Abstract
Background
Vancomycin and piperacillin/tazobactam are reported in clinical studies to increase acute kidney injury (AKI). However, no clinical study has demonstrated synergistic toxicity, only that serum creatinine increases.
Objectives
To clarify the potential for synergistic toxicity between vancomycin, piperacillin/tazobactam and vancomycin + piperacillin/tazobactam treatments by quantifying kidney injury in a translational rat model of AKI and using cell studies.
Methods
(i) Male Sprague–Dawley rats (n = 32) received saline, vancomycin 150 mg/kg/day intravenously, piperacillin/tazobactam 1400 mg/kg/day intraperitoneally or vancomycin + piperacillin/tazobactam for 3 days. Urinary biomarkers and histopathology were analysed. (ii) Cellular injury was assessed in NRK-52E cells using alamarBlue®.
Results
Urinary output increased from Day −1 to Day 1 with vancomycin but only after Day 2 for vancomycin + piperacillin/tazobactam-treated rats. Plasma creatinine was elevated from baseline with vancomycin by Day 2 and only by Day 4 for vancomycin + piperacillin/tazobactam. Urinary KIM-1 and clusterin were increased with vancomycin from Day 1 versus controls (P < 0.001) and only on Day 3 with vancomycin + piperacillin/tazobactam (P < 0.001, KIM-1; P < 0.05, clusterin). The histopathology injury score was elevated only in the vancomycin group when compared with piperacillin/tazobactam as a control (P = 0.04) and generally not so with vancomycin + piperacillin/tazobactam. In NRK-52E cells, vancomycin induced cell death with high doses (IC50 48.76 mg/mL) but piperacillin/tazobactam did not, and vancomycin + piperacillin/tazobactam was similar to vancomycin.
Conclusions
All groups treated with vancomycin demonstrated AKI; however, vancomycin + piperacillin/tazobactam was not worse than vancomycin. Histopathology suggested that piperacillin/tazobactam did not worsen vancomycin-induced AKI and may even be protective.
Introduction
Vancomycin and piperacillin/tazobactam are two of the most commonly utilized antibiotics in hospitalized patients.1 Meta-analyses2 compiling over 25 000 patients suggest that vancomycin + piperacillin/tazobactam increases acute kidney injury (AKI) by an absolute of 9% against vancomycin + comparators with mean ORs ranging from 1.6- to 3.6-fold higher risk. This suggested that synergistic toxicity is concerning, and several experts recommend avoiding vancomycin + piperacillin/tazobactam3,4 even though vancomycin + piperacillin/tazobactam is a drug of choice in numerous national guidelines. Also concerning, other alternatives for piperacillin/tazobactam have equal limitations: cefepime is associated with dose-dependent neurotoxicity,5 fluoroquinolones have numerous black-box warnings,6 carbapenems promote broad antibiotic resistance,7 and aminoglycosides cause worse AKI.8 Thus, defining the AKI profile for vancomycin + piperacillin/tazobactam is imperative; even moderate AKI increases mortality and prolongs hospitalization.9,10
Despite recommendations to avoid vancomycin + piperacillin/tazobactam, the biological plausibility of the increased renal damage has not been established11. Previous studies have relied on serum creatinine (SCr) as an AKI surrogate. While SCr is easily measured, it is neither highly sensitive nor specific for AKI. False positives for AKI exist because SCr transit is defined by secretion and re-absorption (in addition to free filtration)12 and SCr can be elevated for reasons unrelated to filtration even when the kidney is not injured because of drug competition for renal tubular secretion.13 Unlike vancomycin, which causes acute tubular necrosis via oxidative stress at the renal proximal tubule or uromodulin interaction/cast formation,14,15 piperacillin/tazobactam rarely causes AKI.14,16 To date, only acute interstitial nephritis (AIN) has been cited with piperacillin/tazobactam. Given that the absolute increase in the incidence of AKI in patients with vancomycin + piperacillin/tazobactam is very high (9%),2 it is extremely unlikely that the rare AIN drives toxicity. Furthermore, no clinical study has demonstrated that synergistic toxicity occurs because of these mechanisms, only that SCr increases.
While SCr increases are not pathognomonic for AKI, novel urinary biomarkers may be more sensitive and specific for AKI. Additionally, the rat is an excellent model because kidney biomarkers are conserved between rats and humans.17,18 Further validating the rat–human translational link, urinary biomarkers are qualified for rat19 and human drug trials20 by the FDA for drug-induced AKI. We have previously demonstrated that urinary kidney injury molecule-1 (KIM-1) and osteopontin (OPN) predict histopathological damage in a rat model of vancomycin-induced kidney injury (VIKI).21,22 This study sought to clarify the potential for synergistic toxicity between vancomycin, piperacillin/tazobactam and vancomycin + piperacillin/tazobactam treatments by quantifying kidney injury in a translational rat model of AKI and using cell studies.
Materials and methods
Chemicals and reagents
Treatments were clinical grade vancomycin (Hospira, Lake Forrest, IL, USA), piperacillin/tazobactam and cefepime (Apotex Corporation, Weston, FL, USA), gentamicin sulphate USP (Medisca, Plattsburgh, NY, USA) and normal saline (Veterinary 0.9% Sodium Chloride Injection USP, Abbott Laboratories, North Chicago, IL, USA). For LC–MS/MS, creatinine pharmaceutical secondary standard certified reference material (Sigma–Aldrich), creatinine-d3 (Cayman Chemicals, Ann Arbor, MI, USA), LC–MS/MS grade acetonitrile and methanol (VWR International, Radnor, PA, USA), formic acid (Fisher Scientific, Waltham, MA, USA) and frozen, non-medicated, non-immunized, pooled Sprague–Dawley rat EDTA plasma (BioreclamationIVT, Westbury, NY, USA) were used. Other materials were similar to our previous reports.21,22
Experimental design and animals
The animal toxicology study was conducted at Midwestern University (IACUC #2295) in compliance with the National Research Council’s publication, the Guide.23
Male Sprague–Dawley rats (290–320 g, Envigo, Indianapolis, IN, USA) were randomized to receive vancomycin 150 mg/kg/day (n = 8), piperacillin/tazobactam 1400 mg/kg/day (n = 8) and vancomycin + piperacillin/tazobactam at the same doses (n = 10) or normal saline (n = 6, Figure 1a). Surgeries were as previously described.22 The vancomycin dose was chosen based on previous studies21,24,25 and to approximate the human dose (30 mg/kg/day) allometrically scaled for the rat [i.e. 30 mg/kg × 6.2 (rat factor) = 186 mg/kg].26 The piperacillin/tazobactam dose was chosen to approximate the human dose (225 mg/kg/day) allometrically scaled for the rat (i.e. 225 mg/kg × 6.2 = 1395 mg/kg). Vancomycin was given IV via the left jugular catheter as this has been shown to result in VIKI in our model, and piperacillin/tazobactam was given IP to extend residence time. Animals were placed in metabolic cages on Day −1 and from Day 1 to Day 3 (Figure 1b). Rats received the first dose on Day 1. In addition to saline as a control, animals served as their own controls, and each study day was compared with Day −1. Rats were housed in a light- and temperature-controlled room for the duration of the study and allowed free access to water and food, including the time in which they resided in metabolic cages (Lab Products Inc., Seaford, DE, USA).
Figure 1.
Experimental design. (a) Flow chart of animal dosing. (b) Timeline of the experiments. VAN, vancomycin; TZP, piperacillin/tazobactam.
Blood and urine collection
Blood samples (0.125 mL, maximum 15 samples/animal) were drawn from a right-sided jugular vein catheter in a sedation-free manner when possible,21,22 and plasma was stored at −80°C (Figure 1b). Urine was collected continuously, aliquoted every 24 h and centrifuged (400 g, 5 min), and the supernatant was stored at −80°C.
Plasma creatinine
Plasma concentrations of creatinine were assayed by LC–MS/MS similarly to a method previously described.27 The internal standard was isotope-labelled creatinine-d3 (0.1 mg/mL Cre-d3, Cayman Chemical, Ann Arbor, MI, USA). Creatinine was detected using Q1/Q3 ion transitions at mass-to-charge (m/z) ratios 114.1/44.3 amu as quantifier and 114.1/43.4 amu as qualifier. Cr-d3 internal standard was detected at m/z 117.09/89.2 amu. The assay was linear between concentrations of 0.31 and 40 mg/dL (R2 = 0.999). We calculated the endogenous creatinine concentration in the blank plasma from the linear equation of the standard curve and adjusted the standard curve to reflect the true creatinine concentration in the sample. The intra- and inter-assay precisions were <2%. Greater than 92% accuracy was observed in all standards tested, with an overall mean assay accuracy of 99.8%. For plasma creatinine, baseline was Day 1 (post-surgery).
Urinary biomarkers
Urine samples were analysed using MILLIPLEX® MAP Rat Kidney Toxicity Magnetic Bead Panel 1 (EMD Millipore Corporation, Charles, MO, USA), according to the manufacturer’s protocol as previously described.28,29 MILLIPLEX® Analyst v5.1 Flex software was used to calculate analyte concentrations. For urinary biomarkers, baseline was Day −1 (pre-surgery).
Histopathology
Animals were euthanized via exsanguination while under anaesthesia. Kidneys were harvested and washed in cold isotonic saline. The left kidney was preserved in 10% formalin solution; the right kidney was flash frozen in liquid nitrogen. Histopathological scoring was performed by IDEXX BioAnalytics (West Sacramento, CA, USA) on paraffin-embedded haematoxylin and eosin-stained kidney sections as previously described.22 In brief the PSTC Standardized Kidney Histopathology Lexicon was utilized with categorical scoring according to grades from 0 to 5 (0, no observable; 1, minimal; 2, mild; 3, moderate; 4, marked; and 5, severe pathology).30 Composite scores for each animal were the highest ordinal score from any kidney site.30
Cell viability
Normal rat kidney epithelial cells (NRK-52E, ATCC® CRL1571™)31 were cultured in DMEM (GenClone, Genesee Scientific) with 5% bovine calf serum in 5% CO2 at 37°C. Penicillin and streptomycin were not included in the medium. Cells were either sub-cultured or received fresh growth medium two or three times per week. Cells were used between passages 14 and 30. NRK-52E cells were plated in 96-well half-volume black plates in DMEM. The next day the medium was changed to 10 mM HEPES-buffered Hanks’ balanced salt solution (HHBS) in the absence of serum. Drugs were dissolved in normal saline and added to the cells. Cefepime was a negative control and gentamicin, a known nephrotoxin,32 a positive control. After 24 h of incubation with antibiotics at 37°C, 5% CO2, cell viability was assessed using alamarBlue® (5 μL/well, Invitrogen). Cells remained in the same drug–HHBS solution until the end of the experiment. Plates were read at 48 h (24 h after the addition of alamarBlue®) using an Enspire Multimode Plate reader (Perkin Elmer) with Ex530/Em590 filters. Results were analysed by normalizing the cell metabolism of antibiotic-treated cells to saline controls and expressed as relative fluorescence units. Drug concentrations were transformed to log base 10. Sigmoidal three-parameter dose–response curves were generated after 48 h of incubation with antibiotics alone or combined with vancomycin at a concentration below its IC50 (vancomycin, 1 mg/mL). The relative toxicity was compared using IC50 values (the drug concentration resulting in 50% of maximal reduction in cell viability) obtained using GraphPad Prism version 7.02 (La Jolla, CA, USA).
Statistical analysis
For the rat studies, most statistical analyses were performed using Stata IC 15.1 (except where specifically noted). Decisions to perform analyses with repeated-measures ANOVA or mixed models were based on the Breusch–Pagan/Cook–Weisberg test for heteroscedasticity on the ANOVA model. Departure from constant variance at P < 0.05 served as a trigger to use a mixed model. Plasma creatinine, urinary output and urinary biomarker elevations were compared across groups using a mixed-effects, restricted maximum likelihood estimation regression, with repeated measures occurring over days; measures were repeated at the level of the individual rat. Additionally, LOESS models with 95% CI were created using R version 3.5.133 and the package ggplot234 to circumvent fit assumptions. From the mixed model, contrasts of the marginal linear predictions35 facilitated comparisons of treatment groups versus controls of saline and pre-treatment values (i.e. Day −1). Ordinal logistic regression was used to classify the ordered log-odds of being in a higher histopathological scoring group according to treatment group. Logistic regression was utilized to determine the odds of having a histopathological score ≥2 when treatment groups were compared with saline or piperacillin/tazobactam. All tests were two-tailed, with an a priori level of statistical significance set at an α of 0.05.
For the in vitro studies, graphics were generated and inferential statistics were performed in GraphPad Prism version 7.02 (La Jolla, CA, USA) and R 3.5.1. Mean and SD were calculated for triplicate wells. Comparison of IC50 values across treatment groups was facilitated by constraining a shared bottom value between 0 and 0.3 and a top of 1 for all groups. The extra sum of squares F-test was utilized to compare independent fits with a global fit, whereby a conservative α level >0.2 defined IC50 values that did not differ.
Results
Rat model
In all, 32 rats completed the protocol (Figure 1). Mean plasma creatinine at baseline was 0.51 ± 0.03 mg/dL and did not differ between the groups (Figure 2a). Plasma creatinine rose for vancomycin on Days 2, 3 and 4 from baseline and for vancomycin + piperacillin/tazobactam on Day 4 (P < 0.02 for all), but no treatment group was different from saline. The overall mean total urine output on Day −1 was 6.4 ± 1.9 mL (Figure 2b). Mean total urine output significantly increased on Day 1 following the first dose in the vancomycin-treated animals (P < 0.02) compared with baseline and saline-treated animals and remained elevated over 3 days. On the final day urine output began to fall for vancomycin-treated animals. There was a gradual increase in the mean total urine output in the vancomycin + piperacillin/tazobactam-treated animals compared with baseline and saline-treated animals that was significant on Days 2 and 3 (P = 0.007).
Figure 2.
Plasma and urinary biomarker levels in the rat after treatment with vancomycin (VAN) or piperacillin/tazobactam (TZP) alone or in combination with vancomycin (VAN+TZP). (a) Plasma creatinine change from baseline, (b) urinary output, (c) urinary KIM-1, (d) urinary clusterin and (e) urinary osteopontin levels in animals treated with saline (n = 6), TZP (n = 8), VAN (n = 8) and VAN+TZP (n = 10). Values are expressed as mean ± SD; *P < 0.01 versus saline; #P < 0.05 versus saline; ^P < 0.02 versus baseline. (f) Predictive margins with 95% CI of KIM-1 over 3 experimental days according to treatment group. Mild horizontal perturbation/jitter was applied to the points to enhance visualization. The top line (blue, triangles) is VAN, the intermediate line (red, diamonds) is VAN+TZP and the bottom lines are TZP (green, circles) and saline (black, squares). KIM-1, kidney injury molecule-1; OPN, osteopontin. Plasma creatinine is presented as change from baseline for ease of interpretation, but statistics were performed on the raw data. This figure appears in colour in the online version of JAC and in black and white in the printed version of JAC.
The constant variance assumption was not upheld in repeated-measures ANOVA for KIM-1, clusterin or OPN (P < 0.001 for all). Therefore, the mixed model was utilized. Urinary KIM-1 and clusterin differed between the treatment groups as a function of time, whereas OPN did not (Tables S1 and S2, available as Supplementary data at JAC Online). Changes in these biomarkers over time for each treatment group are graphically displayed in Figure 2(c–f). In the vancomycin group, urinary KIM-1 and clusterin were increased on Days 1, 2 and 3 when compared with saline (P < 0.001 for all; Table S1). For vancomycin + piperacillin/tazobactam versus saline, KIM-1 and clusterin elevations were seen only after 3 days of treatment (P < 0.001 and P = 0.04, respectively); however, both were elevated at Day 3 compared with baseline values (P < 0.001). Similar findings were obtained with the LOESS model and non-overlapping CIs (Figure 2f). OPN did not increase between treatment groups or treatment days (Figure 2e, P > 0.05 for all).
Ordinal histopathological scores were increased after treatment with vancomycin, piperacillin/tazobactam and vancomycin + piperacillin/tazobactam compared with the controls (Figure 3a). Control animals that received normal saline displayed normal kidney tissue histology (Figure 3b). When piperacillin/tazobactam was used as the referent group, histopathological scores were significantly elevated in the vancomycin-treated group (score 2.16, P = 0.044), significantly lower in the saline group (score −2.8, P = 0.02), and not different in the vancomycin + piperacillin/tazobactam group (score 0.29, P = 0.76). In the model to predict histopathological score ≥2, the saline group accurately classified the absence of injury (Table 1). Treatment with vancomycin was associated with a borderline increased risk of injury (OR 11.67, P = 0.058, 95% CI 0.922–147.56). On the other hand, vancomycin + piperacillin/tazobactam treatment was not associated with injury (OR 1.11, P = 0.91, 95% CI 0.16–7.5). The presence of casts was observed in all, but vancomycin, either alone or in combination with piperacillin/tazobactam had a higher incidence of casts. A representative cast is visualized in a stained kidney section from a piperacillin/tazobactam-treated rat (Figure 3c). More casts were observed among vancomycin (eight casts) and vancomycin + piperacillin/tazobactam (nine casts) groups compared with either saline (two casts) or piperacillin/tazobactam groups (four casts). Tubular dilatation and tubular basophilia were observed in all treatment groups but absent in controls. Notably, tubular basophilia and tubular dilatation were evident in every single kidney section in the vancomycin group (n = 8/8, Figure 3d). However, these findings were less common among the piperacillin/tazobactam and vancomycin + piperacillin/tazobactam groups (n = 2 and 4/8 and n = 2 and 4/10, respectively), suggesting possible nephron protection. Tubular degeneration was observed only among vancomycin and vancomycin + piperacillin/tazobactam groups but not in control and piperacillin/tazobactam groups. Tubular degeneration was more pronounced in rats treated with vancomycin (Figure 3d) compared with the vancomycin + piperacillin/tazobactam group (Figure 3e).
Figure 3.
Histopathological assessment of rat kidney tissue after treatment with vancomycin (VAN) alone, piperacillin/tazobactam (TZP) alone or TZP in combination with VAN (VAN+TZP). (a) Quantitative analysis of histopathology score in kidney tissues of rats. Values are expressed as mean ± SD; *P < 0.044 versus saline. Representative photomicrographs of haematoxylin and eosin-stained rat kidney sections. (b) Normal histology of kidney tissue in control rats administered normal saline, (c) tubular casts (arrow, ×100) in cortex of rats treated with TZP 1400 mg/kg IP once daily for 3 days, (d) tubular dilatation, tubular degeneration (arrows) and tubular basophilia (asterisk, ×100) in cortex of rats treated with VAN 150 mg/kg IV once daily for 3 days, and (e) tubular dilatation, tubular degeneration (arrows, ×100) in cortex of rats treated with VAN+TZP once daily for 3 days. This figure appears in colour in the online version of JAC and in black and white in the printed version of JAC.
Table 1.
Ordered logistic regression on Day 4 histopathology score when comparing with either saline or piperacillin/tazobactam
Comparison | Coefficient | P value | Lower 95% CI | Upper 95% CI |
---|---|---|---|---|
Groups compared with saline | ||||
TZP | 2.834 | 0.020 | 0.438 | 5.230 |
VAN | 4.993 | ≤0.001 | 2.287 | 7.699 |
VAN+TZP | 3.123 | 0.010 | 0.753 | 5.494 |
Groups compared with TZP | ||||
saline | −2.834 | 0.020 | −5.230 | −0.438 |
VAN | 2.159 | 0.044 | 0.061 | 4.257 |
VAN+TZP | 0.290 | 0.755 | −1.526 | 2.105 |
VAN, vancomycin; TZP, piperacillin/tazobactam.
NRK-52E cell experiments
In serum-deprived NRK-52E cells, treatment with vancomycin alone produced cell death (IC50 48.76 mg/mL, Figure 4a and b). Treatment with piperacillin/tazobactam and cefepime in the absence of vancomycin produced no cellular death (Figure 4a). When vancomycin was combined with fixed concentrations of piperacillin/tazobactam or cefepime, the degree of cellular death was not different compared with vancomycin alone (Figure 4b, P > 0.2). Gentamicin produced significant cellular death (IC50 11.29 mg/mL). However, when gentamicin was combined with vancomycin, the IC50 of 6.98 mg/mL did not differ compared with gentamicin alone (P > 0.2).
Figure 4.
Serum-deprived normal rat kidney NRK-52E epithelial cell viability after treatment with vancomycin (VAN), piperacillin/tazobactam (TZP), cefepime (FEP) and gentamicin (GEN) alone (a) or in combination with 1 mg/mL vancomycin (b) for 48 h using the alamarBlue® assay. The results presented were obtained from a single experiment in triplicate but are representative of experiments conducted under different conditions. Values are expressed as mean ± SD. RFUs, relative fluorescence units. This figure appears in colour in the online version of JAC and in black and white in the printed version of JAC.
Discussion
While a growing number of clinical studies have reported that vancomycin + piperacillin/tazobactam is associated with increased rates of AKI as measured by SCr, we demonstrated that VIKI from vancomycin + piperacillin/tazobactam is not worse than vancomycin alone. In fact, our rat data suggest that piperacillin/tazobactam may partially protect against VIKI. These data question the biological plausibility of vancomycin + piperacillin/tazobactam resulting in increased kidney toxicity. To the best of our knowledge, this is the first study investigating the effect of vancomycin + piperacillin/tazobactam on kidney injury using translational models focused on histopathology and biomarkers capable of discerning direct toxicity (as opposed to SCr as a surrogate).
Our experimental animal data are somewhat consistent with a previous animal study.15 In that study, vancomycin was administered at 25 mg/day IP for 2 days (which equates to ∼1000 mg/kg/day or an allometrically scaled dose of ∼80 mg/kg/day)26 and acute tubular necrosis was observed with granular material in the tubular lumen and cast formation. Human vancomycin doses are ∼60 mg/kg/day even at the upper end of the dosing range.36 This investigation also employed a very low dose of piperacillin/tazobactam at 100 mg/kg/day IP for 2 days (which is equivalent to 8.2 mg/kg/day human dose).26 A standard human dose for a 70 kg patient is ∼190 mg/kg/day.37 Thus, it was possible that high-dose vancomycin and a low dose of piperacillin/tazobactam was the reason for seeing identical toxicity between vancomycin and vancomycin + piperacillin/tazobactam in their study. In the present study, we allometrically scaled vancomycin and piperacillin/tazobactam to the human dose and did not observe an increase in nephrotoxicity with vancomycin + piperacillin/tazobactam compared with vancomycin alone, demonstrating that adding piperacillin/tazobactam did not worsen VIKI and may even be protective. This protective effect of piperacillin/tazobactam against vancomycin nephrotoxicity could be due to the sodium load associated with it, as observed with ticarcillin sodium.38,39
We demonstrated that vancomycin resulted in cell death in NRK-52E cells. The addition of piperacillin/tazobactam to vancomycin did not affect cell death. Similar results were obtained using other cell lines (HEK-293 and MDCK.2) under different culture conditions (data not shown). Notably, the IC50 of vancomycin from our study is higher than that reported by other groups in kidney cell lines,40–42 but we used clinical grade vancomycin in our cell studies. The pH of the final solution from clinical grade vancomycin should be less toxic to cells.43 This removes a variable that could cause cellular death by means other than drug toxicity. Regardless, we did obtain a dose–response toxicity curve with vancomycin alone.
Mechanistically, it is well established that VIKI is caused by oxidative stress14 and uromodulin interaction/cast formation.15 Piperacillin can rarely cause AIN.16 However, no clinical study has demonstrated that synergistic toxicity occurs because of these mechanisms. It is notable that piperacillin/tazobactam on its own is not generally a nephrotoxin. Prospective randomized controlled trial data support the idea that piperacillin/tazobactam alone is not overtly damaging to the kidney. For instance, when piperacillin/tazobactam was compared with meropenem/vaborbactam in 545 patients with urinary tract infections, piperacillin/tazobactam was reported to increase SCr at a rate of 0.4% versus 0% for meropenem/vaborbactam.44 In a separate trial comparing piperacillin/tazobactam versus meropenem, piperacillin/tazobactam only resulted in n = 1 non-fatal serious adverse event versus n = 0 for meropenem.45 At least some large retrospective studies have also failed to find differences in AKI rates (with SCr as a surrogate).46,47 Given our findings and these others, it remains possible that the reported increase in AKI with vancomycin + piperacillin/tazobactam from the clinical setting is a false positive from an imperfect surrogate of kidney injury (i.e. creatinine).
Clinical studies generally utilize the RIFLE or the AKIN definitions to classify AKI according to SCr.48 SCr is non-specific for kidney injury.12 However, in retrospective studies, SCr is often the only reliable variable to assess a patient’s kidney function. It is possible that competition for secretion or re-absorption can affect SCr with vancomycin + piperacillin/tazobactam treatment, while kidney function (as measured by glomerular filtration rate) remains unaffected.49,50 Yet in our rat study plasma creatinine followed similar trends but lagged 2 days behind urinary output changes and 1 day behind urinary biomarker changes. Because of the study design, we did not obtain blood at Day −1, and thus it is difficult to understand the full relationship between urinary biomarkers and plasma creatinine in our study. An interaction between vancomycin and piperacillin/tazobactam at the level of tubular secretion may not explain the late elevation of SCr for vancomycin + piperacillin/tazobactam. It may be that short-term piperacillin/tazobactam use is protective, but prolonged use triggers other mechanisms. Notably, we have only studied a total of 4 days of combined vancomycin + piperacillin/tazobactam, and longer study durations are necessary.
Novel biomarkers are qualified for use in preclinical animal19 and clinical human20 studies by the FDA to assess for drug-induced AKI, and by the EMA and the Japanese Pharmaceuticals and Medical Devices Agency for pre-clinical rodent studies.19 Urinary biomarker data from this study are interesting because: (i) KIM-1 and clusterin were recently demonstrated to be the most sensitive biomarkers for predicting VIKI defined by histopathology;51 and (ii) multiple samples over time facilitate temporal investigation of toxicity after renal insult. KIM-1 is highly sensitive and specific for proximal tubule damage, the locale of VIKI.21 KIM-1 is highly conserved between rats and humans, and thus the predictive capacity of rat KIM-1 for VIKI is compelling. A human homologue, KIM-1b, is structurally similar except for the cytoplasmic domain.52 Clusterin is present in kidney tubules, is anti-apoptotic and confers cell protection.53 As it is not filtered through the glomeruli, elevation should indicate tubular damage.53
Several limitations bear mention. These data are from a healthy rat model; however, the rat model is a well-accepted surrogate for human pathobiology.20 We have previously demonstrated that the pharmacokinetic/toxicodynamic index between this predictive rat model (i.e. AUC of 482 mg·h/L over 24 h)54 matches that seen in prospective human studies.55 Histopathology was elevated for all three groups compared with saline; however, only vancomycin was elevated compared with piperacillin/tazobactam. This may reflect the inherent subjectivity of histopathology as the control group was identified to the pathologist (per standard practice56); however, the pathologist was blinded to treatments received. Urinary biomarkers may be less biased. We have not yet conducted drug assays, though these experiments are planned. Finally, while we saw signal for KIM-1 and clusterin in our study, OPN did not differ, but it is also not the best biomarker for VIKI51 or proximal tubule necrosis.57
In conclusion, vancomycin + piperacillin/tazobactam did not cause more kidney injury than vancomycin alone as evidenced by a translational rat model measuring plasma creatinine, urinary output, urinary biomarkers and histopathology. Cellular studies supported these conclusions. Novel urinary biomarkers may aid in determining whether higher rates of kidney injury with vancomycin + piperacillin/tazobactam are realized in clinical studies.
Supplementary Material
Funding
This work was supported by internal funding from Midwestern University to the Pharmacometrics Center of Excellence and via equipment usage at the Midwestern University CORE center.
Transparency declarations
K.J.D. has received research support from Merck & Co., Inc. and Pfizer, Inc. M.H.S. reports having received a research grant from Nevakar, Inc. unrelated to the current work. All other authors: none to declare.
References
- 1.Kelesidis T, Braykov N, Uslan DZ et al. Indications and types of antibiotic agents used in 6 acute care hospitals, 2009-2010: a pragmatic retrospective observational study. Infect Control Hosp Epidemiol 2016; 37: 70–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Luther MK, Timbrook TT, Caffrey AR et al. Vancomycin plus piperacillin-tazobactam and acute kidney injury in adults: a systematic review and meta-analysis. Crit Care Med 2018; 46: 12–20. [DOI] [PubMed] [Google Scholar]
- 3.Beckman EJ. Piperacillin-tazobactam plus vancomycin equals acute kidney injury: does it add up? Pediatr Crit Care Med 2017; 18: 1183–4. [DOI] [PubMed] [Google Scholar]
- 4.Watkins RR, Deresinski S. Increasing evidence of the nephrotoxicity of piperacillin/tazobactam and vancomycin combination therapy – what is the clinician to do? Clin Infect Dis 2017; 65: 2137–43. [DOI] [PubMed] [Google Scholar]
- 5.Rhodes NJ, Kuti JL, Nicolau DP et al. An exploratory analysis of the ability of a cefepime trough concentration greater than 22 mg/L to predict neurotoxicity. J Infect Chemother 2016; 22: 78–83. [DOI] [PubMed] [Google Scholar]
- 6.FDA warns about increased risk of ruptures or tears in the aorta blood vessel with fluoroquinolone antibiotics in certain patients. US Food and Drug Administration. Safety Warning. 2018. https://www.fda.gov/Drugs/DrugSafety/ucm628753.htm.
- 7.McLaughlin M, Advincula MR, Malczynski M et al. Correlations of antibiotic use and carbapenem resistance in Enterobacteriaceae. Antimicrob Agents Chemother 2013; 57: 5131–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Mingeot-Leclercq MP, Tulkens PM. Aminoglycosides: nephrotoxicity. Antimicrob Agents Chemother 1999; 43: 1003–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bagshaw SM, George C, Dinu I et al. A multi-centre evaluation of the RIFLE criteria for early acute kidney injury in critically ill patients. Nephrol Dial Transplant 2008; 23: 1203–10. [DOI] [PubMed] [Google Scholar]
- 10.Bagshaw SM, Lapinsky S, Dial S et al. Acute kidney injury in septic shock: clinical outcomes and impact of duration of hypotension prior to initiation of antimicrobial therapy. Intensive Care Med 2009; 35: 871–81. [DOI] [PubMed] [Google Scholar]
- 11.Avedissian SN, Pais GM, Liu J et al. Piperacillin-tazobactam added to vancomycin increases risk for AKI: fact or fiction? Clin Infect Dis 2019; doi:10.1093/cid/ciz1189. [DOI] [PubMed] [Google Scholar]
- 12.Waikar SS, Betensky RA, Emerson SC et al. Imperfect gold standards for kidney injury biomarker evaluation. J Am Soc Nephrol 2012; 23: 13–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Berglund F, Killander J, Pompeius R. Effect of trimethoprim-sulfamethoxazole on the renal excretion of creatinine in man. J Urol 1975; 114: 802–8. [DOI] [PubMed] [Google Scholar]
- 14.Bamgbola O. Review of vancomycin-induced renal toxicity: an update. Ther Adv Endocrinol Metab 2016; 7: 136–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Luque Y, Louis K, Jouanneau C et al. Vancomycin-associated cast nephropathy. J Am Soc Nephrol 2017; 28: 1723–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Soto J, Bosch JM, Alsar Ortiz MJ et al. Piperacillin-induced acute interstitial nephritis. Nephron 1993; 65: 154–5. [DOI] [PubMed] [Google Scholar]
- 17.Zager RA. Exogenous creatinine clearance accurately assesses filtration failure in rat experimental nephropathies. Am J Kidney Dis 1987; 10: 427–30. [DOI] [PubMed] [Google Scholar]
- 18.Darling IM, Morris ME. Evaluation of ‘true’ creatinine clearance in rats reveals extensive renal secretion. Pharm Res 1991; 8: 1318–22. [DOI] [PubMed] [Google Scholar]
- 19.Critical Path Institute. Predictive Safety Testing Consortium. https://c-path.org/programs/pstc/.
- 20.US Food and Drug Administration. Critical Path Institute's Predictive Safety Testing Consortium Nephrotoxicity Working Group (CPATH PSTC-NWG), and Foundation for the National Institutes of Health’s Biomarker Consortium Kidney Safety Biomarker Project Team (FNIH BC-KSP). Safety biomarker panel to aid in the detection of kidney tubular injury in phase 1 trials in healthy volunteers. 2018. https://www.fda.gov/drugs/cder-biomarker-qualification-program/biomarker-qualification-submissions.
- 21.O'Donnell JN, Rhodes NJ, Lodise TP et al. 24-hour pharmacokinetic relationships for vancomycin and novel urinary biomarkers of acute kidney injury. Antimicrob Agents Chemother 2017; 61: e00416-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Rhodes NJ, Prozialeck WC, Lodise TP et al. Evaluation of vancomycin exposures associated with elevations in novel urinary biomarkers of acute kidney injury in vancomycin-treated rats. Antimicrob Agents Chemother 2016; 60: 5742–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.National Research Council (US) Committee for the Update of the Guide for the Care and Use of Laboratory Animals. Guide for the Care and Use of Laboratory Animals. 8th edn. Washington (DC: ): National Academies Press (US; ); 2011. doi:10.17226/12910. https://www.ncbi.nlm.nih.gov/books/NBK54050/. [PubMed] [Google Scholar]
- 24.Fuchs TC, Frick K, Emde B et al. Evaluation of novel acute urinary rat kidney toxicity biomarker for subacute toxicity studies in preclinical trials. Toxicol Pathol 2012; 40: 1031–48. [DOI] [PubMed] [Google Scholar]
- 25.Vaidya VS, Ozer JS, Dieterle F et al. Kidney injury molecule-1 outperforms traditional biomarkers of kidney injury in preclinical biomarker qualification studies. Nat Biotechnol 2010; 28: 478–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.US Food and Drug Administration. Guidance for Industry: Estimating the Maximum Safe Starting Dose in Initial Clinical Trials for Therapeutics in Adult Healthy Volunteers. 2005. https://www.fda.gov/media/72309/download.
- 27.Ou M, Song Y, Li S et al. LC-MS/MS method for serum creatinine: comparison with enzymatic method and Jaffe method. PLoS One 2015; 10: e0133912.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Prozialeck WC, Edwards JR, Lamar PC et al. Expression of kidney injury molecule-1 (Kim-1) in relation to necrosis and apoptosis during the early stages of Cd-induced proximal tubule injury. Toxicol Appl Pharmacol 2009; 238: 306–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Prozialeck WC, Edwards JR, Vaidya VS et al. Preclinical evaluation of novel urinary biomarkers of cadmium nephrotoxicity. Toxicol Appl Pharmacol 2009; 238: 301–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Sistare FD, Dieterle F, Troth S et al. Towards consensus practices to qualify safety biomarkers for use in early drug development. Nat Biotechnol 2010; 28: 446–54. [DOI] [PubMed] [Google Scholar]
- 31.de Larco JE, Todaro GJ. Epithelioid and fibroblastic rat kidney cell clones: epidermal growth factor (EGF) receptors and the effect of mouse sarcoma virus transformation. J Cell Physiol 1978; 94: 335–42. [DOI] [PubMed] [Google Scholar]
- 32.Rybak MJ, Abate BJ, Kang SL et al. Prospective evaluation of the effect of an aminoglycoside dosing regimen on rates of observed nephrotoxicity and ototoxicity. Antimicrob Agents Chemother 1999; 43: 1549–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.R Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing, 2018. https://www.R-project.org/. [Google Scholar]
- 34.Wickham H. ggplot2: Elegant Graphics for Data Analysis. New York: Springer, 2016. [Google Scholar]
- 35.UCLA Institute for Digital Research and Education. Repeated measures analysis with STATA. https://stats.idre.ucla.edu/stata/seminars/repeated-measures-analysis-with-stata/.
- 36.Rybak M, Lomaestro B, Rotschafer JC et al. Therapeutic monitoring of vancomycin in adult patients: a consensus review of the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, and the Society of Infectious Diseases Pharmacists. Am J Health Syst Pharm 2009; 66: 82–98. [DOI] [PubMed] [Google Scholar]
- 37.Zosyn Package Insert. Pfizer. 2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/050684s88s89s90_050750s37s38s39lbl.pdf.
- 38.Ohnishi A, Bryant TD, Branch KR et al. Role of sodium in the protective effect of ticarcillin on gentamicin nephrotoxicity in rats. Antimicrob Agents Chemother 1989; 33: 928–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Sabra R, Branch RA. Role of sodium in protection by extended-spectrum penicillins against tobramycin-induced nephrotoxicity. Antimicrob Agents Chemother 1990; 34: 1020–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Arimura Y, Yano T, Hirano M et al. Mitochondrial superoxide production contributes to vancomycin-induced renal tubular cell apoptosis. Free Radic Biol Med 2012; 52: 1865–73. [DOI] [PubMed] [Google Scholar]
- 41.Humanes B, Jado JC, Camano S et al. Protective effects of cilastatin against vancomycin-induced nephrotoxicity. Biomed Res Int 2015; 2015: 704382.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Im DS, Shin HJ, Yang KJ et al. Cilastatin attenuates vancomycin-induced nephrotoxicity via P-glycoprotein. Toxicol Lett 2017; 277: 9–17. [DOI] [PubMed] [Google Scholar]
- 43.Hospira. Vancomycin Hydrochloride for Injection, USP Prescribing Information. 2014. https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/062911s035lbl.pdf.
- 44.Kaye KS, Bhowmick T, Metallidis S et al. Effect of meropenem-vaborbactam vs piperacillin-tazobactam on clinical cure or improvement and microbial eradication in complicated urinary tract infection: the TANGO I randomized clinical trial. JAMA 2018; 319: 788–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Harris PNA, Tambyah PA, Lye DC et al. Effect of piperacillin-tazobactam vs meropenem on 30-day mortality for patients with E. coli or Klebsiella pneumoniae bloodstream infection and ceftriaxone resistance: a randomized clinical trial. JAMA 2018; 320: 984–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Davies SW, Efird JT, Guidry CA et al. Top Guns: the “Maverick” and “Goose” of empiric therapy. Surg Infect (Larchmt) 2016; 17: 38–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Schreier DJ, Kashani KB, Sakhuja A et al. Incidence of acute kidney injury among critically ill patients with brief empiric use of anti-pseudomonal β-lactams with vancomycin. Clin Infect Dis 2018; 68: 1456–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lopes JA, Jorge S. The RIFLE and AKIN classifications for acute kidney injury: a critical and comprehensive review. Clin Kidney J 2013; 6: 8–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Schaub JA, Parikh CR. Biomarkers of acute kidney injury and associations with short- and long-term outcomes. F1000Res 2016; 5: F1000 Faculty Rev-986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Farkas J. PULMCrit Blog. https://emcrit.org/pulmcrit/piperacillin-tazobactam-nephrotoxic/.
- 51.Pais GM, Avedissian SN, O'Donnell JN et al. Comparative performance of urinary biomarkers for vancomycin-induced kidney injury according to timeline of injury. Antimicrob Agents Chemother 2019; 63: e00079-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Moresco RN, Bochi GV, Stein CS et al. Urinary kidney injury molecule-1 in renal disease. Clin Chim Acta 2018; 487: 15–21. [DOI] [PubMed] [Google Scholar]
- 53.Griffin BR, Faubel S, Edelstein CL. Biomarkers of drug-induced kidney toxicity. Ther Drug Monit 2019; 41: 213–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Avedissian SN, Pais GM, O'Donnell JN et al. Twenty-four hour pharmacokinetic relationships for intravenous vancomycin and novel urinary biomarkers of acute kidney injury in a rat model. J Antimicrob Chemother 2019; 74: 2326–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lodise TP, Rosenkranz SL, Finnemeyer M et al. The Emperor's New Clothes: prospective observational evaluation of the association between initial vancomycin exposure and failure rates among adult hospitalized patients with MRSA bloodstream infections (PROVIDE). Clin Infect Dis 2019; doi:10.1093/cid/ciz460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Burkhardt JE, Ennulat D, Pandher K et al. Topic of histopathology blinding in nonclinical safety biomarker qualification studies. Toxicol Pathol 2010; 38: 666–7. [DOI] [PubMed] [Google Scholar]
- 57.Bonventre JV, Vaidya VS, Schmouder R et al. Next-generation biomarkers for detecting kidney toxicity. Nat Biotechnol 2010; 28: 436–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.