to achieve for ongoing vancomycin TDM. In ad- Sinclair et al174 reported that a 5-mg/kg

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


target AUC/MICBMD.162 The probability dition, vancomycin CL does not always dose augmentation or each additional
of target attainment was not provided, correlate well with renal function (as as- day of vancomycin use increased the
and dosages above 60 mg/kg/day were sessed by creatinine CL) in children, par- risk of AKI. Knoderer and colleagues173
not evaluated in this study. ticularly in those who are acutely ill in the evaluated late-onset AKI (defined as
Two retrospective studies that util- ICU setting and have varying degrees of occurring after 7  days of vancomycin
ized non-Bayesian methods evaluated renal dysfunction. Rapid return of renal therapy) and observed that young age
trough concentration targets of 10 to function may occur over the first few days (<1 year) was independently associated
20 mg/L (a higher range than that used after ICU admission. As such, therapeutic with late AKI.
by Le et al164 and Frymoyer et al163, who monitoring of both serum concentrations One pediatric study evaluated the
also assessed AUC) in children 1 month and renal function should be conducted relationship of AKI with vancomycin
to 18 years of age. An interesting finding during vancomycin therapy.170,171 AUC and trough concentrations, both
of the study of Madigan et al166 was that Loading doses.  Loading doses derived by Bayesian estimation. Le
a dosage of 60  mg/kg/day achieved the of 25 to 30 mg/kg for critically ill adults and colleagues19 conducted a large
target trough concentration in only 17% have been suggested to achieve steady- population-based PK analysis using
of preschool-aged children 2 to 5  years state concentrations more quickly, but 1,576 serum concentrations collected
old, which was the lowest attainment preliminary data on pediatric patients from 680 pediatric subjects. A  contin-
for any pediatric age group. Eiland and suggest that the benefit of a loading uous exposure-response relationship
colleagues161 showed that dosages of dose of 30  mg/kg is quickly lost if the was observed, with 10%, 33%, and 57%
70 to 80  mg/kg/day were necessary to maintenance dose is insufficient to of patients who respectively achieved
achieve trough concentrations of 10 to provide adequate ongoing exposure.167 AUC values of ≥400, 800, and 1,000
20 mg/L. Another study, by Abdel et al,168 However, the concept of a loading dose mg·h/L experiencing AKI. Even after
demonstrated that dosages higher than accompanied by a daily maintenance adjusting for ICU stay and concomi-
60 mg/kg/day were necessary to achieve dose sufficient to achieve the target tant use of nephrotoxic drugs, an AUC
an AUC/MIC of ≥400 in children with exposure and initiated at a specified of ≥800 mg·h/L and trough concentra-
cancer. The mean age in this study co- time after the loading dose should be tions of ≥15  mg/L were independently
hort was 6 (SD, 2.5) years; it is possible investigated. associated with a greater than 2.5-fold
that young age with greater CL may have Minimizing AKI risk.  Similar to increased risk of AKI. The linkage of
been a contributing factor for the need the literature on adults, the literature AUC to AKI, along with the strong cor-
for an increased dose, an observation in pediatrics suggests, in aggregate, relation between AUC and trough con-
uncovered in studies by Le et  al164 and that the risk of AKI increases as a func- centrations (Spearman’s coefficient,
Madigan et al.166 tion of vancomycin exposure, espe- 0.963; P  <  0.001), reinforces AUC as a
As a drug that demonstrates renal cially when the trough concentration plausible PK/PD parameter for thera-
elimination, vancomycin requires dosage exceeds 15 to 20  mg/L. In fact, Fiorito peutic monitoring that encompasses
adjustment in children with acute or and colleagues158 reported in a recent both therapeutic and toxic responses.
chronic renal insufficiency. Le and col- meta-analysis of 10 pediatric studies Vancomycin exposure should be main-
leagues169 conducted a population-based that troughs of ≥15 mg/L increased the tained at an AUC of <800 mg·h/L to
PK analysis with Bayesian methods that OR for AKI by 2.7-fold (95% confidence minimize AKI risk. As such, vanco-
evaluated 63 case-control pairs (matched interval, 1.82-4.05) and that AKI was mycin dosages of ≥100  mg/kg/day
by age and weight) with 319 vancomycin further correlated with a stay in the pe- should be avoided given that the pro-
serum concentrations. The mean age of diatric ICU. McKamy and colleagues172 jected median AUC and trough values
this study cohort was 13 (SD, 6) years. The published results of the first study that are 843 mg·h/L and 21  mg/L, respec-
investigators reported that a vancomycin uncovered the association between tively, at a dosage of 100 mg/kg/day.164
dosage of 45  mg/kg/day (ie, 15  mg/kg vancomycin trough concentrations However, enhanced renal clearance of

850    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

vancomycin may transiently occur in in the various pediatric age groups, and concentrations to achieve the AUC/MIC
specific situations in children, in which the differences in tissue site-of-infection target. Early monitoring of observed
case the dose of vancomycin may need drug exposure (eg, common occurrence concentrations is recommended when
to be higher than is usually prescribed of multifocal complicated osteomyelitis doses exceed 2,000 to 3,000 mg/day (A-
to achieve an AUC of 400 mg·h/L, in children requiring therapeutic bone III). Furthermore, close monitoring of
highlighting the need for therapeutic concentrations, with rare occurrence observed concentrations and renal func-
monitoring. of MRSA endocarditis) suggest that fur- tion is prudent in patients with poor or
Therapeutic monitoring. Recent ther studies in children that incorporate augmented renal clearance, as resolu-
literature on vancomycin in pediatrics prospective assessment of clinical out- tion of their renal function abnormal-
has focused primarily on PK analysis comes and large sample size are needed ities may occur within the first 5 days of
to support optimal dosing. Le and col- to identify the optimal dosing strat- therapy.
leagues177 conducted a population- egies for MRSA infections in pediatrics.

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


based PK analysis in 138 pediatric Until additional data are available, the 18. AUC-guided therapeutic moni-
subjects who were more than 3 months AUC target used in adults (ie, from 400 toring for vancomycin, preferably
of age, evaluating 712 serum vanco- up to 600 mg·h/L [assuming a MIC of with Bayesian estimation, is sug-
mycin concentrations (collected mostly 1 mg/L]) appears to be the most appro- gested for all pediatric age groups,
after the third or fourth dose). They priate initial target for vancomycin ex- based on developmental changes
showed that both accuracy and preci- posures in all pediatric age groups. For of vancomycin CL documented
sion for estimating AUC24 (calculated most children across the pediatric age from the newborn to the adolescent.
by total daily dose over vancomycin CL, groups, assuming a vancomycin MIC Based on current available data,
with the integration of Bayesian estima- of 1  mg/L, published data suggest that the suggestion for AUC-guided
tion) were improved using 2 concentra- a dosage of 60 to 80 mg/kg/day (given in monitoring in pediatrics aligns with
tions (peak and trough), compared with divided doses every 6 hours) is required the approach for adults, including
trough-only monitoring. Furthermore, to achieve an AUC target of 400 to 600 the application of Bayesian esti-
the 2-concentration approach im- mg·h/L. mation for 1 trough concentration
proved the prediction of future AUC or first-order PK equations with 2
exposure in patients.177 Another study, Summary and concentrations (B-II). The Bayesian
by Stockmann et  al,178 evaluated AUC‐ recommendations: AUC-guided dosing strategy may be
based vancomycin monitoring in 23 an optimal approach to individu-
pediatric patients with cystic fibrosis. 17. Based on an AUC target of 400 alize vancomycin therapy in pediat-
The researchers demonstrated that mg·h/L (but potentially up to 600 rics since it can incorporate varying
2 concentrations calculated using a mg·hr/L, assuming a vancomycin ages, weights, and renal function.
standard PK equation and a trough MIC of ≤1 mg/L for MRSA) from Both serum concentrations of van-
concentration calculated using a adult data, the initial recommended comycin and renal function should
Bayesian population-based PK model vancomycin dosage for children with be monitored since vancomycin CL
produced similar AUC estimations. normal renal function and suspected and CLcr are not always well correl-
Despite the availability of limited serious MRSA infections (including ated in pediatrics. Furthermore, ag-
studies on vancomycin monitoring in pneumonia, pyomyositis, multifocal gressive dosing to maintain target
pediatrics, the findings appear con- osteomyelitis, complicated bacte- AUC exposure and decrease the
gruent with adult data supporting remia, and necrotizing fasciitis) is: risk of potential AKI in treatment
AUC-guided therapeutic moni- of MRSA infection necessitates drug
toring that incorporates the Bayesian 60 to 80 mg/kg/day, in divided
•  monitoring.
method, especially if only a single doses given every 6 hours, for chil- 19. Therapeutic monitoring may begin
trough concentration is available. dren ages 3 months to less than within 24 to 48 hours of vancomycin
Furthermore, this AUC-guided moni- 12 years or therapy for serious MRSA infections
toring approach also appears prudent 60 to 70 mg/kg/day, in divided
•  in children, as in adults (B-III). Any
in order to predict toxicity in light of doses given every 6 to 8 hours, for delay in therapeutic monitoring
AKI data in pediatrics. those ≥12 years old (A-II). should be based on severity of infec-
Overall, there are limited pediatric tion and clinical judgment. Dosing
outcomes data to support the AUC The maximum empiric daily dose is adjustment should be made for those
target correlated with drug effectiveness usually 3,600  mg in children with ad- with renal insufficiency, those with
in adults. Some of the differences found equate renal function (C-III). Most obesity (see Pediatric Obesity), and
between adults and children treated for children generally should not require those receiving concurrent nephro-
MRSA infections with vancomycin in- more than 3,000  mg/day, and doses toxic drug therapy. Following the
clude the complexity of vancomycin CL should be adjusted based on observed initial dose, dosing adjustment is

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  851


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

important for those with acute renal studies by Heble et  al181 and Miller the use of a 20-mg/kg loading dose
insufficiency, but subsequent adjust- et al182 documented higher vancomycin based on total body weight in obese
ment (particularly within the first trough concentrations in overweight children increased achievement of an
5 days of therapy) may be necessary and obese children, as compared to AUC/MIC of ≥400, especially within the
for those experiencing recovery of normal-weight children, with dosing first 12 hours of therapy. In addition, 1
renal function. Sustained or subse- based on total body weight. No increase of every 5 obese children had an AUC
quent decreases in dosage may be in AKI was noted in the overweight of ≥ 800 mg·h/L, indicating that routine
needed, particularly for those with children.182 therapeutic and safety monitoring is
chronic renal insufficiency and those Collectively, non-Bayesian studies prudent.188
receiving concurrent nephrotoxic of obese children have evaluated main-
drug therapy (B-III). tenance regimens ranging from 40 to Summary and
20. Vancomycin exposure may be op- 80  mg/kg/day (calculated using total recommendations:

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


timally maintained below the thresh- body weight), with some instituting
olds for AUC of 800 mg·h/L and for maximum doses of 1 to 2 g over 1 to 2 22. Data suggest that obese children are
trough concentrations of 15 mg/L to hours.180,181,183,184 As an alternative to likely to have vancomycin exposures
minimize AKI (B-II). The safety of total body weight, one study recom- that may be statistically greater than
vancomycin dosages above 80 mg/ mended the use of body surface area to those in normal-weight children when
kg/day has not been prospectively dose vancomycin, which necessitates doses are calculated on a mg/kg basis,
evaluated. Avoiding vancomycin establishing a different dosing regimen but these differences are not known
dosages of ≥100 mg/kg/day is sug- and obtaining height measurements to be of sufficient clinical importance
gested since they are likely to surpass that may not always be readily avail- to suggest different mg/kg empiric
these thresholds (B-III). able in clinical practice.185 Body sur- vancomycin dosages in obese children
21. Insufficient data exist on which face area is not typically used for dosing at this time. Similar to nonobese chil-
to base a recommendation for a medications in children, except for dren, obese children < 12 years old,
loading dose among the nonobese chemotherapeutic agents.186 compared with those ≥ 12 years, may
pediatric population. Loading Using a Bayesian population-based require a higher mg/kg dose (B-II).
doses from adult studies may be PK analysis of 389 vancomycin serum 23. Therapeutic monitoring is likely to be
considered, but further studies are concentrations collected from 87 of particular value in obese children,
needed to elucidate the appropriate pairs of obese and nonobese children both for therapeutic response and
dose for the various pediatric popu- (matched by age and baseline SCr), Le the risk of AKI. The specific recom-
lations from the neonate to adoles- and colleagues187 showed that the Vd mendations for therapeutic moni-
cent (C-III). was strongly correlated with actual or toring in nonobese children may also
total body weight and that CL correl- apply for obese children (B-II).
Pediatric obesity.  Vancomycin ated with allometric weight (ie, weight x 24. A loading dose of 20 mg/kg by total
is a large glycopeptide molecule that 0.75) and body surface area. Using this body weight is recommended in
is hydrophilic, suggesting that distri- PK model, Nguyen and colleagues164,166 obese children (A-III).
bution into tissues with high lipid con- concluded, using Monte Carlo simu-
centrations, such as adipose tissue, is lations with Bayesian estimation, that Neonates.  Vancomycin ther-
decreased, as noted above for adults vancomycin 60  mg/kg/day dosed by apeutic monitoring is important in
(see Dosing in Obesity section). When total body weight, as compared with neonates, based on developmental
vancomycin dosing is based on total other weight measures, resulted in the considerations of prominent increasing
body weight (mg/kg) for both obese highest rate of achievement of the target renal function that occurs over the
and nonobese children, serum con- AUC/MIC of ≥400 in obese children (ie, first several weeks of life.189 Models to
centrations have been documented to the target was achieved in 76% when predict vancomycin dosing have var-
be higher in obese children, assuming vancomycin was given by total body iously incorporated weight-based
that renal CL is similar between the 2 weight, in 66% when given by adjusted dosing, chronologic age–based dosing,
populations.179 Moffett retrospectively body weight, and in 31% when given postmenstrual age–based dosing, SCr-
compared vancomycin pharmacoki- by allometric weight). Furthermore, based dosing (except for the first week of
netics in 24 obese children who were when given dosages of vancomycin life, when transplacental maternal cre-
matched with 24 nonobese control of 60  mg/kg/day by total body weight, atinine in the neonatal circulation ren-
children.180 Vancomycin dose admin- fewer obese children of <12 vs ≥12 years ders neonatal SCr values inaccurate in
istration per child was slightly higher of age achieved an AUC/MIC of ≥400 estimating renal function), or combin-
in the obese children, which resulted (70% and 84%, respectively), an age- ations of these strategies. Regardless of
in increased trough concentrations. based observation also identified in which model is used, therapeutic moni-
Similarly, 2 retrospective non-Bayesian nonobese children.164,166 Interestingly, toring in the neonate is essential due to

852    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

the rapid maturation of renal function PK analysis was conducted to create not be evaluated rigorously in this study
over the first weeks of life. a model for vancomycin CL that was due to the small sample size. Overall,
Mehrotra et al190 compared 4 models based on weight, postmenstrual age, the clinical utility of CI in neonates re-
for predicting vancomycin serum con- and SCr (measured by a modified ki- quires further evaluation, as the most
centrations, based on their population netic Jaffé reaction). Monte Carlo common pathogen causing late-onset
PK model, using a standard weight- simulations with Bayesian estimation sepsis requiring vancomycin therapy is
based dose, a postmenstrual age–based demonstrated that trough concentra- Staphylococcus epidermidis, with limited
dose, a postmenstrual and postnatal tions ranging from 7 to 11  mg/L were cases of S.  aureus sepsis. While the op-
age–based dose, and a SCr-based dose. highly predictive of an AUC24 of >400 timal AUC/MIC target for S. epidermidis
Serum creatinine–based dosing pre- mg·h/L in at least 90% of neonates. is not well studied, a lower target may be
dicted trough concentrations with the Dosages to achieve this PK/PD target reasonable, but further data to support
smallest variability in both term and pre- ranged from 15 to 20 mg/kg every 8 to 12 this recommendation are needed.

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


term neonates. However, for those who hours, depending on postmenstrual age The incidence of vancomycin-
wish to achieve a target exposure based and SCr.194 Stockmann et al196 later sup- associated AKI reported in neonates
on high trough concentrations within a ported the predictive performance and has been low, ranging from 1% to
narrow range of 15 to 20 mg/L, it should generalizability of this model in their 9%.200 Nonetheless, a positive correla-
be noted that only 13% to 21% of neo- study of 243 neonates with 734 vanco- tion between increasing vancomycin
nates were within this range across the mycin concentrations. While a trough trough concentrations and AKI has
4 dosing regimens. Marqués-Miñana concentration of 11 mg/L predicted the been reported by Bhargava et  al.201
et  al191 also developed a population PK attainment of an AUC of ≥400 mg·h/L Furthermore, in a large, retrospec-
model and proposed dosing based on in 93% of neonates, Stockmann and tive, multicenter, propensity score–
postmenstrual age. SCr-based rather colleagues noted that a trough concen- matched cohort study of 533 neonates
than postmenstrual or postconceptional tration alone did not precisely predict receiving vancomycin and gentamicin
age–based dosing has been supported AUC and concluded that Bayesian ap- and 533 receiving gentamicin alone,
by Irikura et al192 and Capparelli et al.193 proaches to support vancomycin dosing Constance et al202 concluded that AKI
However, when evaluating published decisions for neonates in the clinical was not associated with vancomycin
neonatal PK models, no consensus on setting are needed.196 Furthermore, Cies alone but may occur in the presence
an optimal dosing regimen was achieved et  al197 reported differences in vanco- of other recognized risk factors, in-
by experts on neonatal vancomycin, mycin pharmacokinetics, in particular cluding patent ductus arteriosus,
Zhao et  al reported.194 After evaluating rapid vancomycin CL, in neonates with concomitant nonsteroidal anti-in-
the predictive performance of 6 models, extracorporeal oxygenation life sup- flammatory drug use, 1 or more pos-
Zhao et al concluded the importance of port, reiterating the need for Bayesian- itive blood cultures, low birth weight,
evaluating analytical techniques for SCr derived dosing decision support in this and higher scores for severity of ill-
and vancomycin concentrations best vulnerable population. Lastly, Leroux ness and risk of mortality.
explained the variability of predictions et al198 demonstrated the success of the
between the models. Zhao et  al found clinical integration of a model-based Summary and
that the Jaffé method overestimated SCr vancomycin dosing calculator, devel- recommendations:
concentrations when compared to the oped from a population PK study, that
enzymatic method and that for vanco- was successful in improving the rate of 25. Doses recommended to achieve an
mycin concentrations, the fluorescence attainment of a serum concentration of AUC of 400 mg·h/L (assuming a MIC
polarization immunoassay method 15 to 25 mg/L from 41% to 72% without of 1 mg/L) in neonates and infants
and enzyme-multiplied immunoassay any cases of AKI. up to 3 months old range from 10
method assays showed different predic- As an alternative to intermittent ad- to 20 mg/kg every 8 to 48 hours,
tive performances as well.194 ministration, CI of vancomycin has been depending on postmenstrual age,
With the knowledge that AUC, as evaluated in infants. In a multicenter, weight, and SCr (A-II). AUC-guided
compared with trough concentrations, randomized controlled trial involving therapeutic dosing and monitoring,
is a more achievable target in pediat- 111 infants less than 90 days of age, Gwee preferably with Bayesian estimation,
rics, Frymoyer and colleagues195 evalu- et al199 showed that the use of CI resulted can best achieve the target vanco-
ated the association between AUC and in fewer dose adjustments and a lower mycin exposure likely to be required
trough concentrations in neonates. mean daily dose than intermittent ad- for a successful outcome of treat-
Using 1,702 vancomycin concentrations ministration. The target trough concen- ment for a MRSA infection for all
(measured by the homogenous particle- trations were 10 to 20 mg/L for II, and the neonates, regardless of gestational
enhanced turbidimetric inhibition steady-state concentrations were 15 to and chronologic age. The specific
immunoassay) collected from 249 neo- 25 mg/L for CI. The AUC and clinical out- recommendations for AUC-guided
nates up to 3 months of age, population comes, including nephrotoxicity, could therapeutic monitoring in children

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  853


Table 2. Primary Recommendations for Vancomycin Dosing and Therapeutic Drug Monitoring

854 
A. ADULTS AND PEDIATRIC PATIENTS
1. In patients with suspected or definitive serious MRSA infections, an individualized target AUC/MICBMD ratio of 400 to 600 (assuming a vancomycin MICBMD of 1 mg/L) should be
advocated to achieve clinical efficacy while improving patient safety (A-II).
2. When transitioning to AUC/MIC monitoring, clinicians should conservatively target AUCs for patients with suspected or documented serious infections due to MRSA, assuming
a vancomycin MICBMD of 1 mg/L or less at most institutions. Given the importance of early, appropriate therapy, vancomycin targeted exposure should be achieved early during
ASHP REPORT

the course of therapy, preferably within the first 24 to 48 hours (A-II). As such, the use of Bayesian-derived AUC monitoring may be prudent in these cases since it does not re-
quire steady-state serum vancomycin concentrations to allow for early assessment of AUC target attainment.
3. Trough-only monitoring, with a target of 15 to 20 mg/L, is no longer recommended, based on efficacy and nephrotoxicity data in patients with serious infections due to MRSA
(A-II). There is insufficient evidence to provide recommendations on whether trough-only or AUC-guided vancomycin monitoring should be used among patients with noninva-
sive MRSA or other infections.
4. Vancomycin monitoring is recommended for patients receiving vancomycin for serious MRSA infections to achieve a sustained targeted AUC (assuming a MICBMD of 1 mg/L
unless it is known to be greater or less than 1 mg/L by BMD). Independent of MRSA infection, vancomycin monitoring is also recommended for all patients at high risk for
nephrotoxicity (eg, critically ill patients receiving concurrent nephrotoxins), patients with unstable (ie, deteriorating or significantly improving) renal function, and those receiving
prolonged courses of therapy (more than 3 to 5 days). We suggest the frequency of monitoring be based on clinical judgment; frequent or daily monitoring may be prudent for
hemodynamically unstable patients (eg, those with end-stage renal disease), with once-weekly monitoring for hemodynamically stable patients (B-II).
5. Based on current national vancomycin susceptibility surveillance data, under most circumstances for empiric dosing, the vancomycin MIC should be assumed to be 1 mg/L.
When the MICBMD is greater than 1 mg/L, the probability of achieving an AUC/MIC target of ≥400 is low with conventional dosing; higher doses may risk unnecessary tox-
icity, and the decision to change therapy should be based on clinical judgment. In addition, when the MICBMD is less than 1 mg/L, we do not recommend decreasing the dose
to achieve the AUC/MIC target. It is important to note the limitations in automated susceptibility testing methods, including the lack of precision and variability in MIC results
depending on the method used (B-II).
6. The pharmacokinetics of continuous infusion suggest that such regimens may be a reasonable alternative to conventional intermittent infusion dosing when the AUC target
cannot be achieved (B-II).
7. Incompatibility of vancomycin with other drugs commonly coadministered in the ICU requires the use of independent lines or multiple catheters when vancomycin is being con-

  AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


sidered for continuous infusion (A-III).
B. ADULTS
8. Given the narrow vancomycin AUC range for therapeutic effect and minimal associated risk of acute kidney injury (AKI), the most accurate and optimal way to manage vanco-
mycin dosing should be through AUC-guided dosing and monitoring (A-II). We recommend to accomplish this in one of two ways:
a. One approach relies on the collection of 2 concentrations (obtained near the steady-state, post-distributional peak concentration at 1 to 2 hours after infusion and trough at end
of dosing interval), preferably but not required during the same dosing interval (if possible) and utilizing first-order pharmacokinetic (PK) equations to estimate the AUC (A-II).
b. The preferred approach to monitor AUC involves the use of Bayesian software programs, embedded with a PK model based on richly sampled vancomycin data as the
Bayesian prior, to optimize the delivery of vancomycin based on the collection of 1 or 2 vancomycin concentrations, with at least 1 trough. It is preferred to obtain 2 PK samples
(ie, 1 to 2 hours post infusion and at the end of the dosing interval) to estimate the AUC with the Bayesian approach (A-II). A trough concentration alone may be sufficient to
estimate the AUC with the Bayesian approach in some patients, but more data across different patient populations are needed to confirm the viability of using trough-only data
(B-II).
9. Doses of 15 to 20 mg/kg (based on actual body weight) administered every 8 to 12 hours as an intermittent infusion are recommended for most patients with normal renal func-
tion when assuming a MICBMD of 1 mg/L (A-II). In patients with normal renal function, these doses may not achieve therapeutic AUC/MIC targets when the MIC is 2 mg/L.

Continued on next page


GUIDELINE ON VANCOMYCIN MONITORING

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022



Continued from previous page
Table 2.  Primary Recommendations for Vancomycin Dosing and Therapeutic Drug Monitoring
B. ADULTS

10. Continuous Infusion: Based on current available data, a loading dose of 15 to 20 mg/kg, followed by daily maintenance CI of 30 to 40 mg/kg (up to 60 mg/kg), to achieve a
target steady-state concentration of 20 to 25 mg/L may be considered for critically ill patients (B-II). AUC24 can be simply calculated when multiplying the steady-state concen-
tration (ie, desired therapeutic range of 20 to 25 mg/L throughout the entire dosing interval) by a factor of 24 (B-II). Attaining the desired drug exposure may be more readily
accomplished, given the ease of sampling time and dosage adjustment, by changing the rate of infusion, which is a highly desirable feature in critically ill patients (B-II).
11. The risk of developing nephrotoxicity with CI appears to be similar or lower compared to intermittent dosing when targeting a steady-state concentration of 15 to 25 mg/L and
a trough concentration of 10 to 20 mg/L, respectively (B-II). Definitive studies are needed to compare drug exposure based on measured AUC24 and factors that predispose to
development of nephrotoxicity, such as receipt of concomitant nephrotoxins, diuretics, and/or vasopressor therapy in patients receiving continuous vs intermittent infusion of
vancomycin.
GUIDELINE ON VANCOMYCIN MONITORING

12. In order to achieve rapid attainment of targeted concentrations in critically ill patients with suspected or documented serious MRSA infections, a loading dose of 20 to 35 mg/
kg can be considered for intermittent administration of vancomycin (B-II). Loading doses should be based on actual body weight and not exceed 3,000 mg. More intensive
and early therapeutic monitoring should also be performed in obese patients (B-II).
13. Adult Obesity: A vancomycin loading dose of 20 to 25 mg/kg using actual body weight, with a maximum of 3,000 mg, may be considered in obese adult patients with serious
infections (B-II). Empiric maintenance doses for most obese patients usually do not exceed 4,500 mg/day, depending on their renal function (B-II). Early and frequent moni-
toring of AUC exposure is recommended for dose adjustment, especially when empiric doses exceed 4,000 mg/day (A-II).
14. Intermittent Hemodialysis: Since efficacy data are unavailable for an AUC of <400 mg · h/L, monitoring based on predialysis serum concentrations and extrapolating these
values to estimate AUC is most practical. Maintaining predialysis concentrations between 15 and 20 mg/L is likely to achieve the AUC of 400 to 600 mg · h/L in the previous
24 hours (C-III). Predialysis serum concentration monitoring should be performed not less than weekly and should drive subsequent dosing rather than a strict weight-based
recommendation, although these recommended doses provide a useful starting point until serum concentrations have been determined (B-II).
15. Hybrid Dialysis Therapies (eg, Slow-Low Efficiency Dialysis [SLED]): Loading doses of 20 to 25 mg/kg actual body weight should be used, recognizing that these hybrid dialysis
therapies efficiently remove vancomycin (B-III). Initial doses should not be delayed to wait for a dialysis treatment to end. Maintenance doses of 15 mg/kg should be given
after hybrid hemodialysis ends or during the final 60 to 90 minutes of dialysis, as is done with standard hemodialysis (B-III). Concentration monitoring should guide further
maintenance doses.
16. 
Continuous Renal Replacement Therapy (CRRT): Loading doses of 20 to 25 mg/kg by actual body weight should be used in patients receiving CRRT at conventional, KDIGO-
recommended effluent rates of 20 to 25 mL/kg/h (B-II). Initial maintenance dosing for CRRT with effluent rates of 20 to 25 mL/kg/h should be 7.5 to 10 mg/kg every 12 hours (B-
II). Maintenance dose and dosing interval should be based on serum concentration monitoring, which should be conducted within the first 24 hours to ensure AUC/MIC targets
are met. In fluid overloaded patients, doses may be reduced as patients become euvolemic and drug Vd decreases. The use of CI vancomycin in patients receiving CRRT ap-
pears to be growing, and this method could be used in place of intermittent vancomycin dosing, especially when high CRRT ultrafiltrate/dialysate flow rates are employed (B-II).
C. PEDIATRIC PATIENTS
17. Based on an AUC target of 400 mg · h/L (but potentially up to 600 mg · hr/L assuming a MIC of ≤1 mg/L) from adult data, the initial recommended vancomycin dosage for chil-
dren with normal renal function and suspected serious MRSA infections is 60 to 80 mg/kg/day, divided every 6 to 8 hours, for children ages 3 months and older (A-II).

Continued on next page


ASHP REPORT

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  855


Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022
856 
Continued from previous page
Table 2.  Primary Recommendations for Vancomycin Dosing and Therapeutic Drug Monitoring
C. PEDIATRIC PATIENTS
ASHP REPORT

18. The maximum empiric daily dose is usually 3,600 mg/day in children with adequate renal function (C-III). Most children generally should not require more than 3,000 mg/day,
and doses should be adjusted based on observed concentrations to achieve the AUC/MIC target. Early monitoring of observed concentrations is recommended when doses
exceed 2,000 to 3,000 mg/day (A-III). Furthermore, close monitoring of observed concentrations and renal function is prudent in patients with poor or augmented renal clear-
ance as resolution of their renal function may occur within the first 5 days of therapy.
19. AUC-guided therapeutic monitoring for vancomycin, preferably with Bayesian estimation, is suggested for all pediatric age groups, based on developmental changes of vanco-
mycin CL documented from the newborn to the adolescent. Based on current available data, the suggestion for AUC-guided monitoring in pediatrics aligns with the approach
for adults, including the application of Bayesian estimation for 1 trough concentration, or first-order PK equations with 2 concentrations (B-II). The Bayesian AUC-guided
dosing strategy may be an optimal approach to individualize vancomycin therapy in pediatrics since it can incorporate varying ages, weights, and renal function. Both serum
concentrations of vancomycin and renal function should be monitored since vancomycin CL and creatinine CL are not always well correlated in pediatrics. Furthermore, ag-
gressive dosing to maintain target AUC exposure and decrease the risk of potential AKI in treatment of MRSA infection necessitates drug monitoring.
20. Therapeutic monitoring may begin within 24 to 48 hours of vancomycin therapy for serious MRSA infections in children, as in adults (B-III). Any delay in therapeutic monitoring
should be based on severity of infection and clinical judgment. Dosing adjustment should be made for those with renal insufficiency, or those with obesity, or for those re-
ceiving concurrent nephrotoxic drug therapy. Following the initial dose, dosing adjustment is important for those with acute renal insufficiency, but subsequent adjustment (par-
ticularly within the first 5 days of therapy) may be necessary for those experiencing recovery of renal function. Sustained or subsequent decreases in dosage may be needed,
particularly for those with chronic renal insufficiency and those receiving concurrent nephrotoxic drug therapy (B-III).
21. Vancomycin exposure may be optimally maintained below the thresholds for AUC of 800 mg · h/L and for trough concentrations of 15 mg/L to minimize AKI (B-II). The safety
of vancomycin above 80 mg/kg/day has not been prospectively evaluated. Avoiding vancomycin dosages of ≥100 mg/kg/day is suggested since they are likely to surpass
these thresholds (B-III).

  AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


22. Insufficient data exist on which to base a recommendation for a loading dose among the nonobese pediatric population. Loading doses from adult studies may be considered,
but further studies are needed to elucidate the appropriate dose for the various pediatric populations, from neonates to adolescents (C-III).
23. Pediatric Obesity: Data suggest that obese children are likely to have vancomycin exposures that may be statistically greater than in normal-weight children when doses are
calculated on a mg/kg basis, but these differences are not known to be of sufficient clinical importance to suggest different mg/kg empiric vancomycin dosages in obese chil-
dren at this time. Similar to nonobese children, obese children < 12 years old, compared with those ≥ 12 years, may require higher mg/kg doses (B-II).
24. Pediatric Obesity: Therapeutic monitoring is likely to be of particular value in obese children, both for therapeutic response and to minimize the risk of AKI. The specific re-
commendations for therapeutic monitoring in nonobese children may also apply for obese children (B-II). A loading dose of 20 mg/kg by total body weight is recommended in
obese children (A-III).
25. Neonates: Dosages recommended to achieve an AUC of 400 mg · hr/L (assuming a MIC of 1 mg/L) in neonates and infants up to 3 months old range from 10 to 20 mg/kg
every 8 to 48 hours depending on postmenstrual age, weight, and SCr (A-II).

Abbreviations (not defined in body of table): AUC, area under the curve; BMD, broth micodilution; CL, clearance; ICU, intensive care unit; KDIGO, Kidney Disease: Improving Global Outcomes; MIC,
minimum inhibitory concentration; MRSA, methicillin-resistant Staphylococcus aureus; SCr, serum creatinine; Vd, volume of distribution.
GUIDELINE ON VANCOMYCIN MONITORING

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

should also apply for neonates (see Disclosures was a speaker at the American Society for
recommendation 18, A-III). Dr. Wong-Beringer received a grant from Microbiology and European Society for
Merck & Co. and consulted for Rempex Clinical Microbiology and Infectious Diseases
Pharmaceuticals, INSMED, Merck & Co., ASM/ESCMID conference; served on the 2015–
Conclusion Nabriva Therapeutics, GlaxoSmithKline, 2019 Program Committee and was 2016–2018
Paratek Pharmaceuticals, Achaogen, Inc., Program Co-Chairperson for the American
To optimize vancomycin use for the
Bayer HealthCare, and SIGA Technologies. Microbiology Society; and was a member
treatment of serious infections caused of the 2017–2019 Antimicrobial Resistance
Dr. Bradley served on a planning committee
by MRSA, we recommend targeting an for the US Food and Drug Administration Committee for IDSA. Dr. Rybak received re-
AUC/MICBMD ratio of 400 to 600 (as- and European Medicines Agency at the search grants from Bayer Pharmaceuticals, the
suming an MICBMD of 1  mg/L) for em- American College of Clinical Pharmacy’s NIH Research Project Grant (RO1) Program,
annual meeting; participated in the design Merck, Allergan, the Michigan Department
piric dosing in both adult and pediatric
of a a pediatric clinical trial; served on an of Health and Human Services, Accelerate
patients to maximize clinical efficacy Diagnostics, Inc., NIH, Contrafect, Motif
executive committee for the United States
and minimize AKI risk. Furthermore, Biosciences, and the Michigan Translational

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


Committee on Antimicrobial Susceptibility
the AUC should be therapeutically Testing; and consulted for Achaogen, Research and Commercialization Program;
monitored using 1 or 2 postdose con- Allergan, ContraFect, GlaxoSmithKline, and served as a grant review panel member
Janssen, Melinta, Merck, Nabrive, Pfizer, for NIH. The other authors have declared no
centrations (ie, a peak concentration
Theravance, and Zavante. Dr. Liu received a potential conflicts of interest.
measured after the early vancomycin
research grant from Nohla Therapeutics and
tissue distribution phase and a trough was a member of an Independent Efficacy References
level measured prior to the next dose), Adjudication Committee with Theravance. 1. Rybak MJ, Lomaestro BM,
preferably integrating the Bayesian ap- Dr. Le received research awards from the Rotschafer JC, et al. Therapeutic moni-
proach. The primary recommendations Sternfels Prize for Drug Safety Innovation, toring of vancomycin in adult patients:
are summarized in Table  2. The suc- Duke University, the National Institutes of a consensus review of the American
Health (NIH) and National Institute of Child Society of Health-System Pharmacists,
cessful use of these guideline recom- Health and Human Development, and JMI the Infectious Diseases Society of
mendations to positively impact patient Laboratories; was an invited advisory board America, and the Society of Infectious
outcomes may require multifaceted member for FDA, the Asian Pacific Health Diseases Pharmacists. Am J Health-Syst
interventions, including educational Foundation, and Infectious Diseases and Pharm. 2009; 66(1):82-98.
meetings, guideline implementation, Therapy journal. Dr. Levine served on the data 2. The periodic health examination.
safety monitoring board for Contrafect and Canadian Task Force on the Periodic
and dissemination of educational ma- served on an adjudication panel for Novartis. Health Examination. Can Med Assoc J.
terial on vancomycin dosing, moni- Dr. Lodise received grants from the Antibiotic 1979; 121(9):1193-1254.
toring, and nephrotoxicity.203,204 While Resistance Leadership Group (ARLG), Merck 3. Craig WA. Basic pharmacodynamics
valuable literature pertaining to adults, & Co, and Motif Bio PLC; served as a health of antibacterials with clinical appli-
children, and neonates has emerged outcomes project consultant for Paratek cations to the use of beta-lactams,
Pharmaceuticals, Allergan, Merck & Co., glycopeptides, and linezolid.
since the last vancomycin guideline, and Melinta Therapeutics; served on advi- Infect Dis Clin North Am. 2003;
future studies in all patient popula- sory boards for Paratek Pharmaceuticals, 17(3):479-501.
tions are necessary to address existing Motif Bio PLC, Achaogen, Nabriva, and 4. Drusano GL. Antimicrobial pharma-
gaps, including (1) efficacy data to sup- Tetraphase; served as a consultant to Paratek codynamics: critical interactions of
port vancomycin use in specific pa- Pharmaceuticals, ARLG, Allergan, Merck & ‘bug and drug’. Nat Rev Microbiol. 2004;
Co., Melinta Therapeutics, Motif Bio PLC, 2(4):289-300.
tient populations (including neonates Achaogen, Nabriva, and Tetraphase; and 5. Liu C, Bayer A, Cosgrove SE, Daum RS
and pediatric patients and patients was a speaker for Melinta Therapeutics, et al. Clinical practice guidelines by The
with renal disease and obesity) and for Tetraphase, and Sunovion. Dr. Maples served Infectious Diseases Society of America
other types of infections, (2) efficacy as an international working group member for the treatment of methicillin-
data for specific pathogens, including for the European Cystic Fibrosis Society and resistant Staphylococcus aureus
North American Cystic Fibrosis Society, infections in adults and children: execu-
coagulase-negative staphylococcus served on an advisory panel for the Centers tive summary. Clin Infect Dis. 2011;
and Streptococcus species; (3) robust for Disease Control and Prevention and Pew 52(3):285-292.
pediatric efficacy data for MRSA and Charitable Trust, and was on a committee for 6. Rybak MJ. The pharmacokinetic and
other gram-positive pathogens causing the Arkansas Health Department. Dr. Mueller pharmacodynamic properties of van-
different types of serious infections; received research grants from Merck & Co. comycin. Clin Infect Dis. 2006; 42(Suppl
and Hope Pharmaceutical and served on an 1):S35-S39.
(4) optimal loading and maintenance advisory board for NxStage and Baxter. Dr. 7. Kullar R, Leonard SN, Davis SL et al.
dosing regimens for patients with obe- Pai received a grant from Merck, Inc., served Validation of the effectiveness of a van-
sity and renal insufficiency; (5) efficacy on an advisory board for Shinogi and Paratek comycin nomogram in achieving target
benefit and the need for a dosing al- Pharmaceuticals, and served on the meet the trough concentrations of 15-20 mg/L
gorithm (specifically incorporating a professor program for Merck. Dr. Rodvold re- suggested by the vancomycin con-
ceived a grant from Theravance Biopharm, sensus guidelines. Pharmacotherapy.
loading dose followed by maintenance NIH, ARLG, and Allergan; consulted for BLC, 2011; 31(5):441-448.
infusion); and (6) toxicodynamics of Entasis, Merck, Paratek Pharmaceuticals, 8. Singh NB, Yim J, Jahanbakhsh S,
vancomycin CI in critically ill patients. Shionogi, Tetraphase, and Wockhardt; Sakoulas G, Rybak MJ. Impact of

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  857


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

cefazolin co-administration with patients with methicillin-resistant 28. Roy AK, Mc Gorrian C, Treacy C et al.
vancomycin to reduce develop- Staphylococcus aureus infective endo- A comparison of traditional and novel
ment of vancomycin-intermediate carditis. Antimicrob Agents Chemother. definitions (RIFLE, AKIN, and KDIGO)
Staphylococcus aureus. Diagn Microbiol 2015; 59(6):2978-2985. of acute kidney injury for the prediction
Infect Dis. 2018; 91(4):363-370. 18. Mogle BT, Steele JM, Seabury RW, of outcomes in acute decompensated
9. Vidaillac C, Gardete S, Tewhey R Dang UJ, Kufel WD. Implementation of heart failure. Cardiorenal Med. 2013;
et al. Alternative mutational path- a two-point pharmacokinetic AUC- 3(1):26-37.
ways to intermediate resistance to based vancomycin therapeutic drug 29. van Hal SJ, Paterson DL, Lodise TP.
vancomycin in methicillin-resistant monitoring approach in patients with Systematic review and meta-analysis
Staphylococcus aureus. J Infect Dis. methicillin-resistant Staphylococcus of vancomycin-induced nephrotoxicity
2013; 208(1):67-74. aureus bacteraemia. Int J Antimicrob associated with dosing schedules that
10. Moise-Broder PA, Forrest A, Agents. 2018; 52(6):805-810. maintain troughs between 15 and 20
Birmingham MC, Schentag JJ. 19. Le J, Ny P, Capparelli E et al. milligrams per liter. Antimicrob Agents
Pharmacodynamics of vancomycin and Pharmacodynamic characteristics of Chemother. 2013; 57(2):734-744.
other antimicrobials in patients with nephrotoxicity associated with vanco- 30. Sinha Ray A, Haikal A, Hammoud KA,

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


Staphylococcus aureus lower respiratory mycin use in children. J Pediatric Infect Yu AS. Vancomycin and the risk of
tract infections. Clin Pharmacokinet. Dis Soc. 2015; 4(4):e109-e116. AKI: a systematic review and meta-
2004; 43(13):925-942. 20. Finch NA, Zasowski EJ, Murray KP analysis. Clin J Am Soc Nephrol. 2016;
11. Kullar R, Davis SL, Levine DP, Rybak MJ. et al. A quasi-experiment to study the 11(12):2132-2140.
Impact of vancomycin exposure on impact of vancomycin area under 31. Singbartl K, Kellum JA. AKI in the ICU:
outcomes in patients with methicillin- the concentration-time curve-guided definition, epidemiology, risk stratifi-
resistant Staphylococcus aureus bacte- dosing on vancomycin-associated neph- cation, and outcomes. Kidney Int. 2012;
remia: support for consensus guidelines rotoxicity. Antimicrob Agents Chemother. 81(9):819-825.
suggested targets. Clin Infect Dis. 2011; 2017; 61(12):e01293-17. 32. Linder A, Fjell C, Levin A, Walley KR,
52(8):975-981. 21. Zasowski EJ, Murray KP, Trinh TD et al. Russell JA, Boyd JH. Small acute in-
12. Brown J, Brown K, Forrest A. Identification of vancomycin exposure- creases in serum creatinine are associ-
Vancomycin AUC24/MIC ratio in pa- toxicity thresholds in hospitalized ated with decreased long-term survival
tients with complicated bacteremia patients receiving intravenous vanco- in the critically ill. Am J Respir Crit Care
and infective endocarditis due to mycin. Antimicrob Agents Chemother. Med. 2014; 189(9):1075-81.
methicillin-resistant Staphylococcus 2018; 62(1):e01684-17. 33. Suzuki Y, Kawasaki K, Sato Y et al. Is
aureus and its association with attrib- 22. Neely MN, Kato L, Youn G et al. peak concentration needed in thera-
utable mortality during hospitalization. Prospective trial on the use of trough peutic drug monitoring of vancomycin?
Antimicrob Agents Chemother. 2012; concentration versus area under the A pharmacokinetic-pharmacodynamic
56(2):634-638. curve to determine therapeutic van- analysis in patients with methicillin-
13. Holmes NE, Turnidge JD, Munckhof WJ comycin dosing. Antimicrob Agents resistant Staphylococcus aureus
et al. Vancomycin AUC/MIC ratio Chemother. 2018; 62(2):e02042-17. pneumonia. Chemotherapy. 2012;
and 30-day mortality in patients with 23. Lodise TP, Rosenkranz SL, Finnemeyer M 58(4):308-312.
Staphylococcus aureus bacteremia. et al. The emperor’s new clothes: pro- 34. Lodise TP, Patel N, Lomaestro BM,
Antimicrob Agents Chemother. 2013; spective observational evaluation of the Rodvold KA, Drusano GL. Relationship
57(4):1654-1663. association between initial vancomycin between initial vancomycin
14. Gawronski KM, Goff DA, Brown J, exposure and failure rates among adult concentration-time profile and neph-
Khadem TM, Bauer KA. A steward- hospitalized patients with MRSA blood- rotoxicity among hospitalized patients.
ship program’s retrospective evalua- stream infections (PROVIDE) [published Clin Infect Dis. 2009; 49(4):507-514.
tion of vancomycin AUC24/MIC and online ahead of print June 3, 2019]. Clin 35. Aljefri DM, Avedissian SN, Rhodes NJ,
time to microbiological clearance in Infect Dis. doi.10.1093/cid/ciz460. Postelnick MJ, Nguyen K, Scheetz MH.
patients with methicillin-resistant 24. Neely MN, Youn G, Jones B et al. Vancomycin area under the curve and
Staphylococcus aureus bacteremia Are vancomycin trough concentra- acute kidney injury: a meta-analysis.
and osteomyelitis. Clin Ther. 2013; tions adequate for optimal dosing? Clin Infect Dis. 2019; 69(11):1881-1887.
35(6):772-779. Antimicrob Agents Chemother. 2014; 36. Avedissian SN, Pais GM, O’Donnell JN
15. Jung Y, Song KH, Cho Je et al. Area 58(1):309-316. et al. Twenty-four hour pharmaco-
under the concentration-time curve 25. Lesar TS, Rotschafer JC, Strand LM, kinetic relationships for intravenous
to minimum inhibitory concentra- Zolem LD, Zaske DE. Gentamicin vancomycin and novel urinary bio-
tion ratio as a predictor of vancomycin dosing errors with four commonly markers of acute kidney injury in a rat
treatment outcome in methicillin- used nomograms. JAMA. 1982; model. J Antimicrob Chemother. 2019;
resistant Staphylococcus aureus bacter- 248(10):1190-1193. 74(8):2326-2334.
aemia. Int J Antimicrob Agents. 2014; 26. Patel N, Pai MP, Rodvold KA, 37. Rhodes NJ, Prozialeck WC, Lodise TP
43(2):179-183. Lomaestro B, Drusano GL, Lodise TP. et al. Evaluation of vancomycin expos-
16. Lodise TP, Drusano GL, Zasowski E Vancomycin: we can’t get there ures associated with elevations in novel
et al. Vancomycin exposure in pa- from here. Clin Infect Dis. 2011; urinary biomarkers of acute kidney
tients with methicillin-resistant 52(8):969-974. injury in vancomycin-treated rats.
Staphylococcus aureus bloodstream 27. Mehta RL, Kellum JA, Shah SV et al.; Antimicrob Agents Chemother. 2016;
infections: how much is enough? Clin Acute Kidney Injury Network. Acute 60(10):5742-5751.
Infect Dis. 2014; 59(5):666-675. Kidney Injury Network: report of an 38. Lodise TP, Graves J, Evans A et al.
17. Casapao AM, Lodise TP, Davis SL et al. initiative to improve outcomes in Relationship between vancomycin
Association between vancomycin day 1 acute kidney injury. Crit Care. 2007; MIC and failure among patients with
exposure profile and outcomes among 11(2):R31. methicillin-resistant Staphylococcus

858    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

aureus bacteremia treated with vanco- IMPACT-HAP Database. Clin Ther. 2012; vancomycin MIC creep in methi-
mycin. Antimicrob Agents Chemother. 34(1):149-157. cillin resistant Staphylococcus aureus
2008; 52(9):3315-3320. 48. Vance-Bryan K, Rotschafer JC, infections—a systematic review and
39. Tong SYC, Lye DC, Yahav D, et al; for Gilliland SS, Rodvold KA, meta-analysis. Clin Microbiol Infect.
Australasian Society for Infectious Fitzgerald CM, Guay DR. A compar- 2017; 24(2):97-104.
Diseases Clinical Research Network. ative assessment of vancomycin- 59. Diaz R, Ramalheira E, Afreixo V, Gago B.
Effect of vancomycin or daptomycin associated nephrotoxicity in the Evaluation of vancomycin MIC creep in
with vs without an antistaphylococcal young versus the elderly hospitalized Staphylococcus aureus infections. J Glob
β-lactam on mortality, bacteremia, patient. J Antimicrob Chemother. 1994; Antimicrob Resist. 2017; 10:281-284.
relapse, or treatment failure in pa- 33(4):811-821. 60. Sader HS, Farrell DJ, Flamm RK,
tients with MRSA bacteremia: a 49. Jeffres MN, Isakow W, Doherty JA, Jones RN. Activity of ceftaroline and
randomized clinical trial. JAMA. 2020; Micek ST, Kollef MH. A retrospec- comparator agents tested against
323(6):527-537. tive analysis of possible renal tox- Staphylococcus aureus from patients
40. Moenster RP, Linneman TW, icity associated with vancomycin in with bloodstream infections in US
Finnegan PM, Hand S, Thomas Z, patients with health care-associated medical centres (2009-13). J Antimicrob

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


McDonald JR. Acute renal failure methicillin-resistant Staphylococcus Chemother. 2015; 70(7):2053-2056.
associated with vancomycin and aureus pneumonia. Clin Ther. 2007; 61. Pfaller MA, Sader HS, Flamm RK,
beta-lactams for the treatment of 29(6):1107-1115. Castanheira M, Smart JI, Mendes RE. In
osteomyelitis in diabetics: piperacillin- 50. Fullmer A, McCue D, Feng C. vitro activity of telavancin against clini-
tazobactam as compared with Retrospective review of vancomycin- cally important gram-positive pathogens
cefepime. Clin Microbiol Infect. 2014; induced nephrotoxicity in patients with from 69 U.S. medical centers (2015): po-
20(6):O384-O389. leukemia. J Oncol Pharm Pract. 2014; tency analysis by U.S. Census Divisions.
41. Gomes DM, Smotherman C, Birch A 20(6):403-408. Microb Drug Resist. 2017; 23(6):718-726.
et al. Comparison of acute kidney 51. Chung J, Oh JM, Cho EM et al. Optimal 62. Duncan LR, Sader HS, Smart JI,
injury during treatment with van- dose of vancomycin for treating Flamm RK, Mendes RE. Telavancin
comycin in combination with methicillin-resistant Staphylococcus activity in vitro tested against a world-
piperacillin-tazobactam or cefepime. aureus pneumonia in critically ill pa- wide collection of gram-positive clinical
Pharmacotherapy. 2014; 34(7):662-669. tients. Anaesth Intensive Care. 2011; isolates (2014). J Glob Antimicrob Resist.
42. Burgess LD, Drew RH. Comparison 39(6):1030-1037. 2017; 10:271-276.
of the incidence of vancomycin- 52. Hermsen ED, Hanson M, 63. Mouton JW, Meletiadis J, Voss A,
induced nephrotoxicity in hospi- Sankaranarayanan J, Stoner JA, Turnidge J. Variation of MIC measure-
talized patients with and without Florescu MC, Rupp ME. Clinical outcomes ments: the contribution of strain and
concomitant piperacillin-tazobactam. and nephrotoxicity associated with vanco- laboratory variability to measurement
Pharmacotherapy. 2014; 34(7):670-676. mycin trough concentrations during treat- precision. J Antimicrob Chemother.
43. Navalkele B, Pogue JM, Karino S et al. ment of deep-seated infections. Expert 2018; 73(9):2374-2379.
Risk of acute kidney injury in patients Opin Drug Saf. 2010; 9(1):9-14. 64. Heil EL, Claeys KC, Mynatt RP et al.
on concomitant vancomycin and 53. Hidayat LK, Hsu DI, Quist R, Making the change to area under the
piperacillin-tazobactam compared to Shriner KA, Wong-Beringer A. curve-based vancomycin dosing.
those on vancomycin and cefepime. High-dose vancomycin therapy for Am J Health-Syst Pharm. 2018;
Clin Infect Dis. 2017; 64(2):116-123. methicillin-resistant Staphylococcus 75(24):1986-1995.
44. Karino S, Kaye KS, Navalkele B aureus infections: efficacy and 65. Jelliffe RW, Schumitzky A. Modeling,
et al. Epidemiology of acute kidney toxicity. Arch Intern Med. 2006; adaptive control, and optimal drug
injury among patients receiving 166(19):2138-2144. therapy. Med Prog Technol. 1990;
concomitant vancomycin and 54. Kralovicová K, Spanik S, Halko J et al. 16(1-2):95-110.
piperacillin-tazobactam: opportun- Do vancomycin serum levels predict 66. Bayard DS, Jelliffe RW. A Bayesian
ities for antimicrobial stewardship. failures of vancomycin therapy or approach to tracking patients having
Antimicrob Agents Chemother. 2016; nephrotoxicity in cancer patients? J changing pharmacokinetic parameters.
60(6):3743-3750. Chemother. 1997; 9(6):420-426. J Pharmacokinet Pharmacodyn. 2004;
45. Lodise TP, Lomaestro B, Graves J, 55. Zimmermann AE, Katona BG, 31(1):75-107.
Drusano GL. Larger vancomycin doses Plaisance KI. Association of vanco- 67. Fuchs A, Csajka C, Thoma Y, Buclin T,
(at least four grams per day) are as- mycin serum concentrations with out- Widmer N. Benchmarking therapeutic
sociated with an increased incidence comes in patients with gram-positive drug monitoring software: a review
of nephrotoxicity. Antimicrob Agents bacteremia. Pharmacotherapy. 1995; of available computer tools. Clin
Chemother. 2008; 52(4):1330-1336. 15(1):85-91. Pharmacokinet. 2013; 52(1):9-22.
46. Minejima E, Choi J, Beringer P, Lou M, 56. Mohr JF, Murray BE. Point: vancomycin 68. Turner RB, Kojiro K, Shephard EA et al.
Tse E, Wong-Beringer A. Applying new is not obsolete for the treatment of in- Review and validation of Bayesian
diagnostic criteria for acute kidney fection caused by methicillin-resistant dose-optimizing software and equa-
injury to facilitate early identification of Staphylococcus aureus. Clin Infect Dis. tions for calculation of the vancomycin
nephrotoxicity in vancomycin-treated 2007; 44(12):1536-1542. area under the curve in critically ill
patients. Antimicrob Agents Chemother. 57. Pai MP, Neely M, Rodvold KA, patients. Pharmacotherapy. 2018;
2011; 55(7):3278-3283. Lodise TP. Innovative approaches to op- 38(12):1174-1183.
47. Cano EL, Haque NZ, Welch VL, et al. timizing the delivery of vancomycin in 69. Al-Sulaiti FK, Nader AM, Saad MO et al.
Incidence of nephrotoxicity and as- individual patients. Adv Drug Deliv Rev. Clinical and pharmacokinetic outcomes
sociation with vancomycin use in 2014; 77:50-57. of peak-trough-based versus trough-based
intensive care unit patients with pneu- 58. Diaz R, Afreixo V, Ramalheira E, vancomycin therapeutic drug monitoring
monia: retrospective analysis of the Rodrigues C, Gago B. Evaluation of approaches: a pragmatic randomized

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  859


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

controlled trial. Eur J Drug Metab continuous infusion for treatment 92. Raverdy V, Ampe E, Hecq JD,
Pharmacokinet. 2019; 44(5):639-652. of ventilator-associated pneumonia Tulkens PM. Stability and compat-
70. Begg EJ, Barclay ML, Duffull SB. A in trauma patients. Am Surg. 2013; ibility of vancomycin for admin-
suggested approach to once-daily 79(11):1185-1190. istration by continuous infusion.
aminoglycoside dosing. Br J Clin 82. Tafelski S, Nachtigall I, Troeger U, et al. J Antimicrob Chemother. 2013;
Pharmacol. 1995; 39(6):605-609. Observational clinical study on the 68(5):1179-1182.
71. Bauer LA. Applied Clinical effects of different dosing regimens on 93. Wade J, Cooper M, Ragan R. Simulated
Pharmacokinetics. 2nd ed. New York, vancomycin target levels in critically ill Y-site compatibility of vancomycin
NY: McGraw-Hill Medical; 2008. patients: continuous versus intermittent and piperacillin-tazobactam. Hosp
72. Beringer P. Winter’s Basic Clinical application. J Infect Public Health. 2015; Pharm. 2015; 50(5):376-379.
Pharmacokinetics. 6th ed. Philadelphia, 8(4):355-363. 94. de Waele JJ, Danneels I, Depuydt P,
PA: Wolters Kluwer Health; 2018. 83. Hutschala D, Kinstner C, Skhirdladze K, Decruyenaere J, Bourgeois M, Hoste E.
73. Marsot A, Boulamery A, Bruguerolle B, Thalhammer F, Muller M, Tschernko E. Factors associated with inadequate
Simon N. Vancomycin: a review of pop- Influence of vancomycin on renal func- early vancomycin levels in critically
ulation pharmacokinetic analyses. Clin tion in critically ill patients after cardiac ill patients treated with continuous

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


Pharmacokinet. 2012; 51(1):1-13. surgery: continuous versus intermit- infusion. Int J Antimicrob Agents. 2013;
74. Lodise TP, McKinnon PS, Swiderski L, tent infusion. Anesthesiology. 2009; 41(5):434-438.
Rybak MJ. Outcomes analysis of 111(2):356-365. 95. Ocampos-Martinez E, Penaccini L,
delayed antibiotic treatment for 84. Akers KS, Cota JM, Chung KK, Renz EM, Scolletta S et al. Determinants of early
hospital-acquired Staphylococcus au- Mende K, Murray CK. Serum vanco- inadequate vancomycin concentra-
reus bacteremia. Clin Infect Dis. 2003; mycin levels resulting from continuous tions during continuous infusion
36(11):1418-1423. or intermittent infusion in critically ill in septic patients. Int J Antimicrob
75. Diaz R, Afreixo V, Ramalheira E, burn patients with or without contin- Agents. 2012; 39(4):332-337.
Rodrigues C, Gago B. Evaluation of uous renal replacement therapy. J Burn 96. Mohammedi I, Descloux E, Argaud L,
vancomycin MIC creep in methicillin- Care Res. 2012; 33(6):e254-e262. Le Scanff J, Robert D. Loading dose of
resistant Staphylococcus aureus 85. Saugel B, Nowack MC, Hapfelmeier A et al. vancomycin in critically ill patients:
infections-a systematic review and Continuous intravenous administration of 15 mg/kg is a better choice than
meta-analysis. Clin Microbiol Infect. vancomycin in medical intensive care unit 500 mg. Int J Antimicrob Agents. 2006;
2018; 24(2):97-104. patients. J Crit Care. 2013; 28(1):9-13. 27(3):259-262.
76. Diekema DJ, Pfaller MA, Shortridge D, 86. Hanrahan TP, Harlow G, Hutchinson J 97. Li J, Udy AA, Kirkpatrick CM, Lipman J,
Zervos M, Jones RN. Twenty-year et al. Vancomycin-associated Roberts JA. Improving vancomycin
trends in antimicrobial susceptibilities nephrotoxicity in the critically ill: a prescription in critical illness through
among Staphylococcus aureus from the retrospective multivariate regres- a drug use evaluation process: a
SENTRY antimicrobial surveillance sion analysis*. Crit Care Med. 2014; weight-based dosing intervention
program. Open Forum Infect Dis. 2019; 42(12):2527-2536. study. Int J Antimicrob Agents. 2012;
6(suppl 1):S47-S53. 87. Vuagnat A, Stern R, Lotthe A et al. 39(1):69-72.
77. Kruzel MC, Lewis CT, Welsh KJ et al. High dose vancomycin for osteomy- 98. Pea F, Furlanut M, Negri C et al.
Determination of vancomycin and elitis: continuous vs. intermittent Prospectively validated dosing nomo-
daptomycin MICs by different testing infusion. J Clin Pharm Ther. 2004; grams for maximizing the pharmaco-
methods for methicillin-resistant 29(4):351-357. dynamics of vancomycin administered
Staphylococcus aureus. J Clin Microbiol. 88. Ingram PR, Lye DC, Fisher DA, Goh WP, by continuous infusion in critically
2011; 49(6):2272-2273. Tam VH. Nephrotoxicity of contin- ill patients. Antimicrob Agents
78. Hsu DI, Hidayat LK, Quist R et al. uous versus intermittent infusion of Chemother. 2009; 53(5):1863-1867.
Comparison of method-specific vanco- vancomycin in outpatient parenteral 99. Roberts JA, Taccone FS, Udy AA,
mycin minimum inhibitory concentra- antimicrobial therapy. Int J Antimicrob Vincent JL, Jacobs F, Lipman J.
tion values and their predictability for Agents. 2009; 34(6):570-574. Vancomycin dosing in critically ill
treatment outcome of meticillin-resistant 89. Verrall AJ, Llorin R, Tam VH et al. patients: robust methods for im-
Staphylococcus aureus (MRSA) infections. Efficacy of continuous infusion of van- proved continuous-infusion regimens.
Int J Antimicrob Agents. 2008; 32(5):378-385. comycin for the outpatient treatment of Antimicrob Agents Chemother. 2011;
79. Rybak MJ, Vidaillac C, Sader HS et al. methicillin-resistant Staphylococcus au- 55(6):2704-2709.
Evaluation of vancomycin suscepti- reus infections. J Antimicrob Chemother. 100. Truong J, Levkovich BJ, Padiglione AA.
bility testing for methicillin-resistant 2012; 67(12):2970-2973. Simple approach to improving van-
Staphylococcus aureus: comparison 90. Ingram PR, Lye DC, Tambyah PA, comycin dosing in intensive care: a
of etest and three automated testing Goh WP, Tam VH, Fisher DA. Risk standardised loading dose results in
methods. J Clin Microbiol. 2013; factors for nephrotoxicity associated earlier therapeutic levels. Intern Med J.
51(7):2077-2081. with continuous vancomycin infusion 2012; 42(1):23-29.
80. Wysocki M, Delatour F, Faurisson F in outpatient parenteral antibiotic 101. Wang JT, Fang CT, Chen YC, Chang SC.
et al. Continuous versus intermit- therapy. J Antimicrob Chemother. 2008; Necessity of a loading dose when
tent infusion of vancomycin in severe 62(1):168-171. using vancomycin in critically ill pa-
staphylococcal infections: prospec- 91. Dubée V, Zeller V, Lhotellier L et al. tients. J Antimicrob Chemother. 2001;
tive multicenter randomized study. Continuous high-dose vancomycin 47(2):246.
Antimicrob Agents Chemother. 2001; combination therapy for methicillin- 102. Barth RH, DeVincenzo N. Use
45(9):2460-2467. resistant staphylococcal prosthetic of vancomycin in high-flux he-
81. Schmelzer TM, Christmas AB, hip infection: a prospective cohort modialysis: experience with 130
Norton HJ, Heniford BT, Sing RF. study. Clin Microbiol Infect. 2013; courses of therapy. Kidney Int. 1996;
Vancomycin intermittent dosing versus 19(2):E98-E105. 50(3):929-936.

860    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

103. Brown M, Polisetty R, Gracely EJ, among adults and youth: United exposure in obese patients with
Cuhaci B, Schlecht HP. Weight-based States, 2015–2016. NCHS Data Brief. limited pharmacokinetic sampling.
loading of vancomycin in patients on 2017; (288):1-8. Antimicrob Agents Chemother. 2017;
hemodialysis. Clin Infect Dis. 2011; 116. Choi YC, Saw S, Soliman D et al. 61(5):e02478-16.
53(2):164-166. Intravenous vancomycin associated 128. Fu CF, Huang JD, Wang JT, Lin SW,
104. Vandecasteele SJ, de Bacquer D, with the development of nephro- Wu CC. The ratio of pre-dialysis van-
de Vriese AS. Implementation of a dose toxicity in patients with class III comycin trough serum concentration
calculator for vancomycin to achieve obesity. Ann Pharmacother. 2017; to minimum inhibitory concentra-
target trough levels of 15-20 microg/mL 51(11):937-944. tion is associated with treatment
in persons undergoing hemodialysis. 117. Vance-Bryan K, Guay DR, Gilliland SS, outcomes in methicillin-resistant
Clin Infect Dis. 2011; 53(2):124-129. Rodvold KA, Rotschafer JC. Effect of Staphylococcus aureus bacteremia.
105. Covajes C, Scolletta S, Penaccini L obesity on vancomycin pharmaco- PLoS One. 2018; 13(3):e0193585.
et al. Continuous infusion of vanco- kinetic parameters as determined by 129. Zelenitsky SA, Ariano RE, McCrae ML,
mycin in septic patients receiving using a Bayesian forecasting tech- Vercaigne LM. Initial vancomycin
continuous renal replacement nique. Antimicrob Agents Chemother. dosing protocol to achieve therapeutic

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


therapy. Int J Antimicrob Agents. 2013; 1993; 37(3):436-440. serum concentrations in patients
41(3):261-266. 118. Ducharme MP, Slaughter RL, undergoing hemodialysis. Clin Infect
106. Decker BS, Kays MB, Chambers M, Edwards DJ. Vancomycin pharma- Dis. 2012; 55(4):527-533.
Kraus MA, Moe SM, Sowinski KM. cokinetics in a patient population: 130. Lewis SJ, Mueller BA. Contemporary
Vancomycin pharmacokinetics and effect of age, gender, and body vancomycin dosing in chronic hemo-
pharmacodynamics during short daily weight. Ther Drug Monit. 1994; dialysis patients does not meet AUC
hemodialysis. Clin J Am Soc Nephrol. 16(5):513-518. targets: development of a new dosing
2010; 5(11):1981-1987. 119. Bauer LA, Black DJ, Lill JS. model using Monte Carlo simulation.
107. Alfandari S, Levent T, Descamps D Vancomycin dosing in morbidly obese Presentation at: Infectious Diseases
et al. Evaluation of glycopeptide use in patients. Eur J Clin Pharmacol. 1998; Week; 2016; New Orleans LA.
nine French hospitals. Med Mal Infect. 54(8):621-625. 131. Scott MK, Mueller BA, Clark WR.
2010; 40(4):232-237. 120. Adane ED, Herald M, Koura F. Vancomycin mass transfer character-
108. Hochart C, Berthon C, Corm S et al. Pharmacokinetics of vancomycin in istics of high-flux cellulosic dialysers.
Vancomycin serum concentration extremely obese patients with sus- Nephrol Dial Transplant. 1997;
during febrile neutropenia in patients pected or confirmed Staphylococcus 12(12):2647-2653.
with acute myeloid leukemia. Med Mal aureus infections. Pharmacotherapy. 132. Clark WR, Mueller BA, Kraus MA,
Infect. 2011; 41(12):652-656. 2015; 35(2):127-139. Macias WL. Extracorporeal therapy
109. Lemaire X, Loiez C, Valette M et al. 121. Hong J, Krop LC, Johns T, Pai MP. requirements for patients with acute
Comparison of vancomycin and Individualized vancomycin dosing in renal failure. J Am Soc Nephrol. 1997;
teicoplanin trough serum levels in obese patients: a two-sample meas- 8(5):804-812.
patients with infected orthopedic urement approach improves target 133. Mueller BA, Smoyer WE. Challenges
devices: new data for old therapies. J attainment. Pharmacotherapy. 2015; in developing evidence-based drug
Infect Chemother. 2011; 17(3):370-374. 35(5):455-463. dosing guidelines for adults and
110. Grace E. Altered vancomycin phar- 122. Pai MP. Drug dosing based on weight children receiving renal replacement
macokinetics in obese and morbidly and body surface area: mathemat- therapy. Clin Pharmacol Ther. 2009;
obese patients: what we have learned ical assumptions and limitations in 86(5):479-482.
over the past 30 years. J Antimicrob obese adults. Pharmacotherapy. 2012; 134. Vancomycin hydrochloride for injec-
Chemother. 2012; 67(6):1305-1310. 32(9):856-868. tion [package insert]. Lake Forest, IL:
111. Blouin RA, Bauer LA, Miller DD, 123. Cockcroft DW, Gault MH. Prediction Hospira, Inc.; March 2017.
Record KE, Griffen WO Jr. Vancomycin of creatinine clearance from serum 135. Lanese DM, Alfrey PS, Molitoris BA.
pharmacokinetics in normal creatinine. Nephron. 1976; 16(1):31-41. Markedly increased clearance of van-
and morbidly obese subjects. 124. Matzke GR, Aronoff GR, Atkinson AJ comycin during hemodialysis using
Antimicrob Agents Chemother. 1982; Jr et al. Drug dosing consideration polysulfone dialyzers. Kidney Int. 1989;
21(4):575-580. in patients with acute and chronic 35(6):1409-1412.
112. Crass RL, Dunn R, Hong J, Krop LC, kidney disease-a clinical update from 136. Quale JM, O’Halloran JJ,
Pai MP. Dosing vancomycin in the Kidney Disease Improving Global DeVincenzo N, Barth RH.
super obese: less is more. J Antimicrob Outcomes (KDIGO). Kidney Int. 2011; Removal of vancomycin by high-
Chemother. 2018; 73(11):3081-3086. 80(11):1122-1137. flux hemodialysis membranes.
113. Clinical guidelines on the identifi- 125. Rodvold KA, Blum RA, Fischer JH Antimicrob Agents Chemother. 1992;
cation, evaluation, and treatment et al. Vancomycin pharmacokinetics 36(7):1424-1426.
of overweight and obesity in adults: in patients with various degrees of 137. Scott MK, Macias WL, Kraus MA,
executive summary. Expert Panel on renal function. Antimicrob Agents Clark WR, Carfagna MA, Mueller BA.
the Identification, Evaluation, and Chemother. 1988; 32(6):848-852. Effects of dialysis membrane on
Treatment of Overweight in Adults. 126. Pai MP, Hong J, Krop L. Peak measure- intradialytic vancomycin admin-
Am J Clin Nutr. 1998; 68(4):899-917. ment for vancomycin AUC estimation istration. Pharmacotherapy. 1997;
114. Pastor PN, Reuben C, Xia H. in obese adults improves precision 17(2):256-262.
Chartbook on Trends in the Health of and lowers bias. Antimicrob Agents 138. Mason NA, Neudeck BL, Welage LS,
Americans. Hyattsville, MD: National Chemother. 2017; 61(4):e02490-16. Patel JA, Swartz RD. Comparison of 3
Center for Health Statistics; 2002. 127. Carreno JJ, Lomaestro B, Tietjan J, vancomycin dosage regimens during
115. Hales CM, Carroll MD, Fryar CD, Lodise TP. Pilot study of a Bayesian hemodialysis with cellulose triace-
Ogden CL. Prevalence of obesity approach to estimate vancomycin tate dialyzers: post-dialysis versus

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  861


ASHP REPORT GUIDELINE ON VANCOMYCIN MONITORING

intradialytic administration. Clin modeled continuous renal replace- 162. Frymoyer A, Hersh AL, Benet LZ,
Nephrol. 2003; 60(2):96-104. ment therapy. Blood Purif. 2011; Guglielmo BJ. Current recommended
139. Ariano RE, Fine A, Sitar DS, Rexrode S, 32(2):82-88. dosing of vancomycin for children
Zelenitsky SA. Adequacy of a van- 152. Frazee EN, Kuper PJ, Schramm GE with invasive methicillin-resistant
comycin dosing regimen in patients et al. Effect of continuous venovenous Staphylococcus aureus infections is
receiving high-flux hemodialysis. Am J hemofiltration dose on achievement of inadequate. Pediatr Infect Dis J. 2009;
Kidney Dis. 2005; 46(4):681-687. adequate vancomycin trough concen- 28(5):398-402.
140. Lucksiri A, Scott MK, Mueller BA, trations. Antimicrob Agents Chemother. 163. Frymoyer A, Guglielmo BJ, Hersh AL.
Hamburger RJ, Sowinski KM. 2012; 56(12):6181-6185. Desired vancomycin trough serum
CAHP-210 dialyzer influence on 153. Kidney Disease: Improving Global concentration for treating invasive
intra-dialytic vancomycin removal. Outcomes Acute Kidney Injury Work methicillin-resistant staphylococcal
Nephrol Dial Transplant. 2002; Group. KDIGO clinical practice guide- infections. Pediatr Infect Dis J. 2013;
17(9):1649-1654. line for acute kidney injury. Kidney Int 32(10):1077-1079.
141. Nyman HA, Agarwal A, Senekjian HO, Suppl. 2012(2):S1-S138. 164. Le J, Bradley JS, Murray W et al.
Leypoldt JK, Cheung AK. Removal of 154. Chaijamorn W, Jitsurong A, Improved vancomycin dosing in

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


vancomycin administered during dial- Wiwattanawongsa K, children using area under the curve
ysis by a high-flux dialyzer. Hemodial Wanakamanee U, Dandecha P. exposure. Pediatr Infect Dis J. 2013;
Int. 2018; 22(3):383-387. Vancomycin clearance during contin- 32(4):e155-e163.
142. Pollard TA, Lampasona V, Akkerman S uous venovenous haemofiltration in 165. Ploessl C, White C, Manasco K.
et al. Vancomycin redistribution: critically ill patients. Int J Antimicrob Correlation of a vancomycin pharma-
dosing recommendations following Agents. 2011; 38(2):152-156. cokinetic model and trough serum
high-flux hemodialysis. Kidney Int. 155. Colin PJ, Allegaert K, Thomson AH concentrations in pediatric patients.
1994; 45(1):232-237. et al. Vancomycin Pharmacokinetics Pediatr Infect Dis J. 2015; 34(10):e244
143. Crawford BS, Largen RF, Walton T, throughout life: results from a pooled -e247.
Doran JJ. Once-weekly vancomycin for population analysis and evaluation of 166. Madigan T, Sieve RM, Graner KK,
patients receiving high-flux hemodi- current dosing recommendations. Clin Banerjee R. The effect of age and
alysis. Am J Health-Syst Pharm. 2008; Pharmacokinet. 2019; 58(6):767-780. weight on vancomycin serum
65(13):1248-1253. 156. McNeil JC, Kok EY, Forbes AR et al. trough concentrations in pediatric
144. El Nekidy WS, El-Masri MM, Healthcare-associated Staphylococcus patients. Pharmacotherapy. 2013;
Umstead GS, Dehoorne-Smith M. aureus bacteremia in children: evi- 33(12):1264-1272.
Predicting maintenance doses of dence for reverse vancomycin creep 167. Demirjian A, Finkelstein Y, Nava-
vancomycin for hospitalized patients and impact of vancomycin trough Ocampo A et al. A randomized con-
undergoing hemodialysis. Can J Hosp values on outcome. Pediatr Infect Dis J. trolled trial of a vancomycin loading
Pharm. 2016; 69(5):341-347. 2016; 35(3):263-268. dose in children. Pediatr Infect Dis J.
145. Pallotta KE, Manley HJ. Vancomycin 157. McNeil JC, Kaplan SL, Vallejo JG. The 2013; 32(11):1217-1223.
use in patients requiring hemodialysis: influence of the route of antibiotic 168. Abdel Hadi O, Al Omar S, Nazer LH,
a literature review. Semin Dial. 2008; administration, methicillin suscepti- Mubarak S, Le J. Vancomycin phar-
21(1):63-70. bility, vancomycin duration and serum macokinetics and predicted dosage
146. Pai AB, Pai MP. Vancomycin dosing trough concentration on outcomes of requirements in pediatric cancer
in high flux hemodialysis: a limited- pediatric Staphylococcus aureus bacte- patients. J Oncol Pharm Pract. 2016;
sampling algorithm. Am J Health-Syst remic osteoarticular infection. Pediatr 22(3):448-453.
Pharm. 2004; 61(17):1812-1816. Infect Dis J. 2017; 36(6):572-577. 169. Le J, Vaida F, Nguyen E et al.
147. Maxson R, Pate J, Starr J. Evaluation of 158. Fiorito TM, Luther MK, Dennehy PH, Population-based pharmacokinetic
weight-based vancomycin dosing for LaPlante KL, Matson KL. modeling of vancomycin in children
hospitalized hemodialysis patients. Nephrotoxicity with vancomycin in the with renal insufficiency. J Pharmacol
Ren Fail. 2016; 38(10):1677-1682. pediatric population: a systematic re- Clin Toxicol. 2014; 2(1):1017-1026.
148. Kielstein JT, Czock D, Schöpke T et al. view and meta-analysis. Pediatr Infect 170. Avedissian SN, Bradley E, Zhang D
Pharmacokinetics and total elimina- Dis J. 2018; 37(7):654-661. et al. Augmented renal clearance using
tion of meropenem and vancomycin 159. Hahn A, Frenck RW Jr, Allen-Staat M, population-based pharmacokinetic
in intensive care unit patients under- Zou Y, Vinks AA. Evaluation of target modeling in critically ill pediatric
going extended daily dialysis. Crit Care attainment of vancomycin area under patients. Pediatr Crit Care Med. 2017;
Med. 2006; 34(1):51-56. the curve in children with methicillin- 18(9):e388-e394.
149. Scoville BA, Mueller BA. Medication resistant Staphylococcus aureus 171. Zhang H, Wang Y, Gao P et al.
dosing in critically ill patients with bacteremia. Ther Drug Monit. 2015; Pharmacokinetic characteristics and
acute kidney injury treated with renal 37(5):619-625. clinical outcomes of vancomycin in
replacement therapy. Am J Kidney Dis. 160. Yoo RN, Kim SH, Lee J. Impact of initial young children with various degrees of
2013; 61(3):490-500. vancomycin trough concentration on renal function. J Clin Pharmacol. 2016;
150. Harris LE, Reaves AB, Krauss AG, clinical and microbiological outcomes 56(6):740-748.
Griner J, Hudson JQ. Evaluation of an- of methicillin-resistant Staphylococcus 172. McKamy S, Hernandez E, Jahng M,
tibiotic prescribing patterns in patients aureus bacteremia in children. J Korean Moriwaki T, Deveikis A, Le J. Incidence
receiving sustained low-efficiency di- Med Sci. 2017; 32(1):22-28. and risk factors influencing the de-
alysis: opportunities for pharmacists. 161. Eiland LS, English TM, Eiland EH 3rd. velopment of vancomycin nephro-
Int J Pharm Pract. 2013; 21(1):55-61. Assessment of vancomycin dosing and toxicity in children. J Pediatr. 2011;
151. Pasko DA, Churchwell MD, subsequent serum concentrations in 158(3):422-426.
Salama NN, Mueller BA. Longitudinal pediatric patients. Ann Pharmacother. 173. Knoderer CA, Gritzman AL,
hemodiafilter performance in 2011; 45(5):582-589. Nichols KR, Wilson AC. Late-occurring

862    AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020


GUIDELINE ON VANCOMYCIN MONITORING ASHP REPORT

vancomycin-associated acute kidney and obese pediatric patients. J Pediatr the concentration-time curve in neo-
injury in children receiving prolonged Pharmacol Ther. 2014; 19(3):182-188. nates. Antimicrob Agents Chemother.
therapy. Ann Pharmacother. 2015; 185. Camaione L, Elliott K, Mitchell- 2014; 58(11):6454-6461.
49(10):1113-1119. Van Steele A, Lomaestro B, Pai MP. 196. Stockmann C, Hersh AL, Roberts JK
174. Sinclair EA, Yenokyan G, McMunn A, Vancomycin dosing in children and et al. Predictive performance of a van-
Fadrowski JJ, Milstone AM, Lee CK. young adults: back to the drawing comycin population pharmacokinetic
Factors associated with acute kidney board. Pharmacotherapy. 2013; model in neonates. Infect Dis Ther.
injury in children receiving vanco- 33(12):1278-1287. 2015; 4(2):187-198.
mycin. Ann Pharmacother. 2014; 186. Natale S, Bradley J, Nguyen WH et al. 197. Cies JJ, Moore WS 2nd, Nichols K,
48(12):1555-1562. Pediatric obesity: pharmacokinetic Knoderer CA, Carella DM,
175. Matson KL, Shaffer CL, Beck GL, alterations and effects on antimicro- Chopra A. Population pharmaco-
Simonsen KA. Assessment of initial bial dosing. Pharmacotherapy. 2017; kinetics and pharmacodynamic
serum vancomycin trough concentra- 37(3):361-378. target attainment of vancomycin in
tions and their association with initial 187. Le J, Capparelli EV, Wahid U et al. neonates on extracorporeal life sup-
empirical weight-based vancomycin Bayesian estimation of vancomycin port. Pediatr Crit Care Med. 2017;

Downloaded from https://academic.oup.com/ajhp/article/77/11/835/5810200 by guest on 19 April 2022


dosing and development of neph- pharmacokinetics in obese children: 18(10):977-985.
rotoxicity in children: a multicenter matched case-control study. Clin Ther. 198. Leroux S, Jacqz-Aigrain E, Biran V
retrospective study. Pharmacotherapy. 2015; 37(6):1340-1351. et al. Clinical utility and safety of
2015; 35(3):337-343. 188. Nguyen WN, Bradley JS, Capparelli EV a model-based patient-tailored
176. Ragab AR, Al-Mazroua MK, et al. Optimal weight-based vanco- dose of vancomycin in neonates.
Al-Harony MA. Incidence and mycin dosing in obese children using Antimicrob Agents Chemother. 2016;
predisposing factors of vancomycin- Bayesian estimation. Presentation 60(4):2039-2042.
induced nephrotoxicity in children. at: Interscience Conference 199. Gwee A, Cranswick N, McMullan B
Infect Dis Ther. 2013; 2(1):37-46. on Antimicroial Agents and et al. Continuous versus inter-
177. Le J, Ngu B, Bradley JS et al. Chemotherapy; September 21, 2015; mittent vancomycin infusions in
Vancomycin monitoring in children San Diego, CA. infants: a randomized controlled
using Bayesian estimation. Ther Drug 189. Rhodin MM, Anderson BJ, Peters AM trial. Pediatrics. 2019; 143(2),
Monit. 2014; 36(4):510-518. et al. Human renal function matu- pii:e20182179.
178. Stockmann C, Olson J, Rashid J ration: a quantitative description 200. Lestner JM, Hill LF, Heath PT,
et al. An evaluation of vancomycin using weight and postmenstrual age. Sharland M. Vancomycin toxicity
area under the curve estimation Pediatr Nephrol. 2009; 24(1):67-76. in neonates: a review of the evi-
methods for children treated for 190. Mehrotra N, Tang L, Phelps SJ, dence. Curr Opin Infect Dis. 2016;
acute pulmonary exacerbations of Meibohm B. Evaluation of vanco- 29(3):237-247.
cystic fibrosis due to methicillin- mycin dosing regimens in preterm 201. Bhargava V, Malloy M, Fonseca R.
resistant Staphylococcus aureus. J Clin and term neonates using Monte Carlo The association between vancomycin
Pharmacol. 2019; 59(2):198-205. simulations. Pharmacotherapy. 2012; trough concentrations and acute
179. Kendrick JG, Carr RR, Ensom MH. 32(5):408-419. kidney injury in the neonatal inten-
Pharmacokinetics and drug dosing in 191. Marqués-Miñana MR, Saadeddin A, sive care unit. BMC Pediatr. 2017;
obese children. J Pediatr Pharmacol Peris JE. Population pharmacokinetic 17(1):50.
Ther. 2010; 15(2):94-109. analysis of vancomycin in neonates. 202. Constance JE, Balch AH, Stockmann C
180. Moffett BS, Kim S, Edwards MS. A new proposal of initial dosage et al. A propensity-matched cohort
Vancomycin dosing in obese pediatric guideline. Br J Clin Pharmacol. 2010; study of vancomycin-associated
patients. Clin Pediatr (Phila). 2011; 70(5):713-720. nephrotoxicity in neonates. Arch
50(5):442-446. 192. Irikura M, Fujiyama A, Saita F et al. Dis Child Fetal Neonatal Ed. 2016;
181. Heble DE Jr, McPherson C, Nelson MP, Evaluation of the vancomycin dosage 101(3):F236-F243.
Hunstad DA. Vancomycin trough regimen based on serum creatinine 203. Phillips CJ, Wisdom AJ,
concentrations in overweight or obese used in the neonatal intensive care McKinnon RA, Woodman RJ,
pediatric patients. Pharmacotherapy. unit. Pediatr Int. 2011; 53(6):1038-1044. Gordon DL. Interventions targeting
2013; 33(12):1273-1277. 193. Capparelli EV, Lane JR, Romanowski GL the prescribing and monitoring of
182. Miller M, Miller JL, Hagemann TM, et al. The influences of renal function vancomycin for hospitalized patients:
Harrison D, Chavez-Bueno S, and maturation on vancomycin elimi- a systematic review with meta-
Johnson PN. Vancomycin dosage nation in newborns and infants. J Clin analysis. Infect Drug Resist. 2018;
in overweight and obese children. Pharmacol. 2001; 41(9):927-934. 11:2081-2094.
Am J Health-Syst Pharm. 2011; 194. Zhao W, Lopez E, Biran V, 204. Phillips CJ, McKinnon RA,
68(21):2062-2068. Durrmeyer X, Fakhoury M, Jacqz- Woodman RJ, Gordon DL. Sustained
183. Nassar L, Hadad S, Gefen A et al. Aigrain E. Vancomycin continuous improvement in vancomycin dosing
Prospective evaluation of the dosing infusion in neonates: dosing optimisa- and monitoring post-implementation
regimen of vancomycin in children of tion and therapeutic drug monitoring. of guidelines: results of a three-year
different weight categories. Curr Drug Arch Dis Child. 2013; 98(6):449-453. follow-up after a multifaceted inter-
Saf. 2012; 7(5):375-381. 195. Frymoyer A, Hersh AL, El-Komy MH vention in an Australian teaching
184. Eiland LS, Sonawane KB. Vancomycin et al. Association between vancomycin hospital. J Infect Chemother. 2018;
dosing in healthy-weight, overweight, trough concentration and area under 24(2):103-109.

AM J HEALTH-SYST PHARM | VOLUME 77 | NUMBER 11 | JUNE 1, 2020  863

You might also like