Risk of Acute Kidney Injury in Patients on Concomitant Vancomycin and Piperacillin–Tazobactam Compared to Those on Vancomycin and Cefepime


Abstract

Background.

Recent evidence suggests that among patients receiving vancomycin, receipt of concomitant piperacillin–tazobactam increases the risk of nephrotoxicity. Well-controlled, adequately powered studies comparing rates of acute kidney injury (AKI) among patients receiving vancomycin + piperacillin–tazobactam (VPT) compared to similar patients receiving vancomycin + cefepime (VC) are lacking. In this study we compared the incidence of AKI among patients receiving combination therapy with VPT to a matched group receiving VC.

Methods.

A retrospective, matched, cohort study was performed. Patients were eligible if they received combination therapy for ≥48 hours. Patients were excluded if their baseline serum creatinine was >1.2mg/dL or they were receiving renal replacement therapy. Patients receiving VC were matched to patients receiving VPT based on severity of illness, intensive care unit status, duration of combination therapy, vancomycin dose, and number of concomitant nephrotoxins. The primary outcome was the incidence of AKI. Multivariate modeling was performed using Cox proportional hazards.

Results.

A total of 558 patients were included. AKI rates were significantly higher in the VPT group than the VC group (81/279 [29%] vs 31/279 [11%]). In multivariate analysis, therapy with VPT was an independent predictor for AKI (hazard ratio = 4.27; 95% confidence interval, 2.73–6.68). Among patients who developed AKI, the median onset was more rapid in the VPT group compared to the VC group (3 vs 5 days P =< .0001).

Conclusion.

The VPT combination was associated with both an increased AKI risk and a more rapid onset of AKI compared to the VC combination.

vancomycin, piperacillin–tazobactam, cefepime, acute kidney injury, nephrotoxicity.

Topic:

Issue Section:

Major Article

Empiric antimicrobial therapy for the treatment of healthcare-associated infections frequently includes coverage for both methicillin-resistant Staphylococcus aureus and Pseudomonas aeruginosa. Common regimens include vancomycin in combination with an antipseudomonal b-lactam [1]. Piperacillin–tazobactam and cefepime are among the most common agents used for empiric antipseudomonal coverage.

A hospital’s selection of piperacillin–tazobactam vs cefepime as the “workhorse” antipseudomonal antibiotic has traditionally been based on institutional susceptibility trends, acquisition costs, and other formulary considerations. Concerns regarding nephrotoxicity have become increasingly prominent. While vancomycin has long been associated with acute kidney injury (AKI), recent evidence suggests that patients receiving combination therapy with piperacillin–tazobactam have a higher incidence of AKI compared to patients receiving vancomycin monotherapy [2] or those receiving combination therapy with vancomycin and cefepime (VC) [3].

However, the finding of increased toxicity in patients receiving vancomycin and piperacillin–tazobactam (VPT) combination therapy compared to VC has not been universal. A recent analysis showed no difference in AKI rates among intensive care unit (ICU) patients receiving either combination [4]. Prior studies have been limited by relatively small sample sizes, notable diversity in the patients receiving the different combination therapy regimens, and suboptimal study design.

In light of the conflicting results and methodological limitations of prior studies as well as the importance of clearly understanding whether or not combination therapy with VPT is associated with an increased AKI risk, this retrospective, matched, cohort study was designed to definitively address the following questions: is combination therapy with VPT associated with greater AKI risk compared to VC? If so, how much greater is the risk?

METHODS

Study Settings and Design

This was a retrospective, matched, cohort study that compared the incidence of AKI among patients on concomitant VC and those receiving VPT. The study was conducted at the Detroit Medical Center (DMC), a tertiary care health system in metropolitan Detroit, Michigan, comprised of 5 acute care hospitals with more than 2000 inpatient beds. The institutional review boards at the DMC and Wayne State University approved the study prior to initiation.

Study Population

The study population consisted of patients aged ≥18 years admitted to the DMC between 1 December 2011 and 31 December 2013. Patients included in the study received combination therapy with VC or VPT for ≥48 hours and had the 2 antibiotics initiated within 24 hours of one another. For patients who received combination therapy multiple times during hospitalization, only the initial regimen was included. Patients were excluded if the baseline serum creatinine was >1.2 mg/dL or they required renal replacement therapy at the time of initiation of combination therapy.

Patients were divided into 2 groups based on the combination regimen received. The patients in the VC group were matched to the VPT group on 5 variables associated with the development of AKI in a 1:1 ratio. The matching was performed based on severity of sepsis at the time that the combination antibiotics were started (dichotomized to presence or absence of severe sepsis/septic shock) [5], ICU status at onset of combination therapy, duration of combination therapy (divided into 3 categories: ≤3 days, 4–7 days, >7 days), the daily dose of vancomycin received (divided into 3 categories: < 2 grams/day, 2–4 grams/day, and >4 grams/day), and number of concomitant nephrotoxic agents received while on combination therapy.

Covariates Collected

Data abstracted from medical records included patient demographics; comorbidities, including Charlson comorbidity index [6]; severity of sepsis based on systemic inflammatory response syndrome criteria [5]; mechanical ventilation; infectious diagnosis; and receipt of concomitant nephrotoxins while receiving combination therapy. Antibiotic therapy variables collected included dose and duration of therapy. Vancomycin trough levels were also collected. Vancomycin loading dose was defined as an initial vancomycin dose that was higher than subsequent maintenance doses. The variables used for matching were extracted during the time period between 2 days prior and 2 days after initiation of combination therapy, with the highest values used for this purpose. Vasopressors, aminoglycosides, colistin, amphotericin B, angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, diuretics, and intravenous contrast were considered as nephrotoxic agents.

Vancomycin Trough Value Assessment

In order to assess the impact of vancomycin exposures on development of AKI, the median trough of vancomycin prior to AKI was calculated. For patients who did not develop AKI, median vancomycin troughs during the entire duration of combination therapy were analyzed, whereas among patients who developed AKI, only trough values obtained before the onset of AKI were included. Patients in whom trough values were not obtained during therapy and those who did not have trough values obtained prior to the development of AKI were excluded from trough analyses.

Acute Kidney Injury Definitions

Determination of AKI was based on 3 definitions: According to the RIFLE (Risk, Injury, Failure, Loss, End Stage Renal Disease) criteria [7], the Acute Kindey Injury Network (AKIN) criteria [8], and vancomycin consensus guideline definition [9]. For RIFLE criteria, the terms risk, injury, and failure were defined as follows: risk, a rise in creatinine by 1.5 times baseline or a decrease in glomerular filtration rate (GFR) by 25%; injury, a rise in creatinine of 2 times baseline or a decrease in the GFR by 50%; and failure, a rise in creatinine by 3 times baseline or a GFR decrease by 75%. AKIN criteria were categorized into 3 stages: a rise in creatinine by 1.5-fold or 0.3 mg/dL was categorized as stage 1, a 2-fold rise in creatinine was categorized as stage 2, and a rise in creatinine by 3-fold or ≥ 4 mg/dL or initiation of renal replacement therapy was categorized as stage 3. For the vancomycin consensus guidelines, AKI was defined as a rise in baseline serum creatinine by ≥50% or >0.5 mg/dL sustained over at least 2 consecutive measurements ranging from the time of initiation until 72 hours post-completion of vancomycin therapy. RIFLE-defined AKI was used for all multivariate analyses, where meeting any stage of the RIFLE criteria was considered AKI.

Statistical Analyses

All statistical analyses were performed using SAS software, version 9.3 (Cary, North Carolina). Matched bivariate analyses comparing patients receiving VC to patients receiving VPT were conducted using conditional logistic regression modeling. For bivariate unmatched analysis, Fisher exact test and χ2 test were used to analyze dichotomous variables, and Student t test and Wilcoxon rank-sum test were used for continuous variables.

To determine the impact of VPT on AKI risk in both bivariate and multivariate analyses, Cox proportional hazards methodology was used. In multivariate analysis to control for residual differences between the VPT and VC groups, all variables with a P value <.1 in the bivariate matched analysis comparing VPT and VC groups were included, along with treatment group (VPT vs VC), in a multivariate model for AKI. In this model the event of interest was development of RIFLE-defined AKI. All P values were 2 sided and a P value <.05 was considered statistically significant. Crude rates of AKI of the 2 study groups were compared using a Kaplan–Meier curve and the log-rank test.

RESULTS

Baseline Characteristics

A total of 320 patients who received VPT and 803 patients who received VC during the study period were identified. Of the 320 VPT patients, adequate VC matches were identified for 279. Thus, 279 VPT–VC pairs were included in the final study population, for a total of 558 patients. The mean age was 55.9 ± 16.6 years. Patients in both VC and VPT groups had similar baseline characteristics in terms of age, length of ICU stay, Charlson comorbidity index score, baseline creatinine, and use of concomitant nephrotoxins (Table 1). There were more females in the VC group, and more patients were white in the VPT group. Patients were more likely to have had connective tissue disease and hypertension in the VC group compared to those in the VPT group. Patients in the VPT group had a higher incidence of septic shock and skin and soft tissue infections. Combination therapy with both VPT and VC was initiated as empiric therapy in all patients. There were no differences in the number of patients receiving vancomycin loading doses, the median loading or maintenance doses of vancomycin given, or the median vancomycin trough values between the 2 groups.



Table 1.

Baseline Characteristics of Cohort Comparing Patients Receiving Vancomycin and piperacillin–Tazobactam Combination to Patients Receiving Vancomycin–Cefepime Combination

VariableVancomycin–Cefepime
n = 279 (%)
Vancomycin and Piperacillin–Tazobactam
n = 279 (%)
Odds Ratio (95% Confidence Interval)P Value
Age, ya56.5 ± 16.455.3 ± 16.8.39
Female153 (55)128 (46)0.69(0.50–0.97).034
Race<.0001
White56 (20)93 (33).0005
Black191 (68)175 (63)
Others32 (11)11 (4)1.98 (1.35–2.92)
Admission source226 (81)242 (87)1.53 (0.97–2.42).005
Home30 (11)31 (11).07
Nursing home23 (8)6 (2)
Other hospital226 (81)242 (87)
Height, cma170 ± 10171 ± 11.34
Median weight, kg74 (63.8–90)78.4 (66–95).42
Median body mass index, kg/m225 (21.4–30.3)26.5 (22.5–31.7).55
Comorbid conditions
Myocardial Infarction24 (9)13 (5)0.52 (0.25–1.04).06
Congestive heart failure36 (13)34 (12)0.94 (0.56–1.55).79
Peripheral vascular disease26 (9)30 (11)1.17 (0.67–2.04).57
Dementia32 (11)23 (8)0.69 (0.39–1.22).20
Chronic pulmonary disease79 (28)82 (29)1.05 (0.73–1.52).78
Connective tissue disease22 (8)10 (4)0.43 (0.20–0.94).03
Chronic kidney disease10 (4)5 (2)0.49 (0.16–1.45).19
Malignant solid tumor51 (18)40 (14)0.75 (0.47–1.17).20
Cerebrovascular disease42 (15)29 (10)0.65 (0.39–1.08).10
Liver disease9 (3)15 (5)1.70 (0.73–3.96).21
Diabetes mellitus62 (22)69 (24)1.15 (0.77–1.70).48
Hypertension177 (63)149 (53)0.66 (0.47–0.93).02
Median Charlson comorbidity index (IQR)1 (0–3)1 (0–3).21
Hospital and infection-related variables
Systemic inflammatory response syndrome criteriab
No sepsis44 (16)52 (19)
Sepsis166 (60)159 (57)
Severe sepsis62 (22)48 (17)
Septic shock7 (3)20 (7)
Any sepsis235 (84)227 (81)0.82 (0.53–1.27).37
Severe sepsis/septic shock69 (25)68 (24)0.98 (0.66–1.44).92
Intensive care unit stay b63 (23)58 (21)0.90 (0.60–1.35).61
Mechanical ventilation b43 (15)44 (16)1.03 (0.65–1.62).91
Median white blood cell count b
>10
11.3 (7.7–15.5)
166 (60)
10.5 (7.4–14.4)
146 (52)
0.74 (0.53–1.04).06
.08
Mean baseline creatininea0.86 ± 0.200.86 ± 0.21.64
Median length of stay before combination therapy (IQR)0 (0.0–3.0)0 (0.0–2.0).63
Infection type and diagnosis
Physician-diagnosis with positive culture86 (31)91 (33)1.08 (0.76–1.55).65
Pneumonia12 (4)14 (5)1.17 (0.53–2.59).68
Endocarditis4 (1)2 (1)0.49 (0.09–2.73).42
Intraabdominal infection5 (2)9 (3)1.82 (0.60–5.51).28
Skin/soft tissue infection21 (8)37 (13)1.88 (1.07–3.29).02
Bone/joint infection19 (7)16 (6)0.83 (0.42–1.65).60
Urinary tract infection11 (4)8 (3)0.72 (0.28–1.82).48
Bacteremia22 (8)25 (9)1.15 (0.63–2.09).65
Catheter-associated bloodstream infection6 (2)2 (1)0.33 (0.06–1.64).17
Other/unknown4 (1)3 (1)0.75 (0.16–3.37).70
Invasive infectionc35 (13)32 (12)0.90 (0.54–1.50).69
Polymicrobial infection36 (13)40 (14)1.13 (0.69–1.83).62
Pathogens
Gram-positive bacteria50 (18)60 (22)1.25 (0.83–1.90).28
Methicillin-resistant Staphylococcus aureus23 (8)16 (6)0.68 (0.35–1.31).25
Methicillin-susceptible Staphylococcus aureus7 (3)14 (5)2.05 (0.81–5.15).13
Gram-negative bacteria51 (18)43 (15)0.81 (0.52–1.27).37
Pseudomonas5 (2)13 (5)2.67 (0.94–7.59).06
Enterobacteriaceae48 (17)31 (11)0.60 (0.37–0.97).04
Concomitant nephrotoxins
Median number of nephrotoxins (IQR)1 (0–2)1 (0–2)0.98 (0.82–1.17).86
Vasopressors9 (3)16 (6 )1.82 (0.79–4.20).16
Aminoglycoside10 (4)16 (6)1.64 (0.73–3.67).23
Colistin7 (3)2 (1)0.28 (0.06–1.36).12
Angiotensin converting enzyme inhibitors/ angiotensin II receptor blockers76 (27)70 (25)0.89 (0.61–1.30).56
Diuretics78 (28)75 (27)0.95 (0.65–1.37).78
Intravenous contrast76 (27)74 (27)0.96 (0.66–1.40).85
Vancomycin dosing and monitoring
Loading dose given237 (85)233 (84)0.89 (0.57–1.42).64
Loading dose, mga1544.8 ± 762.3
(n = 237)
1610.2 ± 827.6
(n = 233)
.12
Vancomycin dose a2818 ± 12022968 ± 1320.21
Median trough before AKId (IQR)17.7 (13.4–20.9)17.3 (12.6–21.6).98
MT before AKI >15d160 (68)
n = 236
146 (65)
n = 225
.55
.55
MT before AKI >20d77 (33)
n = 236
80 (27)
n = 225

Statistically significant values are shown in bold.

Abbreviations: AKI, acute kidney injury; IQR, interquartile range; MT, median trough.

aData are presented as mean ± standard deviation. The vancomycin maintenance dose listed is the first maintenance vancomycin dose, as subsequent doses were based on serum concentrations.

b Variables such as systemic inflammatory response syndrome criteria, intensive care unit stay, mechanical ventilation, and wild blood cell count were assessed during a window period of 2 days prior to and 2 days after initiation of combination therapy.

c Invasive infections were defined as presence of pneumonia or endocarditis or bone/joint infection.

d Data based on median troughs before AKI in patients with AKI and includes median troughs for entire duration of therapy in patients without AKI.

Open in new tab

Comparative Rates of AKI in VC and VPT Patients

The rate of AKI was higher among patients receiving VPT compared to those receiving VC combination therapy. Based on RIFLE criteria, 81 patients in the VPT group developed AKI compared to 31 patients in the VC group (29.0% vs 11.1%; hazard ratio [HR] = 4.0; 95% confidence interval [CI], 2.6–6.2; P < .0001). Rates of AKI were also higher per AKIN criteria (32% in the VPT vs 14% in the VC group; HR = 3.5; 95% CI, 2.3–5.2; P < .0001) and per vancomycin consensus guidelines definition (24% in VPT vs 8.2% in VC; HR = 4.4; 95% CI, 2.7–7.3; P < .0001). In multivariate analysis, after controlling for residual differences between the VPT and VC groups (race, gender, admission from home, comorbid conditions, presence of septic shock, baseline serum white blood cell count, and source of infection), VPT was independently associated with RIFLE-defined AKI (HR = 4.3; 95% CI, 2.7–6.7; P < .0001).

Characterization of AKI

Of the patients who developed RIFLE-defined AKI (n = 31 in the VC group and n = 81 in the VPT group), the onset of AKI was more rapid in patients receiving VPT. The median duration of combination therapy prior to development of AKI was 5 days (interquartile range [IQR], 3–7 days) in the VC and 3 days (IQR, 2–5 days) in the VPT group; P < .0001. Survival curves depicting time to AKI in the 2 treatment groups were constructed (Figures 1 and 2) and demonstrate the increased incidence and more rapid onset of AKI among patients in the VPT group compared to those in the VC group (P < .0001). Importantly, the Kaplan–Meier curves also show that the daily rate of AKI among at-risk patients remained consistently higher in the VPT group compared to the VC group throughout the entire first week of combination therapy.

Figure 1.

Comparison of cumulative rates of acute kidney injury in patients receiving combination therapy with vancomycin–cefepime and those receiving vancomycin and piperacillin–tazobactam. Abbreviations: AKI, acute kidney injury; VC, vancomycin–cefepime; VPT, vancomycin and piperacillin–tazobactam.

Open in new tabDownload slide

Comparison of cumulative rates of acute kidney injury in patients receiving combination therapy with vancomycin–cefepime and those receiving vancomycin and piperacillin–tazobactam. Abbreviations: AKI, acute kidney injury; VC, vancomycin–cefepime; VPT, vancomycin and piperacillin–tazobactam.

Figure 2.

Kaplan–Meier survival analysis for acute kidney injury as a function of treatment group.

Open in new tabDownload slide

Kaplan–Meier survival analysis for acute kidney injury as a function of treatment group.

Table 2.

Outcomes Associated With Receipt of Vancomycin Plus Piperacillin–Tazobactam Combination Therapy Compared to Receipt of Vancomycin Plus Cefepime

VariableVancomycin– Cefepime
n = 279 (%)
Vancomycin Plus Piperacillin– Tazobactam
n = 279 (%)
Bivariate HR (95% CI)P ValueMultivariate Adjusted HR
(95% CI)
P Value
RIFLE criteria
AKI any class31 (11.1)81 (29.0)4.00 (2.59–6.18)<.00014.27 (2.73–6.68)a<.0001
Risk12 (4.3)40 (14.3)
Injury8 (2.9)21 (7.5)
Failure11 (3.9)20 (7.2)
AKIN criteria
AKI any stage39 (13.9)89 (31.9)3.49 (2.35–5.18)<.0001
Stage 120 (7.2)48 (17.2)
Stage 28 (2.9)21 (7.5)
Stage 311(3.9)20 (7.2)
AKI per Vancomycin consensus guidelines23 (8.2)67 (24.0)4.44 (2.69–7.32)<.0001
AKI requiring hemodialysis3 (1.1)2 (0.7)0.66 (0.11–4.00).65
Median length of stay after initiation of combination therapy (interquartile range)6 [4–11]8 [5–12].01
Mortality24 (8.6)16 (5.7)0.64 (0.34–1.24).19

Statistically significant values are shown in bold.

Abbreviations: AKI, acute kidney injury; AKIN, acute kindey injury network; CI, confidence interval; HR, hazard ratio; RIFLE, risk, injury, failure, loss, end stage.

a Controlling for race, gender, admission from home, comorbid conditions, baseline serum white blood cell count >10, 000/µL, and source of infection being skin or soft tissue.

Open in new tab

Other Outcome Variables

The median length of stay after initiation of combination therapy was longer for VPT patients compared to VC patients (8 days vs 6 days; P = .01). There was no difference in mortality between the 2 groups.

Impact of Vancomycin Troughs on AKI

Although there were no differences in median vancomycin trough values or the number of patients who had troughs >15 mg/L or >20 mg/L between the VC and VPT groups, additional analyses were performed to further assess the impact of vancomycin trough on incidence of AKI (Figures 3a, 3b). Interestingly, when the trough was dichotomized, there was no association between vancomycin trough and AKI for patients in the VPT group (trough <15 mg/L or ≥15 mg/L). Additionally, there was no association when troughs were categorized into 3 ascending groups: <15, 15–20, or >20 mg/L.

Figure 3.

<img src="https://oup.silverchair-cdn.com/oup/backfile/Content_public/Journal/cid/64/2/10.1093_cid_ciw709/3/m_ciw7093a.jpeg?Expires=1708556068&Signature=Raj7ZWyZSjetAqBveqHTiYcZJusIpV5LI7s2BwRdDQRN-Zpcv511HwE~dB8lmYnvL-B7ga4mcQ6woiUDpPFccpu6N5o5AQooYG9wmoYxFKsXMU6HU0~i2nV6Oe7sFS5ays4F4udFFTGHMQNgVDkMYxMEBhOQ05GjgjY0W31FXrcdu1y0uq4JbPqtFEV4tIDfDJ~WcUl5RuntpDXwS~ONzKJHoW~WLbgkXPege0wmKGMWlNuXMwBn75fVAdt916ypjt9N1JdPmf0zomROpg8z7LzcJBOzv6D7y6CVZJzi8DQcWnkdJ6TmfZhu3Qeuj3e8xgpq~u6cQVIoXUutuWGr~g__&Key-Pair-Id=APKAIE5G5CRDK6RD3PGA&quot; alt="A, Acute kidney injury (AKI) rates as a function of vancomycin troughs. B, AKI rates in patients with median troughs

Open in new tabDownload slide

<img src="https://oup.silverchair-cdn.com/oup/backfile/Content_public/Journal/cid/64/2/10.1093_cid_ciw709/3/m_ciw7093b.jpeg?Expires=1708556068&Signature=s86LYkAAPdl6Ied5mATHhcl-Tqzptrv~RsvUtXqE-W-n7g6fB1JZpgKM6adl7OT-5ADqpE~uz-hsFdd7M6I8OWxv7F0CSxcFB4wX0PSsCwZt1EqQbJmsVUco-hZtCmN7mWTgmKIRPZUX6-aHIrjep9SdhruSqOcQM3B-FG-87SWLGl57sZnqhu9Ms0GPJFYImdTkt3qCb6J6OQqwJTkKe5SuYd0xpIXWPIrzmA8XK898QF4igfJ4REgFDdpV5pzdV-Z8idmg0veoPvJjEEvbTyO-ZKJoEr-m~4HODWGG1Q2JHs2A7fhZVbJWL5yBByfm0T4PNKv4I6KB8LZNtxa03g__&Key-Pair-Id=APKAIE5G5CRDK6RD3PGA&quot; alt="A, Acute kidney injury (AKI) rates as a function of vancomycin troughs. B, AKI rates in patients with median troughs

Open in new tabDownload slide

A, Acute kidney injury (AKI) rates as a function of vancomycin troughs. B, AKI rates in patients with median troughs <15 mg/L vs ≥ 15 mg/L.

Abbreviations: AKI, acute kidney injury; NA, not applicable; VC, vancomycin–cefepime; VPT, vancomycin and piperacillin–tazobactam.

Conversely, a direct relationship was seen between vancomycin trough and AKI among patients in the VC group. When the vancomycin troughs were dichotomized, AKI occurred in 1/76 (1%) patients with median trough values <15 mg/L vs 20/160 (13%) of patients with values ≥15 mg/L; P = 003. Additionally, when vancomycin troughs were analyzed in ascending categories, a significant association was also seen. AKI occurred in 1% of patients with troughs <15 mg/L, in 5% (4/83) of patients with troughs of 15–20 mg/L, and in 21% (16/77) of patients with median troughs >20 mg/L. AKI rates among patients in the VC group were significantly different when patients with troughs of <15 mg/L were compared to patients with troughs >20 mg/L (P = .0001) and when patients with vancomycin troughs of 15–20 mg/L were compared to patients with troughs >20 mg/L (P = .003).

DISCUSSION

Rates of AKI among patients receiving VPT were approximately 3 times greater than rates in patients receiving VC, regardless of type of AKI definition used. In multivariate modeling and controlling for residual differences between these 2 closely matched groups, receipt of VPT was associated with a greater than 4-fold increased risk of AKI. These findings are particularly robust and convincing as, unlike previous analyses comparing toxicity risk in patients on VPT and VC, this analysis was adequately powered and groups were matched on 5 widely recognized risk factors for AKI in patients receiving vancomycin.

These findings are strengthened by 3 additional important and notable findings. First, among patients who developed AKI, the onset was more rapid in VPT patients compared to VC patients (3 days vs 5 days; P < .0001.) Second, the daily rate of AKI among the at-risk population remained higher throughout the first week of therapy among VPT patients. This rapid onset and persistently increased AKI risk are both consistent with VPT being more toxic than VC.

The third finding supporting an association between VPT and increased toxicity was both interesting and unexpected. Data from this study show discordance in the impact of vancomycin troughs on toxicity in patients receiving VPT compared to those receiving VC. Among patients receiving VPT, there was no discernable impact of vancomycin trough on the incidence of AKI. Conversely, a distinct trough–toxicity association was noted in patients receiving VC. These discordant trough associations strengthen the finding that the VPT combination was a significant driver of AKI. These data suggest that the concomitant use of VPT had such a nephrotoxic effect that it muted the impact of vancomycin trough concentrations on AKI. However, when patients received VC (and the toxic effect of VPT was not present), the association between vancomycin troughs and AKI was apparent. These findings could help to explain the discordant literature with respect to the impact of vancomycin trough on AKI, as the type of concomitant antipseudomonal therapy received by patients is rarely reported, let alone controlled for. Of note, the associations between vancomycin trough and AKI are particularly robust, as only trough values obtained before the onset of AKI were included. Because elevated vancomycin troughs that occurred as a result of AKI were excluded, the association between vancomycin trough and AKI was unbiased.

The findings of this study are largely consistent with those found in other studies that analyzed comparative AKI risks of VPT and VC. In a smaller analysis, Gomes and colleagues demonstrated similar findings, with 35% of VPT and 13% of VC patients developing AKI [3]. In a propensity score-matched subgroup, VPT was independently associated with increased AKI risk (OR, 5.67; 95% CI, 1.66–19.33). Similarly, in an analysis that was conducted to assess the impact of generic vancomycin product on development of AKI, Sutton and colleagues reported concomitant VPT to be the strongest predictor of AKI in the cohort (OR, 3.97; 95% CI, 1.66–9.50) [11].

However, the association between VPT and AKI is not a universal finding. Although Moenster and colleagues reported that AKI occurred in 29% of patients on VPT and 13% of patients on VC, this difference failed to reach statistical significance (OR, 3.45; 95% CI, 0.96–12.4) [12]. Importantly, the study was underpowered, and numerically these findings are consistent with those from the aforementioned studies. Hammond and colleagues also recently analyzed comparative toxicity rates in an ICU population. In their analysis AKI was reported in 33% of patients on VPT and 29% of patients on VC; P = .65 [4]. It warrants mention that this study was powered to detect a difference in AKI rates of 36.5% vs 15% in the 2 groups and therefore was underpowered to identify more subtle differences in AKI rates, particularly in an ICU population with competing AKI risks.

The data presented here are robust, overcome several limitations found in the previous literature, and convincingly demonstrate that, compared to VC, combination therapy with VPT is associated with a higher overall incidence of AKI, a more rapid onset of AKI, and a persistently increased daily AKI risk throughout the first week of therapy. Despite the robustness of our methodology, there are a few limitations. This was a single-center, retrospective analysis and is thus subject to the inherit biases associated with this type of study design, and the results should be confirmed in other patient populations. In addition, only approximately 20% of patients in this study were cared for in the ICU; therefore, the results might not be generalizable to the ICU patient population. Furthermore, while the definition of combination therapy used in this manuscript is rational (≥48 hours of combination therapy where each agent was started within 24 hours of the other), definitions used by investigators in other analyses differ slightly (ranging from a requirement of administration of the combination for ≥48–72 hours, with or without the requirement that the agents were started within 48 hours of one other). However, these relatively minor differences are unlikely to explain differences between the findings presented here and those in prior publications. Finally, we chose to exclude patients with baseline renal insufficiency. Patients with baseline renal insufficiency represent an important patient population at risk for developing AKI and warrant evaluation in future studies.

In conclusion, combination therapy with VPT was independently associated with a 4-fold increased risk of AKI compared to combination therapy with VC. Additionally, AKI with VPT occurred in a more rapid fashion. Despite this rapid onset of AKI, there are opportunities for providers to limit the incidence of this adverse event. Data recently published by our group [10] demonstrated that the highest daily incidence of AKI among patients receiving VPT occurred on day 4 and day 5 of therapy. Therefore, timely de-escalation or discontinuation of 1 or both of the combination agents would likely mitigate AKI risk. However, given the association between VPT and increased AKI risk, it is critical that clinicians consider all risks and benefits of therapy (both efficacy and toxicity) when selecting empiric combination regimens. Clinicians might choose an alternative to piperacillin–tazobactam in settings where vancomycin is coadministered. If antibiogram data demonstrate an advantage with regard to activity against likely gram-negative pathogens of empiric piperacillin–tazobactam, clinicians might combine piperacillin–tazobactam with an alternative gram- positive agent. Because overuse of vancomycin alternatives might be concerning from a stewardship perspective, one approach might be to limit use of combination therapy with vancomycin alternatives and piperacillin–tazobactam to patients who are hemodynamically unstable and thus more likely to be significantly harmed by ineffective empiric gram-negative coverage, while using vancomycin plus cefepime in more stable patients.

Swimming in a Sea of Vancomycin and Piperacillin-Tazobactam: What About the Kidneys?


Rounding in the intensive care unit (ICU), you’re three patients deep with 20-plus to go. Already you’re bobbing in a vancomycin-piperacillin-tazobactam (vanc-PT) sea, your mouth salty from the carrier. Every patient is on both, and you suspect the nurses and therapists are too. The intern appears to have red man syndrome, and you begin to wonder about your own vancomycin level. Is it therapeutic? Has it affected your kidneys?

Rather than outsource 100% of your thought processes to house staff and hospital culture, you pause to reflect. Empirical coverage with broad-spectrum antibiotics in the ICU is common and consistent with guidelines. Sepsis is more frequent than a Detroit Pistons loss, and time to coverage drives outcomes. But doesn’t vanc-Zosyn come at a cost? Doesn’t it damage the kidneys?

Vancomycin’s effects on the kidney border on urban legend. I’ve been told it doesn’t cause kidney injury since they changed from the Mississippi mud formulation — or was that term just used for the title of an editorial? Another gadfly might tell you that the levels required to cause harm are above the therapeutic range and we need not worry. There’s some truth to both legends, given that formulation impurities have been corrected and the Infectious Diseases Society of America (IDSA) says the vancomycin and nephrotoxicity data are subject to confounding. They also note that vancomycin toxicity is most likely after extended infusions targeting trough levels > 15-20 mg/L and is otherwise uncommon. Urban legend somewhat dispelled, vancomycin may still cause nephrotoxicity, but less so sans Mississippi mud or higher targets.

What about the vanc-PT combination? In 2017, my academic program reviewed a paper comparing acute kidney injury (AKI) with vanc-PT vs vanc-cefepime. AKI rates were three times higher with vanc-PT. Vanc trough levels were associated with AKI in the presence of cefepime but not PT, a finding the authors attributed to synergistic vanc-PT toxicity that’s unrelated to dosing. I never considered abandoning vanc-PT, but the nephrotoxicity concern remained buried in my subconscious beside maintenance of certification (MOC) exams, aspirational recycling, and other phenomena I don’t want to deal with but can’t seem to forget.

A review published in 2021 provided the deep dive I never had time to perform. Although I recommend reading it for yourself, I’ll try and summarize here. Just like the 2023 college football playoff rankings, the reality is complicated. To start, most studies (including the 2017 paper I cited earlier) are retrospective and observational. Vanc-PT is typically used for hospitalized patients who are pretty sick and often have sepsis. There’s usually suspicion for drug-resistant organisms, thus further selecting for the presence of comorbid disease — in short, confounding by indication and related to multiple patient factors that increase the risk for renal injury. Many meta-analyses have been done, but grouping flawed studies just amplifies the bias and instills false confidence. Finally, whereas serum creatinine changes are well described, the effects on long-term clinical outcomes are not. After reading this review, I thought vanc-PT nephrotoxicity should have stayed buried in my subconscious.

Sepsis and hospital/ventilator-associated pneumonia (HAP/VAP) guidelines don’t help much here. Surviving Sepsis makes no comment on specific antibiotics. The American Thoracic Society/IDSA HAP/VAP guidelines make a case against pairing vancomycin with carbapenems, aminoglycosides, and other drugs for various reasons but make no distinction between vanc-PT vs vanc plus a cephalosporin (cefepime or ceftazidime). Cost may be an issue within a given system, but PT, cefepime, and ceftazidime are all generic.

This past year we finally got what we needed: a prospective randomized study called ACORN. The authors randomly assigned 2511 patients to receive PT vs cefepime (77% of whom were also receiving vancomycin). Renal outcomes weren’t different in the PT group but, patients in the cefepime arm experienced fewer days alive and free of coma or delirium at day 14 (a secondary outcome). On balance, these results favor PT. These data are better than anything else we have. It’s not even close.

In summary, the vanc-PT association with nephrotoxicity was overblown. It is no more. In a high-acuity unit, you’d better be drowning in it. You can stop worrying about effects on the kidney. Now it’s time to go sign up for my MOC exam.

Reducing Inappropriate Vancomycin Use in Cancer Patients


While he was an attending physician at Montefiore Medical Center in New York City, Adam F. Binder, MD, and his colleagues developed an initiative to encourage the appropriate use of intravenous vancomycin for cancer patients with neutropenic fever.

For this work, he was hailed as a Choosing Wisely Champion here at the recent American Society for Hematology (ASH) 2018 annual meeting.

This program is run by ASH in cooperation with the American Board of Internal Medicine Foundation (ABIM), which began the Choosing Wisely initiative in 2012. The annual Choosing Wisely Champions campaign recognizes the efforts of practitioners who are working to eliminate the costly and potentially harmful overuse of tests and procedures.

In a presentation about his work, Binder explained that febrile neutropenia is a relatively common adverse event associated with myelosuppressive cancer therapies, and current guidelines for empiric therapy do not recommend the use of vancomycin.  So he was rather concerned to find that clinicians were overprescribing vancomycin for neutropenic fever.

“There was some anecdotal concern from oncologists and the antibiotic stewardship team that we may have been overprescribing vancomycin,” he said. “Some of the preliminary data that turned this from just anecdotal to objective evidence came out of a larger project that was looking at methicillin-resistant Staphylococcus aureus (MRSA) screening as a way to de-escalate the use of vancomycin in patients presenting with pneumonia.”

For that project, they looked at a cohort of 88 patients with hematologic malignancies who either presented to the emergency department with neutropenic fever or developed it in hospital.

Of this group, 45 patients (51%) had an inappropriate initiation of vancomycin.

“If vancomycin was started in the ER and then discontinued when they got to the floor, that was considered to be inappropriate, because the oncology team made the decision that it wasn’t necessary,” explained Binder.

Next, Binder and colleagues identified what he believed were the top three barriers to more appropriate use of vancomycin. One was changing individual practice patterns. “Physicians get used to practicing a certain way, even though there’s evidence suggesting that vancomycin up front doesn’t improve overall mortality,” he said. “But some may feel comfortable starting broader and then de-escalating.”

Another challenge was the ever-changing residency staff. “We had residents rounding on service and switching every 2 to 4 weeks, and most were second- and third-year residents and most of them had never rounded on an oncology floor,” he said. “So they didn’t know the appropriate management of neutropenic fever.”

A third barrier was a lack of nursing empowerment in driving change or for enforcing guidelines.

An interdisciplinary team was created, which included an antibiotic stewardship team, pharmacists, and hematologists, and together they developed an institutional algorithm to guide prescriptions related to febrile neutropenia. They also conducted recurring educational initiatives emphasizing criteria for appropriate vancomycin initiation based on well-established guidelines.

“The whole team gave their input and having everyone’s consensus meant that it was more likely that the algorithm would be followed than if it had been developed by just a small group,” Binder commented.

Once the guidelines were in place, they wanted to make sure that they were easily accessible. “That’s one of the problems,” he said. “One of our concerns is that guidelines are put online and you can click on them to read them, but no one ever looks at them. We wanted to make sure that the guidelines were seen.”

To circumvent that problem, the guidelines were printed out and laminated and these boards were placed at all work stations. “So no matter where you were, and no matter where you sat down, you’d have them staring at you in the face,” Binder emphasized. “A year later and the boards are still up and people are still using them. It’s a nice example of sustainability.”

After this intervention was put in place, along with educational initiative, inappropriate initiation of vancomycin dropped to 34%. There was also a 1-day reduction in the duration of vancomycin therapy.

Binder said this is an improvement to what they had seen previously (51% inappropriate vancomycin), but there is still a lot of work to be done, “because 34% is still high.”

“This is really just the beginning,” he said. “The changes need to be sustainable, and we need to continue to look for new interventions. One of our next projects is to have a clinical decision-making tool within the order set.”

De-escalating Antibiotics: A Nudge in the Right Direction


Failure to De-escalate Empirical Vancomycin

Empirical antimicrobial treatment in acute care settings is often the result of the “diagnosis momentum” heuristic, wherein the antibiotics started in one location for “sepsis” are continued for several additional days after transfer to another location. Vancomycin remains one of the most commonly prescribed inpatient antibiotics, despite a decline in the prevalence of invasive methicillin-resistant Staphylococcus aureus (MRSA) infections. Two recent studies highlight the infrequent isolation of MRSA in patients receiving empirical vancomycin and diagnostic stewardship interventions to promote vancomycin de-escalation.

Waters and colleagues[1] compared the rate of vancomycin use with subsequent positive cultures justifying treatment with vancomycin in a single-center retrospective observational study. Most of these infections were skin and soft-tissue infections, bacteremia, and pneumonia.

Concern about MRSA is probably the main driver of vancomycin use, yet this organism was confirmed in only 8.4% of the positive cultures. During the 3-month study, only 11% of 1662 patients on vancomycin had a positive culture necessitating vancomycin use as definitive therapy.

Empirical vancomycin can probably be safely de-escalated in nearly 90% of patients, especially after 48 hours of negative cultures. However, convincing prescribers to discontinue antibiotics presents a separate challenge for antimicrobial stewardship programs.

A Nudge Toward De-escalation

Musgrove and colleagues[2] capitalized on the fact that most decisions to start empirical vancomycin and piperacillin-tazobactam for pneumonia occur in the context of concern about unidentified MRSA or Pseudomonas aeruginosa. They performed a quasi-experimental study within a four-hospital system in Detroit to evaluate the impact of a modification to respiratory culture reports on antibiotic use. The microbiology lab changed the way in which they report normal “commensal respiratory flora” (which includes Neisseria, Corynebacterium, and Streptococcus, with no dominant growth of any single organism) to clinicians. They added the statement “No S aureus/MRSA or P aeruginosa” to the report, and the antibiotic stewardship program provided a brief education to prescribers.

After this behavioral nudge was implemented, prescribers were 34% (P < .01), or 5.5-fold, more likely to de-escalate antibiotics. Furthermore, with fewer vancomycin/piperacillin-tazobactam combination days of therapy, they noted a 17% reduction in acute kidney injury (P < .03) even after adjustment for severity of illness. The days of therapy for MRSA and Pseudomonas were reduced from 7 to 5 days (P < .01). The investigators observed additional opportunity for de-escalation, because intervention patients still received a median of 5 days of anti-MRSA and anti-pseudomonal therapy.

Multidrug-resistant organisms (MDROs) were not prevalent in either group; however, there was a significant reduction in development of subsequent MDROs after the culture result nudge (8% vs 1%; P =.035). There was no effect on mortality or development of Clostridium difficile infection, or change in length of intensive care unit or hospital length of stay.

Viewpoint

Behavioral nudges, which use positive reinforcement and indirect messaging to influence decision-making, already exist in many areas of our clinical environment. This study highlights the importance of clear communication of microbiology results as a means to influence antibiotic use. As antibiotic stewardship programs aim to collaborate with prescribers to change behaviors, these behavioral nudges can be useful low-effort/high-yield tools to further assist with antibiotic de-escalation.

Reducing acute kidney injury due to vancomycin in trauma patients


Supratherapeutic vancomycin trough levels are common after trauma and associated with both increased acute kidney injury (AKI) and mortality. We sought to limit the adverse effects of vancomycin in trauma patients through more frequent trough monitoring.

METHODS: Beginning in January 2011, trauma patients treated with vancomycin had trough levels (VT) monitored daily until steady state was reached. Trauma patients admitted from January 2011 to May 2015 (POST) were compared with those admitted from January 2006 to December 2010 (PRE). Inclusion criteria required administration of intravenous vancomycin, admission serum creatinine (SCr), and SCr within 72 hours of highest VT. Acute kidney injury was defined as an increase in SCr of at least 0.3 mg/dL or 50% from admission to post–vancomycin administration. Those in the POST group were prospectively followed up until discharge or death.

RESULTS: Two hundred sixty-three patients met inclusion criteria in the PRE-phase and 115 in the POST-phase. The two groups were similar in age, gender, race, body mass index, pre-existing comorbidities, admission systolic blood pressure, Glasgow Coma Scale, and head Abbreviated Injury Scale. Injury Severity Score was higher in the POST cohort (18 PRE vs. 25 POST, p < 0.001). Compared with PRE, the POST cohort had lower rates of supratherapeutic VT (>20 mg/L) (34.6% PRE vs. 22.6% POST, p = 0.02) and AKI (30.4% PRE vs. 19.1% POST, p = 0.026). After adjusting for confounders, the POST group had a significantly lower risk of AKI with an adjusted odds ratio of 0.457 (p = 0.027). There was a trend toward decreased mortality in the POST cohort, but this did not reach significance (10% PRE vs. 5.2% POST, p = 0.162).

CONCLUSIONS: A reduction in AKI was observed in trauma patients with daily vancomycin trough levels monitored until steady state. Increased awareness regarding closer surveillance of VT in trauma patients may limit the incidence of vancomycin-related nephrotoxicity.

MSSA Bloodstream Infection: Beta-Lactams or Vancomycin?


Beta-lactams are superior to vancomycin for definitive therapy, but not empiric treatment of methicillin-susceptible Staphylococcus aureus (MSSA) bloodstream infection, according to a large retrospective study.

Jennifer S. McDanel, PhD, from the University of Iowa in Iowa City and the Iowa City Veterans Affairs Health Care System, and colleagues published their findings online April 21 in Clinical Infectious Diseases.

Although prior research has linked vancomycin therapy to poor patient outcomes such as recurrence, treatment failure, and death, the validity of this construct with respect to empiric therapy has remained unclear.

“Vancomycin is often prescribed empirically for patients suspected of having S. aureus bloodstream infections since it has activity against both methicillin-resistant and methicillin susceptible strains,” the authors write.
“However, for a patient infected with [MSSA], organizations such as the Infectious Diseases Society of America…recommend switching therapy to a beta-lactam, such as cefazolin or an antistaphylococcal penicillin (nafcillin or oxacillin) once the isolate is known to be MSSA.”

Accordingly, the researchers analyzed data for MSSA culture-positive patients at 122 Veterans Affairs hospitals who received either a beta-lactam or vancomycin alone as empiric therapy (n = 2659 and 3125, respectively) or definitive treatment (n = 4698 and 935, respectively) during 2003 to 2010.

The researchers defined empiric therapy as treatment started 2 days before through 4 days after collection of the first culture positive for MSSA; definitive therapy started 4 to 14 days after collection of the first culture positive for MSSA.

Empiric use of beta-lactam monotherapy had no effect on 30-day mortality compared with vancomycin (hazard ratio [HR], 1.03; 95% confidence interval [CI], 0.89 – 1.20).

As expected, however, definitive therapy with a beta-lactam rather than vancomycin was associated with a significant 35% decrease in 30-day mortality (HR, 0.65; 95% CI, 0.52 – 0.80), with use of cefazolin or an antistaphylococcal penicillin yielding further benefit (HR, 0.57; 95% CI, 0.46 – 0.71).
Although the researchers made adjustments for variables such as age, dialysis/end-stage renal disease, Acute Physiology and Chronic Health Evaluation (APACHE) III score, and Charlson Comorbidity index, the authors note that the findings may be limited by the elderly nature of the study population, and some of the reported deaths may not have been attributable to MSSA. In addition, the researchers did not stratify the definitive therapy analysis by empiric therapy, and therefore did not account for potential switching-related survival benefits.

MSSA Bloodstream Infection: Beta-Lactams or Vancomycin?


Beta-lactams are superior to vancomycin for definitive therapy, but not empiric treatment of methicillin-susceptible Staphylococcus aureus (MSSA) bloodstream infection, according to a large retrospective study.

Jennifer S. McDanel, PhD, from the University of Iowa in Iowa City and the Iowa City Veterans Affairs Health Care System, and colleagues published their findings online April 21 in Clinical Infectious Diseases.

Although prior research has linked vancomycin therapy to poor patient outcomes such as recurrence, treatment failure, and death, the validity of this construct with respect to empiric therapy has remained unclear.

“Vancomycin is often prescribed empirically for patients suspected of having S. aureus bloodstream infections since it has activity against both methicillin-resistant and methicillin susceptible strains,” the authors write.
“However, for a patient infected with [MSSA], organizations such as the Infectious Diseases Society of America…recommend switching therapy to a beta-lactam, such as cefazolin or an antistaphylococcal penicillin (nafcillin or oxacillin) once the isolate is known to be MSSA.”

Accordingly, the researchers analyzed data for MSSA culture-positive patients at 122 Veterans Affairs hospitals who received either a beta-lactam or vancomycin alone as empiric therapy (n = 2659 and 3125, respectively) or definitive treatment (n = 4698 and 935, respectively) during 2003 to 2010.

The researchers defined empiric therapy as treatment started 2 days before through 4 days after collection of the first culture positive for MSSA; definitive therapy started 4 to 14 days after collection of the first culture positive for MSSA.

Empiric use of beta-lactam monotherapy had no effect on 30-day mortality compared with vancomycin (hazard ratio [HR], 1.03; 95% confidence interval [CI], 0.89 – 1.20).

As expected, however, definitive therapy with a beta-lactam rather than vancomycin was associated with a significant 35% decrease in 30-day mortality (HR, 0.65; 95% CI, 0.52 – 0.80), with use of cefazolin or an antistaphylococcal penicillin yielding further benefit (HR, 0.57; 95% CI, 0.46 – 0.71).
Although the researchers made adjustments for variables such as age, dialysis/end-stage renal disease, Acute Physiology and Chronic Health Evaluation (APACHE) III score, and Charlson Comorbidity index, the authors note that the findings may be limited by the elderly nature of the study population, and some of the reported deaths may not have been attributable to MSSA. In addition, the researchers did not stratify the definitive therapy analysis by empiric therapy, and therefore did not account for potential switching-related survival benefits.

Tapeworm Drug Effective at Treating MRSA


With the ever persistent threat of bacterial drug-resistance looming like a carrion bird waiting for a meal, scientists are continually on the hunt for new therapeutics to thwart infections like those caused by methicillin-resistant Staphylococcus aureus (MRSA). Fortuitously, scientists from Brown University have come across two drug compounds that are already in use to treat human tapeworm infections, which they report in a new study show great promise in stopping MRSA infections.

The findings from this study were published recently in PLOS ONE under an article entitled “Repurposing Salicylanilide Anthelmintic Drugs to Combat Drug Resistant Staphylococcus aureus.”

The Brown researchers screened over 600 drugs for effectiveness against MRSA, using an in vivo assay that cultures live nematode worms infected with the drug-resistant bacteria. The investigators found two compounds niclosamide, which is on World Health Organization’s list of essential medicines, and oxyclozanide, a closely related veterinary drug, were effective at suppressing MRSA cultures. Moreover, both drugs were observed to be as effective as the current last-line clinical treatment, vancomycin.

“Since niclosamide is FDA approved and all of the salicylanilide anthelmintic drugs are already out of patent, they are attractive candidates for drug repurposing and warrant further clinical investigation for treating staphylococcal infections,” explained Rajmohan Rajamuthiah, Ph.D. a postdoctoral scholar in the Warren Alpert Medical School of Brown University and first author on the current study.

Dr. Rajamuthiah and his colleagues found that oxyclozanide was more effective at killing MRSA, while niclosamide was more bacteriostatic—effectively suppressing, but not completely eradicating the bacteria. However, the researchers speculate that niclosamide may still provide enough of a kick to keep MRSA at bay while the immune system gets up to speed handling the infection.

While results from the current study are very encouraging and have Dr. Rajamuthiah and his colleagues feeling optimistic, the researchers did point out a caveat that the feel warrants further analysis. Drugs such as oxyclozanide and niclosamide are rapidly cleared by the body and are less effective at diffusing out of the bloodstream and into peripheral tissues, where some MRSA infections could reside.

“The low level of systemic circulation coupled with the rapid elimination profile of niclosamide suggests the necessity for further testing of the potential of niclosamide and oxyclozanide for treating systemic infections,” wrote the scientists. “Further studies should include the evaluation of these compounds in systemic and localized infection models in rodents.”

However, the flipside of the rapid clearance scenario is that drugs may impart very limited toxicity to patients. In order to determine the actual effects of these drugs in mammals, the researchers have planned a series of experiments in rodents to determine the two compounds efficacy and overall toxicity, when used to treat MRSA infections.

“The relatively mild toxicity of oxyclozanide is encouraging based on in vitro tests,” stated Dr. Rajamuthiah. “Since it has never been tested in humans and since it belongs to the same structural family as niclosamide, our findings give strong impetus to using oxyclozanide for further investigations.”

Study: elevated vancomycin MICs no cause for concern


Elevated vancomycin minimum inhibitory concentrations do not increase the risk of death in patients with Staphylococcus aureus bacteremia, according to the findings of a comprehensive meta-analysis published in the Oct. 9 issue of JAMA.

Despite widespread speculation about rising vancomycin resistance, or “MIC creep,” the authors find little evidence to challenge the current CLSI susceptibility breakpoint of ≤ 2 µg/mL for vancomycin.

Dr. Kalil

“After an extensive approach and analysis, we found that MICs of one and two have pretty much similar mortality,” says coauthor Andre C. Kalil, MD, MPH, a professor of medicine in the Infectious Diseases Division of the University of Nebraska Medical Center. The results remained consistent across a variety of study designs, microbiological susceptibility assays, MIC cutoffs, clinical outcomes, durations of bacteremia, and histories of previous vancomycin exposure and treatment.

The findings may help boost clinicians’ confidence in vancomycin therapy and stave off opportunities for S. aureus to develop multidrug resistance, the authors note.

“Based on an MIC of two, some physicians might change the patient’s therapy to daptomycin because the literature essentially suggested there were vancomycin failures if the MIC was over one,” says coauthor Paul D. Fey, PhD, D(ABMM), a professor of pathology and microbiology at the University of Nebraska Medical Center and medical director of the clinical microbiology laboratory. Dr. Fey interpreted the susceptibility test results and contributed microbiology expertise to the study.

Dr. Fey

That strategy may be unnecessary, the new findings suggest. “In terms of the bedside, an elevated MIC by itself should not lead clinicians to rush to switch therapies in patients with SAB [S. aureus bacteremia],” Dr. Kalil says. “If we can prevent unnecessary changes to new or alternative antibiotics, we’re not only helping patients who are being treated now—we’re also helping future patients with these infections.”

The Nebraska Medical Center study refutes three previous meta-analyses linking elevated vancomycin MICs to poor patient outcomes. Those analyses have been noted for including heterogeneous patient populations, combining multiple sites of infection, and evaluating bias-laden endpoints such as treatment failure.

“This is certainly an important contribution to the existing evidence in this subject matter,” says Stefan Riedel, MD, PhD, D(ABMM), of the recently published study. He is an assistant professor of pathology at The Johns Hopkins University and director of the clinical pathology laboratories at Johns Hopkins Bayview Medical Center. He is also a member of the CAP’s Microbiology Resource Committee.

“What I liked about this article, contrary to some of the other meta-analyses, is that it really focused on bloodstream infections,” says Dr. Riedel, who was not involved with the study. “They have a very concise patient population in which to assess the question they’re asking, which is this: Does the MIC that the microbiology lab reports influence the outcome of treatment?”

According to the findings, the answer is no—but there’s more to the story.

“MRSA is a significant problem in clinical medicine, but the question is how significant is it?” asks Dr. Riedel. “Clinicians hear that some antimicrobial susceptibility testing methods may render either slightly higher or lower MICs and they say, well, maybe we’re overcalling or undercalling resistance. Maybe there’s a MIC creep; maybe there will be a treatment failure. So they turn to the laboratory and to the pathologists and ask, ‘What are the MIC distributions for MRSA in the laboratory? What are the susceptibility test methods used in your laboratory? Do we need to use alternative drugs for treatment?’”

Pathologists and laboratory professionals are faced with the difficult task of guiding clinicians through these uncertainties, Dr. Riedel says.

To provide definitive answers to these longstanding questions, Dr. Kalil and coauthor Trevor C. Van Schooneveld, MD, spent nearly two years searching reports of mortality and vancomycin MIC data in humans with S. aureus bacteremia. Aiming to find every article published on vancomycin-resistant S. aureus, the authors searched PubMed, Embase, the Cochrane Library, Evidence-Based Medicine BMJ, and the American College of Physicians Journal Club from inception through April 2014. They examined abstracts presented at annual meetings of the Infectious Diseases Society of America, the Interscience Conference on Antimicrobial Agents and Chemotherapy, and the Society for Healthcare Epidemiology of America. Occasionally, they contacted the authors of old studies for clarification.

“It was a very long process, going through literally thousands of abstracts and references,” Dr. Kalil recalls. In all, 38 studies, encompassing 8,291 patients with SAB, met their inclusion criteria. Then they began the painstaking process of extracting information from the reports.

“There were many, many variables. It was fascinating because other meta-analyses had not really examined these many variables,” Dr. Kalil says. The authors tracked patient age, MIC cutoffs, type of susceptibility assay, heteroresistance test results, methicillin resistance status, duration of bacteremia, intensive care unit exposure, APACHE II score, Charlson score, previous vancomycin exposure, presence of endocarditis, antistaphylococcal drugs used for treatment, vancomycin trough levels, and all-cause mortality.

“We looked at every facet of the problem. Literally, the variables that you see here represent everything you could ever collect from these studies. There’s nothing more that could be extracted,” Dr. Kalil says.

The database they created to house the entries grew until it became unwieldy. “You could scroll down the computer screen, and scroll and scroll, and it just kept going,” he says, laughing. “It’s funny because you think it shouldn’t be too hard. I mean, the studies are already published. All you have to do is a literature search. But this was an exhaustive effort.”

As they parsed the various studies, the authors defined high-vancomycin MIC as values greater than or equal to 1.5 mg/L as measured by the Epsilometer test, or E-test. Clinicians tend to be more likely to explore alternative antimicrobial treatment options when the MIC is 1.5 or higher, Dr. Fey notes.

Subgroup analyses were performed to assess mortality with regard to different MIC cutoffs, microbiology susceptibility assays, methicillin-resistant status, and the presence of heteroresistance. The authors focused on all-cause mortality, however, as the primary outcome.
Their findings revealed an overall mortality rate of 26.1 percent for the 8,291 episodes of SAB. When all data were pooled together, mortality was 26.8 percent versus 25.8 percent in patients with high- versus low-vancomycin MICs, respectively.

When the authors looked exclusively at the highest-quality studies performed to date, as scored on the Newcastle-Ottawa scale, they calculated an estimated mortality rate of 26.2 percent versus 27.8 percent in patients with high- versus low-vancomycin MICs, respectively.

Of the 7,232 patients with methicillin-resistant S. aureus infections, mortality was 27.6 percent versus 27.4 percent in patients with high- versus low-vancomycin MICs, respectively.

Dr. Riedel and colleagues conducted a study in which they compared the performance of various commercially available susceptibility test methods against the gold standard method, the CLSI broth dilution method (Riedel S, et al. J Clin Microbiol. 2014;52[6]:2216–2222). They found evidence of variation in MICs—specifically, a difference of one doubling dilution—depending on the test method used. The JAMA study noted a similar variation between broth microdilution and E-test results. But within each method, mortality rates remained similar for high- and low-vancomycin MICs, the authors report.

“The interesting concept is that clinicians may look at a MIC of two and say, ‘Well that’s why the patient doesn’t respond.’ My impression from this study is that it may be simplistic to think that it’s the MIC breakpoint. Clinicians need to search for other possible causes of a perceived or real vancomycin treatment failure rather than turning straight to the MIC,” Dr. Riedel says.

The severity index of the disease may play a role, he postulates, or perhaps a treatment failure could result from the inability to achieve an appropriate serum concentration of vancomycin for a particular patient.

The role of microbial pathophysiology and virulence in regard to patient mortality is complex. Previous studies have observed declining growth rates and reduced virulence in bacteria that acquire vancomycin resistance gradually through the stepwise acquisition of compensatory mutations, as might occur in a patient on long-term vancomycin therapy. The JAMA findings point to the same conclusion, Dr. Kalil notes. “The suggestion here is that not every S. aureus with a high MIC is going to be more harmful. Some may even be less harmful. That’s why, when you look at our results, you see a range that goes from better survival to worse survival. When you put the numbers together, no survival difference was observed.”

Dr. Riedel

Though the study focused on patients with bacteremia, the data set included patients with infections at various sites, including the skin, soft tissue, urinary tract, and lungs. “That’s an important point for understanding the relation to mortality rates,” Dr. Riedel explains. “If someone has a soft tissue infection, a deep-seated abscess, or maybe an osteomyelitis and is now bacteremic with MRSA, then it is certainly conceivable that this type of bloodstream infection will be much more difficult to eliminate and could therefore be perceived as vancomycin failure using a standard approach. Some of the other meta-analyses did not make this point.”

Dr. Kalil notes that the authors’ meticulous endeavor almost didn’t pay off. During the scramble to gather data for the study, other meta-analyses were published on the same topic.
“We thought for sure we had been scooped,” Dr. Kalil recalls. “But the fun part is that even though it took us longer than usual, and other studies had been published, we stuck together and made a decision to go for a high-profile journal, because our message has very important implications for both practice and research.”