Next Article in Journal
The Impact of National Activities on Antibiotic Consumption in Hospitals and Different Departments over a 14-Year Period
Previous Article in Journal
Genetic Characterization and Population Structure of Drug-Resistant Mycobacterium tuberculosis Isolated from Brazilian Patients Using Whole-Genome Sequencing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Is It Still Beneficial to Monitor the Trough Concentration of Vancomycin? A Quantitative Meta-Analysis of Nephrotoxicity and Efficacy

1
Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai 200040, China
2
Key Laboratory of Clinical Pharmacology of Antibiotics, National Population and Family Planning Commission, Shanghai 200040, China
3
National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
4
Phase I Clinical Research Center, Huashan Hospital, Fudan University, Shanghai 200040, China
*
Authors to whom correspondence should be addressed.
Antibiotics 2024, 13(6), 497; https://doi.org/10.3390/antibiotics13060497
Submission received: 24 April 2024 / Revised: 15 May 2024 / Accepted: 20 May 2024 / Published: 28 May 2024
(This article belongs to the Section Pharmacokinetics and Pharmacodynamics of Drugs)

Abstract

:
This study conducted a quantitative meta-analysis to investigate the association of vancomycin indicators, particularly area under the curve over 24 h (AUC24) and trough concentrations (Ctrough), and their relationship with both nephrotoxicity and efficacy. Literature research was performed in PubMed and Web of Science on vancomycin nephrotoxicity and efficacy in adult inpatients. Vancomycin Ctrough, AUC24, AUC24/minimum inhibitory concentration (MIC), nephrotoxicity evaluation and treatment outcomes were extracted. Logistic regression and Emax models were conducted, stratified by evaluation criterion for nephrotoxicity and primary outcomes for efficacy. Among 100 publications on nephrotoxicity, 29 focused on AUC24 and 97 on Ctrough, while of 74 publications on efficacy, 27 reported AUC24/MIC and 68 reported Ctrough. The logistic regression analysis indicated a significant association between nephrotoxicity and vancomycin Ctrough (odds ratio = 2.193; 95% CI 1.582–3.442, p < 0.001). The receiver operating characteristic curve had an area of 0.90, with a cut-off point of 14.55 mg/L. Additionally, 92.3% of the groups with a mean AUC24 within 400–600 mg·h/L showed a mean Ctrough of 10–20 mg/L. However, a subtle, non-statistically significant association was observed between the AUC24 and nephrotoxicity, as well as between AUC24/MIC and Ctrough concerning treatment outcomes. Our findings suggest that monitoring vancomycin Ctrough remains a beneficial and valuable approach to proactively identifying patients at risk of nephrotoxicity, particularly when Ctrough exceeds 15 mg/L. Ctrough can serve as a surrogate for AUC24 to some extent. However, no definitive cut-off values were identified for AUC24 concerning nephrotoxicity or for Ctrough and AUC24/MIC regarding efficacy.

1. Introduction

Vancomycin is the first-line antibiotic for methicillin resistant Staphylococcus aureus (MRSA) infections [1], and is also used to treat suspected or confirmed infections caused by other Gram-positive bacteria. However, a narrow therapeutic index, which requires balancing efficacy with the risk of acute kidney injury (AKI), and large inter-patient variability in pharmacokinetics (PK) makes vancomycin dosing even more challenging, thus necessitating the use of therapeutic drug monitoring (TDM).
Despite being in clinical use for over 60 years, there is still controversy regarding the most appropriate indicator and its respective target value to optimize vancomycin treatment and reduce toxicity. During the past few years, the ratio of area under the curve to minimum inhibitory concentration over 24 h (AUC24/MIC) has been advocated as the preferred parameter for measuring vancomycin’s effectiveness [1,2,3]. Due to difficulty in determining the AUC24 in routine clinical practice and subsequently calculating the AUC24/MIC, the 2009 American guideline suggested the trough concentration (Ctrough) as a surrogate marker for AUC24, which is recommended as the most accurate and practical method to monitor vancomycin [4]. However, with the development of new approaches, such as Bayesian software, estimating AUC24 has become more convenient. Some studies reported AUC24-guided dosing as more clinically effective [5,6,7] and having less risk of AKI over Ctrough-guide dosing [5,6,7,8,9,10,11]. Furthermore, Ctrough was reported as not being substituted for AUC24 in some studies [12,13,14,15,16]. Thus, the 2020 American TDM guideline for vancomycin and the 2022 Japanese TDM guideline recommended AUC24/MIC as a reliable predictor to improve clinical efficacy and avoid nephrotoxicity, targeting a ratio of 400–600 [17,18].
However, some research still reported inconsistent results. Dalton et al. reported that a target AUC24/MIC index could not be established to achieve the optimal effectiveness and safety of vancomycin [19]. Bellos et al. discovered that an increase in Ctrough was significantly associated with a higher risk of nephrotoxicity [20]. Lodise et al. found that vancomycin Ctrough is the pharmacodynamic index that best describes the exposure–toxicity response relationship [21]. Moreover, recent studies provide growing evidence that Ctrough is more strongly correlated with nephrotoxicity [22]. Meanwhile, resource-constrained settings that face challenges in estimating the AUC24 using a Bayesian approach or a first-order PK equation with two concentrations of steady-state samples remain prevalent, especially in developing countries [23]. Therefore, in some countries, not only is AUC24 still recommended, but Ctrough is as well. For instance, the 2020 Chinese guideline suggests maintaining steady-state Ctrough at 10–15 mg/L in adult patients and 10–20 mg/L in adult patients with serious MRSA infections [24]. The Anti-infectives Committee of the International Association of Therapeutic Drug Monitoring and Clinical Toxicology also recommends a target Ctrough of 10–15 mg/L for serious MRSA infections in 2022 [25]. The European Society of Intensive Care Medicine recommended Ctrough at 15–20 mg/L for severe infections in 2020 [26].
Although numerous reviews have examined a large number of clinical studies and proposed target values for the efficacy and nephrotoxicity of vancomycin, providing valuable clinical references, inconsistencies have persisted in previous meta-analysis. Meanwhile, traditional meta-analysis often faces significant heterogeneity among studies, encompassing differences in patient characteristics, definition of nephrotoxicity, and treatment outcomes. This can make it challenging to accurately assess the relationships and target values for vancomycin efficacy and nephrotoxicity.
Therefore, the present study employed a quantitative meta-analysis to investigate the relationship between vancomycin parameters (Ctrough and AUC24 or AUC24/MIC) and both nephrotoxicity and efficacy, taking into account the varying definition of nephrotoxicity and treatment outcomes. This study aimed to evaluate the benefits of monitoring the Ctrough and AUC24 of vancomycin, and to assess the relationship between AUC24 and Ctrough in studies that included both measures.

2. Results

2.1. Characteristics of the Included Studies

A total of 172 from 1420 studies were subjected to further examination, and finally 100 articles (listed in the Supplementary Materials) were included in the nephrotoxicity analysis. Among them, 29 and 97 articles were included for target AUC24 and target Ctrough evaluation, respectively (Figure 1A). Most of the studies adopted a retrospective design (n = 88), while 13 studies were prospective cohorts and 1 study conducted a post hoc analysis of randomized controlled trials (RCT). The 2009 consensus guideline [4] was the most commonly adopted criteria to define vancomycin nephrotoxicity (n = 65), while the Kidney Disease Improving Global Outcomes (KDIGO) [27], Acute Kidney Injury Network (AKIN) [28], and Risk, Injury, Failure, Loss of kidney function and End-stage kidney disease (RIFLE) [29] criteria were applied in 16, 14, and 16 studies, respectively.
A total of 115 from 4434 articles underwent detailed scrutiny for efficacy analysis. Finally, 74 articles (listed in the Supplementary Materials) were screened for inclusion in the efficacy analysis, of which 27 and 68 articles had the assessment of target AUC24/MIC and target Ctrough, respectively (Figure 1B). Out of the 74 studies, 61 adopted a retrospective design. The most frequently reported outcome was all-cause mortality (n = 55), followed by clinical failure (n = 27), microbiological failure (n = 26) and treatment failure (n = 25). The most commonly used methods for MIC testing were broth microdilution (BMD) (n = 16) and the Etest (n = 17) method, while agar dilution, VITEK 2 (https://www.biomerieux-usa.com/vitek-2, accessed on 24 April 2024) and MicroScan (https://www.beckmancoulter.com/products/microbiology/microscan-walkaway-plus-system, accessed on 24 April 2024) were used less frequently (n < 7).
Since 2012, the number of publications has significantly increased (ranging from 1990 to 2022), with the majority of results reported from United States, Japan and China. Further details on the study characteristics are provided in Supplementary Table S1 to Table S4.

2.2. Nephrotoxicity

The non-linear association between vancomycin Ctrough and the incidence of nephrotoxicity, stratified by different nephrotoxicity definition, is illustrated in Figure 2 and Table 1. The data indicate that a higher Ctrough of vancomycin are associated with a higher incidence of nephrotoxicity, with a more obvious positive correlation observed for the KDIGO and RIFLE criteria. This relationship is also evident in the box plot of the probability of nephrotoxicity in different trough categories (Figure S1).
The univariate logistic regression analysis (see Figure 3 for the 2009 consensus guidelines criteria) revealed that nephrotoxicity was significantly associated with vancomycin Ctrough (OR (95%CI) = 2.193 (1.582–3.442), p < 0.001). The area under the receiver operating characteristic (ROC) curve (AUROC) value of 0.90 indicated the potential of vancomycin Ctrough to serve as a predictor of vancomycin nephrotoxicity (Figure S2), with a cut-off of 14.55 mg/L, representing 79.4% sensitivity and 91.2% specificity in the study populations. Covariates with missing values less than 30% (i.e., age, serum creatinine and male percentage) were also evaluated for their association with nephrotoxicity, but none demonstrated a statistically significant relationship. A subgroup analysis of patients not receiving renal replacement therapy showed similar results (Figure S3).
A slight trend towards lower nephrotoxicity in patients with low AUC24 was observed in Figure S4. Logistic regression analysis examining the association between AUC24 and nephrotoxicity according to the 2009 consensus guideline, using data from eight articles, revealed a similar trend with a cut-off value of 510 mg·h/L, although the trend was not significant (OR = 1.008, 95%CI of 1.001–1.02, p > 0.05, Figure 4).

2.3. Efficacy

2.3.1. Treatment Failure

Eleven studies reported treatment failure as an outcome, with ten articles presenting results with BMD method and six using the Etest method. A subtle trend suggesting that a higher AUC24/MIC is associated with a lower treatment failure rate was observed (Figure 5, Table 1). When AUC24/MICBMD reached 400 or 600, the predicted treatment success rate was 52% and 62%, respectively. Similarly, when AUC24/MICEtest attained 400 or 600, the predicted treatment success rate was 59% and 80%. However, no statistically significant difference (OR = 1.017, 95% CI of 0.999–1.051, p > 0.05, Figure S5) in treatment success rates was identified across the range of AUC24/MICBMD in the logistic regression analysis. A similar trend was observed for the relationship between treatment failure rates and Ctrough (Figure S6).

2.3.2. All-Cause Mortality

Out of 18 studies that reported an all-cause mortality outcome, 12 studies reported the 30- or 28-day all-cause mortality, of which 8 articles reported results with BMD and the Etest method, respectively. Due to the limited sample size of studies reporting a binary outcome, only incidence was analyzed. A subtle trend emerged, suggesting a correlation between higher AUC24/MIC and lower 30-day all-cause mortality rates (Figure 5, Table 1). When the AUC24/MICBMD reached 400 or 600, the predicted survival rate was 76% and 83%, respectively. Similarly, when the AUC24/MICEtest reached 400 or 600, the predicted survival rate was 78% and 84%, respectively. A similar trend was observed for the relationship between 30-day all-cause mortality rate and Ctrough (Figure S6).

2.3.3. Microbiologic Failure

Of 17 studies reporting microbiologic failure, 9 articles utilized BMD method and the Etest method, respectively. Microbiologic failure appeared to be lower in patients with higher AUC24/MIC and Ctrough (Figure 5, Table 1 and Figure S6). The AUC24/MICBMD and AUC24/MICEtest equal to 400 resulted in a microbiologic success rate of 63% and 77%, respectively.
Due to the limited number of articles, it is not possible to analyze clinical failure outcomes. In the subgroup of patients with MRSA infections, the trends observed in the above analysis were similar (results not shown). This suggests that the relationship between AUC24/MIC and clinical outcomes may be consistent in this patient population.

2.4. Relationship of Vancomycin Mean AUC24 and Ctrough

The mean Ctrough values were categorized into the following groups: ≤10 mg/L, 10–15 mg/L, 15–20 mg/L and >20 mg/L. Similarly, the mean AUC24 were divided into ≤200 mg·h/L, 200–400 mg·h/L, 400–600 mg·h/L and >600 mg·h/L. The chord diagram vividly demonstrates the relationship between the mean Ctrough and mean AUC24 for each subgroup of studies (Figure 6, Table 2).
Among the paired groups (n = 61) from studies included in the nephrotoxicity analysis, 77.8% of the groups with mean Ctrough ≤ 10 mg/L had mean AUC24 < 400 mg·h/L, while all groups with mean Ctrough > 20 mg/L (n = 5) had AUC24 > 400 mg·h/L, among which 60% had AUC24 values > 600 mg·h/L. In the subgroups with mean AUC24 within 400–600 mg·h/L, 92.3% had mean Ctrough of 10–20 mg/L.
Of the paired groups (n = 83) from studies included in the efficacy analysis with both exposure measures, 67.9% of the groups with mean Ctrough ≤ 10 mg/L had mean AUC24 < 400 mg·h/L, while all groups with Ctrough > 20 mg/L (n = 3) had AUC24 > 400 mg·h/L. When Ctrough reached 10–15 mg/L, the rate of vancomycin AUC24 in 400–600 mg·h/L was 84.4%, and when Ctrough reached 15–20 mg/L, the rate of vancomycin AUC24 in 400–600 mg·h/L was 71.4%. In other words, 79.7% of the subgroups with mean AUC24 within 400–600 mg·h/L had mean Ctrough of 10–20 mg/L.

3. Discussion

The recent American and Japanese TDM guidelines recommend AUC24/MIC as the preferred approach for enhancing vancomycin efficacy and educing nephrotoxicity, and Ctrough is no longer recommended [17,18]. Nevertheless, obtaining timely and accurate AUC24 poses challenges, and measuring Ctrough remains the most efficient and accessible method to monitor vancomycin dosing, especially in source-limited settings. Therefore, the question of whether Ctrough monitoring remains beneficial warrants further investigation.
Our quantitative meta-analysis demonstrated a robust correlation between vancomycin Ctrough and nephrotoxicity (OR (95%CI) = 2.193 (1.582–3.442), p < 0.001), with a cut-off point identified at 14.55 mg/L. This finding aligns with the recommendations of a clinical guideline [24] and a position statement [25]. Moreover, our analysis revealed that studies with a mean Ctrough of 10–15 mg/L and 15–20 mg/L showed that almost 80% had a mean AUC24 400–600 mg·h/L, suggesting that Ctrough can serve as a surrogate for AUC24 to some degree. This is further supported by a recent multicenter, retrospective study in China that focused on critically ill patients without any form of dialysis [23]. Our study supports the clinical utility of Ctrough monitoring, particularly for nephrotoxicity prevention, as it correlates with AUC24 within specific ranges. However, the direct substitution of Ctrough for AUC24 is not always feasible due to individual pharmacokinetic variations. Developing an AUC24-Ctrough equation could establish patient-specific Ctrough targets for individualized management.
In addition, our study revealed that the application of KDIGO and RIFLE criteria for assessing kidney toxicity yielded higher sensitivity in identifying vancomycin-associated nephrotoxicity compared to the 2009 consensus guideline. This disparity in sensitivity could be attributed to the slightly higher threshold defined in the 2009 guideline (an increase in the serum creatinine ≥0.5 mg/dL [4]), which has been updated in the latest guideline [17].
Unfortunately, although we observed a subtle trend that higher Ctrough were associated with lower treatment or microbiologic failure rates and 30-day all-cause mortality rates, no definitive cut-off value was identified. This lack of specificity can be attributed to the fact that the majority of Ctrough falls within the common therapeutic range of 10–20 mg/L for vancomycin. However, considering the observed efficacy outcome (Figure S6) and the simulation using the Emax model, it appears that a Ctrough greater than 15 mg/L nearly reaches the plateau of the efficacy curve.
Furthermore, we evaluated the relationship between AUC24 and vancomycin nephrotoxicity and the relation between AUC24/MIC and efficacy outcome. While a subtle trend of reduced nephrotoxicity in patients with lower AUC24 was observed, it did not reach statistical significance. This could be attributed to the fact that only eight included studies with a mean AUC24 were centered on the range of 400–600 mg·h/L. However, we attempted to identify a cut-off value at 510 mg·h/L. Concerning the correlation between AUC24/MIC and the efficacy outcome, our analysis revealed that when the AUC24/MICEtest and AUC24/MICBMD exceeded 500–600, both the treatment/microbiologic success rate and 30-day survival rates appeared to approach the efficacy curve plateau. Nonetheless, these findings should be interpreted with caution given the narrow range of AUC24/MIC values obtained from clinical settings employing TDM. In summary, AUC24 not exceeding 500 mg·h/L (assuming the MIC as 1 mg/L) may favor both clinical efficacy and nephrotoxicity avoidance.
There are some limitations for the selected studies. The most common limitation the selected studies mentioned is the retrospective nature of the study design, which is also one of limitations of our analysis, i.e., the majority of the included studies (greater than 80%) were retrospective, which introduces a risk of unmeasured confounding effects and bias. In addition, in the selected studies, the small sample size of the study, a single center being included in most studies, and the fact that a limited type of patient population hinders extrapolation to a wider range of people were also mentioned. However, what we conducted was a quantitative meta-analysis including all the patient population data for analysis, which addressed the concern of the small sample size, single center and single type patient population in each study. Furthermore, some other limitations for our analysis needed to be considered when interpreting the results. Firstly, articles reported different detection methods for vancomycin concentration (most are commercial immunoassays), along with various types of Ctrough measurements, including initial or first steady-state values, average, highest or predicted values used in each article, which may introduce bias. Additionally, among different studies, the severity of the disease and the physiological and pathological condition of the patients vary, and the limited data availability hindered the evaluation of covariates on nephrotoxicity or efficacy, such as co-administered medication, renal function (creatinine clearance rate and renal replacement therapy), and critically ill patients’ percentage. Finally, most studies published after 2009 focused on collecting data within the recommended range of vancomycin Ctrough and AUC24 due to the widespread use of TDM. The narrow range of data might obscure the relationship for the two indictors, making it challenging to draw definitive conclusions.

4. Methods

4.1. Search Strategy

The literature search was performed using the PubMed and Web of Science database. The search keywords for the analysis of association between exposure and nephrotoxicity included “vancomycin”, its exposure parameters (“area under the concentration-time curve”, “trough concentration”, “exposure”, “pharmacokinetics” and “pharmacokinetics /pharmacodynamics”) and safety related indicators (“nephrotoxicity”, “acute kidney injury”, “renal failure”, “renal impairment”).
Similarly, for the analysis of association between exposure and efficacy, the search keywords for vancomycin-related ones included the above mentioned keywords and also “area under the concentration-time curve to minimum inhibitory concentration ratio”, while efficacy related indicators included “efficacy”, “clinical outcome”, “clinical failure”, “clinical response”, “microbiological failure”, “treatment failure”, “success”, “mortality” and “eradication”.
The reference lists of the included studies and historical systematic reviews were searched using a snowball method to identify potential additional sources. No language or date restrictions were imposed, but the patients were limited to adults.

4.2. Inclusion Criteria and Outcomes

We included adult inpatients treated with intravenous vancomycin and studies from RCT, as well as prospective and retrospective studies that met the searching criteria.
The inclusion criteria for the analysis of the association between vancomycin exposure (AUC24 and Ctrough) and nephrotoxicity included studies reporting AUC24 or/and Ctrough, along with detailed definitions of nephrotoxicity events. The primary outcome was the incidence of nephrotoxicity. Likewise, for the association between a vancomycin indicator (AUC24/MIC or Ctrough) and efficacy, the inclusion criteria included studies reporting AUC24/MIC or/and Ctrough and respective outcomes, i.e., treatment failure, all-cause mortality, microbiologic failure, or clinical failure. The primary outcome was treatment failure and 30- or 28-day all-cause mortality. Secondary outcomes were microbiologic failure and clinical failure. Treatment failure was defined as any combination of death, clinical non-improvement or worsening, need for antibiotic modification, microbiologic failure or recurrence of bacteremia. No specific patient populations or infections were excluded.

4.3. Data Extraction

The analysis of nephrotoxicity in the extraction of data comprised the following information: characteristics of the literature (year of publication, name of first author, region or country of study); study design (trial type, eligibility criteria, patient population, and sample size); study outcomes of vancomycin exposure (measurement of AUC24 or Ctrough, method of AUC24 calculation, timing of AUC24 calculation and Ctrough collection relative to start of therapy) and nephrotoxicity (continuous (incidence rate) and/or binary (yes vs. no) nephrotoxicity outcome) per different evaluation criterion like the 2009 vancomycin consensus [4], KDIGO [27], AKIN [28] or RIFLE [29] guidelines), and patient characteristics (age, weight, renal function, proportion of male patients, coadministration of nephrotoxins and critically ill /intensive care unit status).
For the efficacy analysis, the data extraction involved literature characteristics, the study design as mentioned above, study outcomes of exposure parameters (measurement of AUC24/MIC or Ctrough, method of AUC24 calculation, timing of AUC24 calculation and Ctrough collection relative to start of therapy, method of MIC determination) and efficacy (the continuous and/or binary clinical outcome measures), and patient characteristics (age, weight, renal function, proportion of male patients and critically ill/intensive care unit status).
Data extraction was conducted independently by two authors who applied the inclusion criteria. In case of any disagreements, alignment was achieved through consensus.

4.4. Data Handling

Two types of outcomes (all-cause mortality, treatment failure, microbiologic failure, clinical failure and nephrotoxicity) were collected, i.e., proportion (incidence rate) and binary variables (yes or no). The mean values of AUC24/MIC, AUC24 and Ctrough were extracted and treated as continuous variables, except for the articles that only reported median values, which were used instead.
The nephrotoxicity outcome was analyzed, stratified by the evaluation criterion (i.e., 2009 vancomycin consensus, KDIGO, AKIN and RIFLE guidelines). A subgroup analysis of patients in the intensive care unit and without receipt of dialysis was also performed separately, provided that enough studies (>5) were available.
The efficacy outcomes, including all-cause mortality, treatment failure, microbiologic failure and clinical failure, were analyzed separately. To account for potential variations in MIC results due to the use of different MIC testing methods, the analysis was stratified by the MIC testing method. Due to the small sample size of articles reporting MIC testing methods of agar dilution, VITEK 2 and MicroScan, only articles reporting BMD and the Etest method were included. Additionally, a subgroup analysis of only MRSA-infected individuals was performed separately.

4.5. Analytical Method

An exploratory analysis revealed a trend of gradually increasing outcome proportions along with increasing vancomycin indicators, reaching a plateau at higher levels. The distributional characteristics of these data were described by the Emax model (Equation (1)). A fit-for-purpose simulation using the typical values of parameters from the Emax model was conducted to obtain the incidence of outcome at a certain value of Ctrough or AUC24:
E = E m a x · C γ E C 50 γ + C γ
where Emax represents the maximum effect, while EC50 represents the indicators required to achieve half the Emax. The slope factor (also known as Hill factor), represented by γ, measures the sensitivity of the response to the indicator’s change, determining the steepness of the curve.
Binary outcomes were analyzed using a logistic regression model (Equation (2)). A univariate logistic regression analysis was first performed to assess the relationship between the vancomycin indicators (as a continuous variable) and the outcomes. Optimal cut-off points were derived from the ROC curves using Youden’s index [30]. The study employed a univariate logistic regression analysis to evaluate patient characteristics, such as age and renal function, as potential factors influencing the outcome. Missing data were imputed by using the median value of the entire study population, and variables with missing proportions exceeding 30% were excluded from evaluation. A corresponding odds ratio (OR) in relation to the reference group, along with the 95% confidence interval (CI) and p-values, were calculated for each univariate logistic regression model. Variables with a p value of <0.05 in the univariate analysis were included in the multivariate analysis. To evaluate the discrimination of the logistic regression model, ROC curves were constructed, and AUROC was calculated as follows:
log p 1 p = β 0 + β 1 X 1 + + β k X k
where p is the probability that an observation is in a specified category of the binary Y variable, p 1 p describes the odds of being in the current category of interest, the (natural) logarithm of the odds log p 1 p is a linear function of the X variables (and is often called the log odds). This is also referred to as the logit transformation of the probability of success. β0 is the coefficient on the constant term, X is the independent variable(s), and β k is the coefficient on the k t h independent variable.
The data management, all the analysis, simulation and plotting were carried out using the R software (version 4.2.0, Comprehensive R Network, http://cran.r-project.org/, accessed on 9 December 2023).

5. Conclusions

In conclusion, our quantitative meta-analysis has provided evidence of the correlation between vancomycin Ctrough and nephrotoxicity incidence. The findings support that monitoring Ctrough is still beneficial and can be a valuable approach in clinical practice, particularly when the concentration exceeds 15 mg/L. Ctrough can serve as a surrogate for AUC24 to some extent. No definite cut-off was determined for AUC24 in relation to nephrotoxicity, and likewise, for Ctrough and AUC24/MIC in terms of efficacy, underscoring the need for additional investigations.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antibiotics13060497/s1, Table S1. Methodological characteristics of the included studies for nephrotoxicity; Table S2. Patients’ characteristics in included studies for nephrotoxicity; Table S3. Methodological characteristics of the included studies for efficacy; Table S4. Patients’ characteristics in included studies for efficacy. Figure S1. Probability of nephrotoxicity for the trough category; Figure S2. Receiver operating characteristic curve of predictive level of vancomycin trough concentration for nephrotoxicity; Figure S3. Logistic regression illustrating the association of the probability of experiencing nephrotoxicity and as a function of vancomycin trough concentrations in patients without any form of dialysis; Figure S4. Correlation between nephrotoxicity and AUC24; Figure S5. Logistic regression illustrating the association of the treatment success rates and as a function of vancomycin AUC24/MICBMD; Figure S6. Correlation between efficacy outcomes and trough levels. References [31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163] are cited in Supplementary Materials.

Author Contributions

Data search and extraction, W.Y. and K.Z.; methodology, W.Y., Y.C., Y.F. and J.Z.; study design, J.Z.; data analysis, W.Y; writing—original draft preparation, W.Y.; writing—review and editing, Y.F., K.Z. and J.Z.; funding acquisition, Y.F. and J.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Municipal Hospital Emerging Frontier Technology Joint Research Project of Shanghai Shenkang Development Center (SHDC12020106), National Natural Science Foundation of China (82204467), the Research Startup Fund of Huashan Hospital, Fudan University (2021QD033) and the Community infectious disease research capacity building project (BCF-XC-SQ-20221206-07).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Acknowledgments

The authors thank team members from Institute of Antibiotics, Huashan Hospital, Fudan University for their valuable contributions.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Liu, C.; Bayer, A.; Cosgrove, S.E.; Daum, R.S.; Fridkin, S.K.; Gorwitz, R.J.; Kaplan, S.L.; Karchmer, A.W.; Levine, D.P.; Murray, B.E.; et al. Clinical practice guidelines by the infectious diseases society of america for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children: Executive summary. Clin. Infect. Dis. 2011, 52, 285–292. [Google Scholar] [CrossRef] [PubMed]
  2. Rybak, M.J. The pharmacokinetic and pharmacodynamic properties of vancomycin. Clin. Infect. Dis. 2006, 42 (Suppl. S1), S35–S39. [Google Scholar] [CrossRef] [PubMed]
  3. Craig, W.A. Basic pharmacodynamics of antibacterials with clinical applications to the use of beta-lactams, glycopeptides, and linezolid. Infect. Dis. Clin. N. Am. 2003, 17, 479–501. [Google Scholar] [CrossRef] [PubMed]
  4. Rybak, M.J.; Lomaestro, B.M.; Rotschafer, J.C.; Moellering, R.C.; Craig, W.A.; Billeter, M.; Dalovisio, J.R.; Levine, D.P. Vancomycin therapeutic guidelines: A summary of consensus recommendations from the Infectious Diseases Society of America, the American Society of Health-System Pharmacists, and the Society of Infectious Diseases Pharmacists. Clin. Infect. Dis. 2009, 49, 325–327. [Google Scholar] [CrossRef] [PubMed]
  5. Tsutsuura, M.; Moriyama, H.; Kojima, N.; Mizukami, Y.; Tashiro, S.; Osa, S.; Enoki, Y.; Taguchi, K.; Oda, K.; Fujii, S.; et al. The monitoring of vancomycin: A systematic review and meta-analyses of area under the concentration-time curve-guided dosing and trough-guided dosing. BMC Infect. Dis. 2021, 21, 153. [Google Scholar] [CrossRef]
  6. Oda, K.; Jono, H.; Nosaka, K.; Saito, H. Reduced nephrotoxicity with vancomycin therapeutic drug monitoring guided by area under the concentration-time curve against a trough 15–20 mug/mL concentration. Int. J. Antimicrob. Agents 2020, 56, 106109. [Google Scholar] [CrossRef] [PubMed]
  7. Rees, M.R.; Carr, D.R.; Trienski, T.; Buchanan, C.; White, K.; Bremmer, D.N. Outpatient vancomycin therapy: Acute kidney injury in individualized AUC-based goal trough ranges versus traditional trough dosing. J. Am. Pharm. Assoc. (2003) 2022, 62, 706–710. [Google Scholar] [CrossRef] [PubMed]
  8. Finch, N.A.; Zasowski, E.J.; Murray, K.P.; Mynatt, R.P.; Zhao, J.J.; Yost, R.; Pogue, J.M.; Rybak, M.J. A quasi-experiment to study the impact of vancomycin area under the concentration-time curve-guided dosing on vancomycin-associated nephrotoxicity. Antimicrob. Agents Chemother. 2017, 61, e01293-17. [Google Scholar] [CrossRef] [PubMed]
  9. Linder, A.; Fjell, C.; Levin, A.; Walley, K.R.; Russell, J.A.; Boyd, J.H. Small acute increases in serum creatinine are associated with decreased long-term survival in the critically ill. Am. J. Respir. Crit. Care Med. 2014, 189, 1075–1081. [Google Scholar] [CrossRef]
  10. Lee, B.V.; Fong, G.; Bolaris, M.; Neely, M.; Minejima, E.; Kang, A.; Lee, G.; Gong, C.L. Cost-benefit analysis comparing trough, two-level AUC and Bayesian AUC dosing for vancomycin. Clin. Microbiol. Infect. 2021, 27, 1346.e1–1346.e7. [Google Scholar] [CrossRef]
  11. Aljefri, D.M.; Avedissian, S.N.; Rhodes, N.J.; Postelnick, M.J.; Nguyen, K.; Scheetz, M.H. Vancomycin Area under the curve and acute kidney injury: A meta-analysis. Clin. Infect. Dis. 2019, 69, 1881–1887. [Google Scholar] [CrossRef] [PubMed]
  12. Pai, M.P.; Neely, M.; Rodvold, K.A.; Lodise, T.P. Innovative approaches to optimizing the delivery of vancomycin in individual patients. Adv. Drug Deliv. Rev. 2014, 77, 50–57. [Google Scholar] [CrossRef] [PubMed]
  13. Neely, M.N.; Youn, G.; Jones, B.; Jelliffe, R.W.; Drusano, G.L.; Rodvold, K.A.; Lodise, T.P. Are vancomycin trough concentrations adequate for optimal dosing? Antimicrob. Agents Chemother. 2014, 58, 309–316. [Google Scholar] [CrossRef] [PubMed]
  14. Clark, L.; Skrupky, L.P.; Servais, R.; Brummitt, C.F.; Dilworth, T.J. Examining the relationship between vancomycin area under the concentration time curve and serum trough levels in adults with presumed or documented Staphylococcal infections. Ther. Drug Monit. 2019, 41, 483–488. [Google Scholar] [CrossRef] [PubMed]
  15. Bel Kamel, A.; Bourguignon, L.; Marcos, M.; Ducher, M.; Goutelle, S. Is Trough concentration of vancomycin predictive of the area under the curve? A clinical study in elderly patients. Ther. Drug Monit. 2017, 39, 83–87. [Google Scholar] [CrossRef] [PubMed]
  16. Lodise, T.P.; Drusano, G. Vancomycin area under the curve-guided dosing and monitoring for adult and pediatric patients with suspected or documented serious methicillin-resistant Staphylococcus aureus infections: Putting the safety of our patients first. Clin. Infect. Dis. 2021, 72, 1497–1501. [Google Scholar] [CrossRef] [PubMed]
  17. Rybak, M.J.; Le, J.; Lodise, T.P.; Levine, D.P.; Bradley, J.S.; Liu, C.; Mueller, B.A.; Pai, M.P.; Wong-Beringer, A.; Rotschafer, J.C.; et al. Therapeutic monitoring of vancomycin for serious methicillin-resistant Staphylococcus aureus infections: A revised consensus guideline and review by the American Society of Health-system Pharmacists, the Infectious Diseases Society of America, the Pediatric Infectious Diseases Society, and the Society of Infectious Diseases Pharmacists. Clin. Infect. Dis. 2020, 71, 1361–1364. [Google Scholar] [PubMed]
  18. Matsumoto, K.; Oda, K.; Shoji, K.; Hanai, Y.; Takahashi, Y.; Fujii, S.; Hamada, Y.; Kimura, T.; Mayumi, T.; Ueda, T.; et al. Clinical practice guidelines for therapeutic drug monitoring of vancomycin in the framework of model-informed precision dosing: A consensus review by the Japanese Society of Chemotherapy and the Japanese Society of Therapeutic Drug Monitoring. Pharmaceutics 2022, 14, 489. [Google Scholar] [CrossRef] [PubMed]
  19. Dalton, B.R.; Rajakumar, I.; Langevin, A.; Ondro, C.; Sabuda, D.; Griener, T.P.; Dersch-Mills, D.; Rennert-May, E. Vancomycin area under the curve to minimum inhibitory concentration ratio predicting clinical outcome: A systematic review and meta-analysis with pooled sensitivity and specificity. Clin. Microbiol. Infect. 2020, 26, 436–446. [Google Scholar] [CrossRef]
  20. Bellos, I.; Daskalakis, G.; Pergialiotis, V. Relationship of vancomycin trough levels with acute kidney injury risk: An exposure-toxicity meta-analysis. J. Antimicrob. Chemother. 2020, 75, 2725–2734. [Google Scholar] [CrossRef]
  21. Lodise, T.P.; Patel, N.; Lomaestro, B.M.; Rodvold, K.A.; Drusano, G.L. Relationship between initial vancomycin concentration-time profile and nephrotoxicity among hospitalized patients. Clin. Infect. Dis. 2009, 49, 507–514. [Google Scholar] [CrossRef] [PubMed]
  22. Filippone, E.J.; Kraft, W.K.; Farber, J.L. The Nephrotoxicity of Vancomycin. Clin. Pharmacol. Ther. 2017, 102, 459–469. [Google Scholar] [CrossRef] [PubMed]
  23. Yu, Z.; Liu, J.; Yu, H.; Zhou, L.; Zhao, Y.; Zhong, L.; Zhu, J.; Liang, G.; Yang, Y.; Zheng, Y.; et al. Should the trough concentration of vancomycin be abandoned in therapeutic drug monitoring? A multicentre, retrospective study of critically ill patients without any form of dialysis. Int. J. Antimicrob. Agents 2023, 61, 106812. [Google Scholar] [CrossRef] [PubMed]
  24. He, N.; Su, S.; Ye, Z.; Du, G.; He, B.; Li, D.; Liu, Y.; Yang, K.; Zhang, X.; Zhang, Y.; et al. Evidence-based Guideline for Therapeutic Drug Monitoring of Vancomycin: 2020 Update by the Division of Therapeutic Drug Monitoring, Chinese Pharmacological Society. Clin. Infect. Dis. 2020, 71, S363–S371. [Google Scholar] [CrossRef] [PubMed]
  25. Reuter, S.E.; Stocker, S.L.; Alffenaar, J.C.; Baldelli, S.; Cattaneo, D.; Jones, G.; Koch, B.C.P.; Kocic, D.; Mathew, S.K.; Molinaro, M.; et al. Optimal practice for vancomycin therapeutic drug monitoring: Position statement from the Anti-infectives Committee of the International Association of Therapeutic Drug Monitoring and Clinical Toxicology. Ther. Drug Monit. 2022, 44, 121–132. [Google Scholar] [CrossRef]
  26. Abdul-Aziz, M.H.; Alffenaar, J.C.; Bassetti, M.; Bracht, H.; Dimopoulos, G.; Marriott, D.; Neely, M.N.; Paiva, J.A.; Pea, F.; Sjovall, F.; et al. Antimicrobial therapeutic drug monitoring in critically ill adult patients: A Position Paper. Intensive Care Med. 2020, 46, 1127–1153. [Google Scholar] [CrossRef]
  27. Khwaja, A. KDIGO clinical practice guidelines for acute kidney injury. Nephron Clin. Pract. 2012, 120, c179–c184. [Google Scholar] [CrossRef]
  28. Mehta, R.L.; Kellum, J.A.; Shah, S.V.; Molitoris, B.A.; Ronco, C.; Warnock, D.G.; Levin, A.; Acute Kidney Injury, N. Acute Kidney Injury Network: Report of an initiative to improve outcomes in acute kidney injury. Crit. Care 2007, 11, R31. [Google Scholar] [CrossRef] [PubMed]
  29. Bellomo, R.; Ronco, C.; Kellum, J.A.; Mehta, R.L.; Palevsky, P.; Acute Dialysis Quality Initiative Workgroup. Acute renal failure—definition, outcome measures, animal models, fluid therapy and information technology needs: The Second International Consensus Conference of the Acute Dialysis Quality Initiative (ADQI) Group. Crit. Care 2004, 8, R204–R212. [Google Scholar] [CrossRef]
  30. Akobeng, A.K. Understanding diagnostic tests 3: Receiver operating characteristic curves. Acta Paediatr. 2007, 96, 644–647. [Google Scholar] [CrossRef]
  31. Sohn, Y.; Rim, J.H.; Cho, Y.; Hyun, J.; Baek, Y.; Kim, M.; Kim, J.H.; Seong, H.; Ahn, J.Y.; Lee, S.G.; et al. Association of vancomycin trough concentration on the treatment outcome of patients with bacteremia caused by Enterococcus species. BMC Infect. Dis. 2021, 21, 1099. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  32. Marko, R.; Hajjar, J.; Nzeribe, V.; Pittman, M.; Deslandes, V.; Sant, N.; Cowan, J.; Kyermentang, K.; Ramsay, T.; Zelenitsky, S.; et al. Therapeutic Drug Monitoring of Vancomycin in Adult Patients with Methicillin-Resistant Staphylococcus aureus Bacteremia or Pneumonia. Can. J. Hosp. Pharm. 2021, 74, 334–343. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  33. Katip, W.; Oberdorfer, P. A Monocentric Retrospective Study of AUC/MIC Ratio of Vancomycin Associated with Clinical Outcomes and Nephrotoxicity in Patients with Enterococcal Infections. Pharmaceutics 2021, 13, 1378. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  34. Al-Sulaiti, F.K.; Nader, A.M.; Saad, M.O.; Shaukat, A.; Parakadavathu, R.; Elzubair, A.; Al-Badriyeh, D.; Elewa, H.; Awaisu, A. Clinical and Pharmacokinetic Outcomes of Peak-Trough-Based Versus Trough-Based Vancomycin Therapeutic Drug Monitoring Approaches: A Pragmatic Randomized Controlled Trial. Eur. J. Drug Metab. Pharmacokinet 2019, 44, 639–652. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  35. Wan, M.; Walker, S.A.N.; Martin, E.; Elligsen, M.; Palmay, L.; Leis, J.A. The impact of vancomycin trough concentrations on outcomes in non-deep seated infections: A retrospective cohort study. BMC Pharmacol. Toxicol. 2018, 19, 47. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  36. Mogle, B.T.; Steele, J.M.; Seabury, R.W.; Dang, U.J.; Kufel, W.D. Implementation of a two-point pharmacokinetic AUC-based vancomycin therapeutic drug monitoring approach in patients with methicillin-resistant Staphylococcus aureus bacteraemia. Int. J. Antimicrob. Agents 2018, 52, 805–810. [Google Scholar] [CrossRef] [PubMed]
  37. Jumah, M.T.B.; Vasoo, S.; Menon, S.R.; De, P.P.; Neely, M.; Teng, C.B. Pharmacokinetic/Pharmacodynamic Determinants of Vancomycin Efficacy in Enterococcal Bacteremia. Antimicrob. Agents Chemother. 2018, 62, e01602-17. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  38. Fukumori, S.; Tsuji, Y.; Mizoguchi, A.; Kasai, H.; Ishibashi, T.; Iwamura, N.; To, H. Association of the clinical efficacy of vancomycin with the novel pharmacokinetic parameter area under the trough level (AUTL) in elderly patients with hospital-acquired pneumonia. J. Clin. Pharm. Ther. 2016, 41, 399–402. [Google Scholar] [CrossRef] [PubMed]
  39. Suzuki, Y.; Tokimatsu, I.; Morinaga, Y.; Sato, Y.; Takano, K.; Kohno, K.; Ogata, M.; Hiramatsu, K.; Itoh, H.; Kadota, J. A retrospective analysis to estimate target trough concentration of vancomycin for febrile neutropenia in patients with hematological malignancy. Clin. Chim. Acta. 2015, 440, 183–187. [Google Scholar] [CrossRef] [PubMed]
  40. Zhang, X.; Wang, D. The characteristics and impact indicator of vancomycin pharmacokinetics in cancer patients complicated with severe pneumonia. J. Infect. Chemother. 2020, 26, 492–497. [Google Scholar] [CrossRef] [PubMed]
  41. Perin, N.; Roger, C.; Marin, G.; Molinari, N.; Evrard, A.; Lavigne, J.P.; Barbar, S.; Claret, P.G.; Boutin, C.; Muller, L.; et al. Vancomycin Serum Concentration after 48 h of Administration: A 3-Years Survey in an Intensive Care Unit. Antibiotics 2020, 9, 793. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  42. Yahav, D.; Abbas, M.; Nassar, L.; Ghrayeb, A.; Shepshelovich, D.; Kurnik, D.; Leibovici, L.; Paul, M. Attention to age: Similar dosing regimens lead to different vancomycin levels among older and younger patients. Age Ageing 2019, 49, 26–31. [Google Scholar] [CrossRef] [PubMed]
  43. Frazee, E.; Rule, A.D.; Lieske, J.C.; Kashani, K.B.; Barreto, J.N.; Virk, A.; Kuper, P.J.; Dierkhising, R.A.; Leung, N. Cystatin C-Guided Vancomycin Dosing in Critically Ill Patients: A Quality Improvement Project. Am. J. Kidney Dis. 2017, 69, 658–666. [Google Scholar] [CrossRef] [PubMed]
  44. Suzuki, Y.; Kawasaki, K.; Sato, Y.; Tokimatsu, I.; Itoh, H.; Hiramatsu, K.; Takeyama, M.; Kadota, J. Is peak concentration needed in therapeutic drug monitoring of vancomycin? A pharmacokinetic-pharmacodynamic analysis in patients with methicillin-resistant Staphylococcus aureus pneumonia. Chemotherapy 2012, 58, 308–312. [Google Scholar] [CrossRef] [PubMed]
  45. Wysocki, M.; Delatour, F.; Faurisson, F.; Rauss, A.; Pean, Y.; Misset, B.; Thomas, F.; Timsit, J.F.; Similowski, T.; Mentec, H. Continuous versus intermittent infusion of vancomycin in severe Staphylococcal infections: Prospective multicenter randomized study. Antimicrob. Agents Chemother. 2001, 45, 2460–2467. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  46. Gawronski, K.M.; Goff, D.A.; Brown, J.; Khadem, T.M.; Bauer, K.A. A stewardship program’s retrospective evaluation of vancomycin AUC24/MIC and time to microbiological clearance in patients with methicillin-resistant Staphylococcus aureus bacteremia and osteomyelitis. Clin. Ther. 2013, 35, 772–779. [Google Scholar] [CrossRef] [PubMed]
  47. Duszynska, W.; Taccone, F.S.; Hurkacz, M.; Wiela-Hojenska, A.; Kübler, A. Continuous vs. intermittent vancomycin therapy for Gram-positive infections not caused by methicillin-resistant Staphylococcus aureus. Minerva Anestesiol. 2016, 82, 284–293. [Google Scholar] [PubMed]
  48. Mizokami, F.; Shibasaki, M.; Yoshizue, Y.; Noro, T.; Mizuno, T.; Furuta, K. Pharmacodynamics of vancomycin in elderly patients aged 75 years or older with methicillin-resistant Staphylococcus aureus hospital-acquired pneumonia. Clin. Interv. Aging 2013, 8, 1015–1021. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Zasowski, E.J.; Murray, K.P.; Trinh, T.D.; Finch, N.A.; Pogue, J.M.; Mynatt, R.P.; Rybak, M.J. Identification of Vancomycin Exposure-Toxicity Thresholds in Hospitalized Patients Receiving Intravenous Vancomycin. Antimicrob. Agents Chemother. 2017, 62, e01684-17. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  50. Wang, J.L.; Xue, M.; Wang, H.F.; Huang, L.L.; Li, Q.; Xu, J.Y.; Xie, J.F.; Huang, Y.Z. An area under curve-based nomogram to predicts vancomycin-associated nephrotoxicity in critically ill patients: A retrospective cohort study. Zhonghua Nei Ke Za Zhi 2022, 61, 291–297. (In Chinese) [Google Scholar] [CrossRef] [PubMed]
  51. Ueda, T.; Takesue, Y.; Nakajima, K.; Ichiki, K.; Ishikawa, K.; Yamada, K.; Tsuchida, T.; Otani, N.; Takahashi, Y.; Ishihara, M.; et al. Validation of Vancomycin Area under the Concentration-Time Curve Estimation by the Bayesian Approach Using One-Point Samples for Predicting Clinical Outcomes in Patients with Methicillin-Resistant Staphylococcus aureus Infections. Antibiotics 2022, 11, 96. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  52. Yasu, T.; Konuma, T.; Oiwa-Monna, M.; Kato, S.; Isobe, M.; Takahashi, S.; Tojo, A. Lower vancomycin trough levels in adults undergoing unrelated cord blood transplantation. Leuk Lymphoma. 2021, 62, 348–357. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, Y.; Dai, N.; Wei, W.; Jiang, C. Outcomes and Nephrotoxicity Associated with Vancomycin Treatment in Patients 80 Years and Older. Clin. Interv. Aging 2021, 16, 1023–1035. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  54. Liu, K.; Zhang, Y.; Xu, X.; Wu, B.; Ni, J.; Li, T.; Xing, C.; Mao, H. Comparative Prevalence of Acute Kidney Injury in Chinese Patients Receiving Vancomycin with Concurrent β-Lactam Antibiotics: A Retrospective Cohort Study. Clin. Ther. 2021, 43, e319–e351. [Google Scholar] [CrossRef] [PubMed]
  55. Johnston, M.M.; Huang, V.; Hall, S.T.; Buckley, M.S.; Bikin, D.; Barletta, J.F. Optimizing outcomes using vancomycin therapeutic drug monitoring in patients with MRSA bacteremia: Trough concentrations or area under the curve? Diagn Microbiol. Infect. Dis. 2021, 101, 115442. [Google Scholar] [CrossRef] [PubMed]
  56. Al Sulaiman, K.; Alshaya, A.; Aljuhani, O.; Alsaeed, A.; Alshehri, N.; Vishwakarma, R.; Alzahrani, H.; Althewaibi, S.; Alghamdi, N.; Alhelal, K.; et al. The impact of early target attainment of vancomycin in critically ill patients with confirmed Gram-positive infection: A retrospective cohort study. BMC Infect. Dis. 2021, 21, 1182. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  57. Zhang, Y.; Wang, T.; Zhang, D.; You, H.; Dong, Y.; Liu, Y.; Du, Q.; Sun, D.; Zhang, T.; Dong, Y. Therapeutic Drug Monitoring Coupled With Bayesian Forecasting Could Prevent Vancomycin-Associated Nephrotoxicity in Renal Insufficiency Patients: A Prospective Study and Pharmacoeconomic Analysis. Ther. Drug Monit. 2020, 42, 600–609. [Google Scholar] [CrossRef] [PubMed]
  58. Ueki, T.; Sanematsu, E.; Furuya, Y.; Shinohara, Y.; Murakami, Y.; Miyazaki, A.; Sakamoto, Y.; Nakashima, M.N.; Nakashima, M. Relationship between vancomycin-associated nephrotoxicity and the number of combined nephrotoxic agents. Pharmazie 2020, 75, 279–283. [Google Scholar] [CrossRef] [PubMed]
  59. Qin, X.; Tsoi, M.F.; Zhao, X.; Zhang, L.; Qi, Z.; Cheung, B.M.Y. Vancomycin-associated acute kidney injury in Hong Kong in 2012–2016. BMC Nephrol. 2020, 21, 41. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  60. Pan, C.; Wen, A.; Li, X.; Li, D.; Zhang, Y.; Liao, Y.; Ren, Y.; Shen, S. Development and Validation of a Risk Prediction Model of Vancomycin-Associated Nephrotoxicity in Elderly Patients: A Pilot Study. Clin. Transl. Sci. 2020, 13, 491–497. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  61. Mcgrady, K.A.; Benton, M.; Tart, S.; Bowers, R. Evaluation of traditional initial vancomycin dosing versus utilizing an electronic AUC/MIC dosing program. Pharm. Pract. 2020, 18, 2024. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  62. Ma, N.H.; Walker, S.A.N.; Elligsen, M.; Kiss, A.; Palmay, L.; Ho, G.; Powis, J.; Bansal, V.; Leis, J.A. Retrospective multicentre matched cohort study comparing safety and efficacy outcomes of intermittent-infusion versus continuous-infusion vancomycin. J. Antimicrob. Chemother. 2020, 75, 1038–1046. [Google Scholar] [CrossRef] [PubMed]
  63. Imai, S.; Takekuma, Y.; Kashiwagi, H.; Miyai, T.; Kobayashi, M.; Iseki, K.; Sugawara, M. Validation of the usefulness of artificial neural networks for risk prediction of adverse drug reactions used for individual patients in clinical practice. PLoS ONE 2020, 15, e0236789. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  64. Brunetti, L.; Song, J.H.; Suh, D.; Kim, H.J.; Seong, Y.H.; Lee, D.S.; Lee, S.M.; Suh, D.C. The risk of vancomycin toxicity in patients with liver impairment. Ann. Clin. Microbiol. Antimicrob. 2020, 19, 13. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  65. Truong, J.; Smith, S.R.; Veillette, J.J.; Forland, S.C. Individualized Pharmacokinetic Dosing of Vancomycin Reduces Time to Therapeutic Trough Concentrations in Critically Ill Patients. J. Clin. Pharmacol. 2018, 58, 1123–1130. [Google Scholar] [CrossRef] [PubMed]
  66. Yahav, D.; Abbas, M.; Nassar, L.; Ghrayeb, A.; Kurnik, D.; Shepshelovich, D.; Leibovici, L.; Paul, M. The association of vancomycin trough levels with outcomes among patients with methicillin-resistant Staphylococcus aureus (MRSA) infections: Retrospective cohort study. PLoS ONE 2019, 14, e0214309. [Google Scholar] [CrossRef] [PubMed]
  67. de Almeida, C.D.C.; Simões E Silva, A.C.; de Queiroz Oliveira, J.A.; Batista, I.S.F.; Pereira, F.H.; Gonçalves, J.E.; Nobre, V.; Martins, M.A.P. Vancomycin-associated nephrotoxicity in non-critically ill patients admitted in a Brazilian public hospital: A prospective cohort study. PLoS ONE 2019, 14, e0222095. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  68. Nakashima, T.; Koido, K.; Baba, H.; Otsuka, R.; Okinaka, K.; Sano, T.; Nishigaki, R.; Hashimoto, H.; Otsuka, T.; Esaki, M.; et al. Contribution of pharmacists with expertise in infectious diseases to appropriate individualized vancomycin dosing. Pharmazie 2018, 73, 422–424. [Google Scholar] [CrossRef] [PubMed]
  69. May, C.C.; Erwin, B.L.; Childress, M.; Cortopassi, J.; Curtis, G.; Kilpatrick, T.; Taylor, J.; Vance, B.; Wylie, D. Assessment of acute kidney injury in neurologically and traumatically injured intensive care patients receiving large vancomycin doses. Int. J. Crit. Illn. Inj. Sci. 2018, 8, 194–200. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  70. Liang, X.; Fan, Y.; Yang, M.; Zhang, J.; Wu, J.; Yu, J.; Tao, J.; Lu, G.; Zhang, H.; Wang, R.; et al. A Prospective Multicenter Clinical Observational Study on Vancomycin Efficiency and Safety with Therapeutic Drug Monitoring. Clin. Infect. Dis. 2018, 67 (Suppl. S2), S249–S255. [Google Scholar] [CrossRef] [PubMed]
  71. Han, Z.; Pettit, N.N.; Landon, E.M.; Brielmaier, B.D. Impact of Pharmacy Practice Model Expansion on Pharmacokinetic Services: Optimization of Vancomycin Dosing and Improved Patient Safety. Hosp Pharm. 2017, 52, 273–279. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  72. Chavada, R.; Ghosh, N.; Sandaradura, I.; Maley, M.; Van Hal, S.J. Establishment of an AUC0-24 Threshold for Nephrotoxicity Is a Step towards Individualized Vancomycin Dosing for Methicillin-Resistant Staphylococcus aureus Bacteremia. Antimicrob. Agents Chemother. 2017, 61, e02535-16. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  73. Anderson, C.W.; Cazares, K.S.; Lustik, M.B.; Patel, S.M.; Denunzio, T.M. Vancomycin vs. Vancomycin/Piperacillin-Tazobactam-Associated Acute Kidney Injury in Noncritically Ill Patients at a Tertiary Care Military Treatment Facility. Mil. Med. 2017, 182, e1773–e1778. [Google Scholar] [CrossRef] [PubMed]
  74. Hammoud, K.; Brimacombe, M.; Yu, A.; Goodloe, N.; Haidar, W.; El Atrouni, W. Vancomycin Trough and Acute Kidney Injury: A Large Retrospective, Cohort Study. Am. J. Nephrol. 2016, 44, 456–461. [Google Scholar] [CrossRef] [PubMed]
  75. Hanrahan, T.P.; Kotapati, C.; Roberts, M.J.; Rowland, J.; Lipman, J.; Roberts, J.A.; Udy, A. Factors associated with vancomycin nephrotoxicity in the critically ill. Anaesth Intensive Care 2015, 43, 594–599. [Google Scholar] [CrossRef] [PubMed]
  76. Dong, M.H.; Wang, J.W.; Wu, Y.; Chen, B.Y.; Yu, M.; Wen, A.D. Evaluation of body weight-based vancomycin therapy and the incidence of nephrotoxicity: A retrospective study in the northwest of China. Int. J. Infect. Dis. 2015, 37, 125–128. [Google Scholar] [CrossRef] [PubMed]
  77. Hanrahan, T.P.; Harlow, G.; Hutchinson, J.; Dulhunty, J.M.; Lipman, J.; Whitehouse, T.; Roberts, J.A. Vancomycin-associated nephrotoxicity in the critically ill: A retrospective multivariate regression analysis*. Crit. Care Med. 2014, 42, 2527–2536. [Google Scholar] [CrossRef] [PubMed]
  78. Hall, R.G., 2nd; Blaszczyk, A.T.; Thompson, K.A.; Brouse, S.D.; Giuliano, C.A.; Frei, C.R.; Forcade, N.A.; Mortensen, E.M.; Bell, T.; Bedimo, R.J.; et al. Impact of empiric weight-based vancomycin dosing on nephrotoxicity and mortality in geriatric patients with methicillin-resistant Staphylococcus aureus bacteraemia. J. Clin. Pharm. Ther. 2014, 39, 653–657. [Google Scholar] [CrossRef] [PubMed]
  79. Burgess, L.D.; Drew, R.H. Comparison of the incidence of vancomycin-induced nephrotoxicity in hospitalized patients with and without concomitant piperacillin-tazobactam. Pharmacotherapy 2014, 34, 670–676. [Google Scholar] [CrossRef] [PubMed]
  80. Mizuno, T.; Mizokami, F.; Fukami, K.; Ito, K.; Shibasaki, M.; Nagamatsu, T.; Furuta, K. The influence of severe hypoalbuminemia on the half-life of vancomycin in elderly patients with methicillin-resistant Staphylococcus aureus hospital-acquired pneumonia. Clin. Interv. Aging 2013, 8, 1323–1328. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  81. Horey, A.; Mergenhagen, K.A.; Mattappallil, A. The Relationship of nephrotoxicity to vancomycin trough serum concentrations in a veteran’s population: A retrospective analysis. Ann. Pharmacother. 2012, 46, 1477–1483. [Google Scholar] [CrossRef] [PubMed]
  82. Hidayat, L.K.; Hsu, D.I.; Quist, R.; Shriner, K.A.; Wong-Beringer, A. High-dose vancomycin therapy for methicillin-resistant Staphylococcus aureus infections: Efficacy and toxicity. Arch Intern Med 2006, 166, 2138–2144. [Google Scholar] [CrossRef] [PubMed]
  83. Flannery, A.H.; Delozier, N.L.; Effoe, S.A.; Wallace, K.L.; Cook, A.M.; Burgess, D.S. First-Dose Vancomycin Pharmacokinetics Versus Empiric Dosing on Area-Under-the-Curve Target Attainment in Critically Ill Patients. Pharmacotherapy 2020, 40, 1210–1218. [Google Scholar] [CrossRef] [PubMed]
  84. Hirai, T.; Hanada, K.; Kanno, A.; Akashi, M.; Itoh, T. Risk factors for vancomycin nephrotoxicity and time course of renal function during vancomycin treatment. Eur. J. Clin. Pharmacol. 2019, 75, 859–866. [Google Scholar] [CrossRef] [PubMed]
  85. Okada, N.; Chuma, M.; Azuma, M.; Nakamura, S.; Miki, H.; Hamano, H.; Goda, M.; Takechi, K.; Zamami, Y.; Abe, M.; et al. Effect of serum concentration and concomitant drugs on vancomycin-induced acute kidney injury in haematologic patients: A single-centre retrospective study. Eur. J. Clin. Pharmacol. 2019, 75, 1695–1704. [Google Scholar] [CrossRef] [PubMed]
  86. Ramírez, E.; Jiménez, C.; Borobia, A.M.; Tong, H.Y.; Medrano, N.; Krauel-Bidwell, L.; Carcas, A.J.; Selgas, R.; Frías, J. Vancomycin-induced acute kidney injury detected by a prospective pharmacovigilance program from laboratory signals. Ther. Drug Monit. 2013, 35, 360–366. [Google Scholar] [CrossRef] [PubMed]
  87. Molina, K.C.; Barletta, J.F.; Hall, S.T.; Yazdani, C.; Huang, V. The Risk of Acute Kidney Injury in Critically Ill Patients Receiving Concomitant Vancomycin with Piperacillin-Tazobactam or Cefepime. J. Intensive Care Med. 2020, 35, 1434–1438. [Google Scholar] [CrossRef] [PubMed]
  88. Sharma, M.; Braekevelt, K.; Kale-Pradhan, P.; Szpunar, S.; Khatib, R. Are Blacks at Higher Risk for Vancomycin-Related Acute Kidney Injury? J. Pharm. Pract. 2020, 33, 592–597. [Google Scholar] [CrossRef] [PubMed]
  89. Hays, W.B.; Tillman, E. Vancomycin-Associated Acute Kidney Injury in Critically Ill Adolescent and Young Adult Patients. J. Pharm. Pract. 2020, 33, 749–753. [Google Scholar] [CrossRef] [PubMed]
  90. Covvey, J.R.; Erickson, O.; Fiumara, D.; Mazzei, K.; Moszczenski, Z.; Slipak, K.; Nemecek, B.D.; Zimmerman, D.E.; Guarascio, A.J. Comparison of Vancomycin Area-Under-the-Curve Dosing Versus Trough Target-Based Dosing in Obese and Nonobese Patients with Methicillin-Resistant Staphylococcus aureus Bacteremia. Ann. Pharmacother. 2020, 54, 644–651. [Google Scholar] [CrossRef] [PubMed]
  91. Zimmermann, A.E.; Katona, B.G.; Plaisance, K.I. Association of vancomycin serum concentrations with outcomes in patients with gram-positive bacteremia. Pharmacotherapy 1995, 15, 85–91. [Google Scholar] [CrossRef] [PubMed]
  92. Brumer, E.; Dubrovskaya, Y.; Scipione, M.R.; Aberle, C.; Rahimian, J.; Papadopoulos, J. Evaluation of Treatment Courses When Vancomycin Is Given Every 8 Hours in Adult Patients. J. Pharm. Pract. 2015, 28, 511–517. [Google Scholar] [CrossRef] [PubMed]
  93. Brown, M.L.; Hutchison, A.M.; McAtee, A.M.; Gaillard, P.R.; Childress, D.T. Allometric versus consensus guideline dosing in achieving target vancomycin trough concentrations. Am. J. Health Syst. Pharm. 2017, 74, 1067–1075. [Google Scholar] [CrossRef] [PubMed]
  94. Cano, E.L.; Haque, N.Z.; Welch, V.L.; Cely, C.M.; Peyrani, P.; Scerpella, E.G.; Ford, K.D.; Zervos, M.J.; Ramirez, J.A.; Kett, D.H. Improving Medicine through Pathway Assessment of Critical Therapy of Hospital-Acquired Pneumonia (IMPACT-HAP) Study Group. Incidence of nephrotoxicity and association with vancomycin use in intensive care unit patients with pneumonia: Retrospective analysis of the IMPACT-HAP Database. Clin. Ther. 2012, 34, 149–157. [Google Scholar] [CrossRef] [PubMed]
  95. Cappelletty, D.; Jablonski, A.; Jung, R. Risk factors for acute kidney injury in adult patients receiving vancomycin. Clin. Drug Investig. 2014, 34, 189–193. [Google Scholar] [CrossRef] [PubMed]
  96. Bhasin, B.; Ber Ce, P.; Szabo, A.; Chhabra, S.; D’Souza, A. Correlates and Outcomes of Early Acute Kidney Injury after Hematopoietic Cell Transplantation. Am. J. Med. Sci. 2021, 362, 72–77. [Google Scholar] [CrossRef] [PubMed]
  97. Huang, M.; Wu, H.; Zhou, J.; Xu, M.; Zhou, S. Efficacy of Vancomycin on Gram-Positive Bacterial Infection in Elderly Critical Patients and Risk Factors Associated with Nephrotoxicity. Arch. Iran. Med. 2018, 21, 349–355. [Google Scholar] [PubMed]
  98. Higashi, T.; Tsukamoto, H.; Kodawara, T.; Igarashi, T.; Watanabe, K.; Yano, R.; Iwasaki, H.; Goto, N. Evaluation of risk factors for nephrotoxicity associated with high-dose vancomycin in Japanese patients. Pharmazie 2021, 76, 114–118. [Google Scholar] [CrossRef] [PubMed]
  99. Fodero, K.E.; Horey, A.L.; Krajewski, M.P.; Ruh, C.A.; Sellick, J.A., Jr.; Mergenhagen, K.A. Impact of an Antimicrobial Stewardship Program on Patient Safety in Veterans Prescribed Vancomycin. Clin. Ther. 2016, 38, 494–502. [Google Scholar] [CrossRef] [PubMed]
  100. Golenia, B.S.; Levine, A.R.; Moawad, I.M.; Yeh, D.D.; Arpino, P.A. Evaluation of a vancomycin dosing nomogram based on the Modification of Diet in Renal Disease equation in intensive care unit patients. J. Crit. Care 2013, 28, 710–716. [Google Scholar] [CrossRef] [PubMed]
  101. Hale, C.M.; Seabury, R.W.; Steele, J.M.; Darko, W.; Miller, C.D. Are Vancomycin Trough Concentrations of 15 to 20 mg/L Associated with Increased Attainment of an AUC/MIC ≥ 400 in Patients with Presumed MRSA Infection? J. Pharm. Pract. 2017, 30, 329–335. [Google Scholar] [CrossRef] [PubMed]
  102. Hall, S.F.; Athans, V.; Wanek, M.R.; Wang, L.; Estep, J.D.; Williams, B. Evaluation of a hospital-wide vancomycin-dosing nomogram in patients with continuous-flow left ventricular assist devices. Int. J. Artif. Organs 2021, 44, 411–417. [Google Scholar] [CrossRef] [PubMed]
  103. Moh’d, H.; Kheir, F.; Kong, L.; Du, P.; Farag, H.; Mohamad, A.; Zurlo, J.J. Incidence and predictors of vancomycin-associated nephrotoxicity. S. Med. J. 2014, 107, 383–388. [Google Scholar] [CrossRef] [PubMed]
  104. Han, H.K.; An, H.; Shin, K.H.; Shin, D.; Lee, S.H.; Kim, J.H.; Cho, S.H.; Kang, H.R.; Jang, I.J.; Yu, K.S.; et al. Trough concentration over 12.1 mg/L is a major risk factor of vancomycin-related nephrotoxicity in patients with therapeutic drug monitoring. Ther. Drug Monit. 2014, 36, 606–611. [Google Scholar] [CrossRef] [PubMed]
  105. Haruki, Y.; Hagiya, H.; Haruki, M.; Inoue, Y.; Sugiyama, T. Concomitant vancomycin and piperacillin/tazobactam treatment is associated with an increased risk of acute kidney injury in Japanese patients. J. Infect. Chemother. 2020, 26, 1026–1032. [Google Scholar] [CrossRef] [PubMed]
  106. Hermsen, E.D.; Hanson, M.; Sankaranarayanan, J.; Stoner, J.A.; Florescu, M.C.; Rupp, M.E. Clinical outcomes and nephrotoxicity associated with vancomycin trough concentrations during treatment of deep-seated infections. Expert Opin. Drug Saf. 2010, 9, 9–14. [Google Scholar] [CrossRef] [PubMed]
  107. Hong, L.T.; Goolsby, T.A.; Sherman, D.S.; Mueller, S.W.; Reynolds, P.; Cava, L.; Neumann, R.; Kiser, T.H. Continuous infusion vs. intermittent vancomycin in neurosurgical intensive care unit patients. J. Crit. Care 2015, 30, 1153.e1–1153.e6. [Google Scholar] [CrossRef] [PubMed]
  108. Ghehi, M.T.; Rezaee, S.; Hayatshahi, A.; Hadjibabaie, M.; Gholami, K.; Javadi, M.; Khoee, S.H.; Radfar, M.; Esfandbod, M.; Ghavamzadeh, A. Vancomycin Pharmacokinetic Parameters in Patients Undergoing Hematopoietic Stem Cell Transplantation (HSCT). Int. J. Hematol. Oncol. Stem. Cell Res. 2013, 7, 1–9. [Google Scholar] [PubMed] [PubMed Central]
  109. Imai, S.; Yamada, T.; Kasashi, K.; Kobayashi, M.; Iseki, K. Usefulness of a decision tree model for the analysis of adverse drug reactions: Evaluation of a risk prediction model of vancomycin-associated nephrotoxicity constructed using a data mining procedure. J. Eval. Clin. Pract. 2017, 23, 1240–1246. [Google Scholar] [CrossRef] [PubMed]
  110. Imai, S.; Yamada, T.; Kasashi, K.; Niinuma, Y.; Kobayashi, M.; Iseki, K. Construction of a risk prediction model of vancomycin-associated nephrotoxicity to be used at the time of initial therapeutic drug monitoring: A data mining analysis using a decision tree model. J. Eval. Clin. Pract. 2019, 25, 163–170. [Google Scholar] [CrossRef] [PubMed]
  111. Ko, A.; Harada, M.Y.; Barmparas, G.; Jay, J.; Sun, B.J.; Chen, E.; Mehrzadi, D.; Patel, B.; Mason, R.; Ley, E.J. Reducing acute kidney injury due to vancomycin in trauma patients. J. Trauma Acute Care Surg. 2016, 81, 352–357. [Google Scholar] [CrossRef] [PubMed]
  112. Kullar, R.; Leonard, S.N.; Davis, S.L.; Delgado, G., Jr.; Pogue, J.M.; Wahby, K.A.; Falcione, B.; Rybak, M.J. Validation of the effectiveness of a vancomycin nomogram in achieving target trough concentrations of 15–20 mg/L suggested by the vancomycin consensus guidelines. Pharmacotherapy 2011, 31, 441–448. [Google Scholar] [CrossRef] [PubMed]
  113. Kullar, R.; Davis, S.L.; Taylor, T.N.; Kaye, K.S.; Rybak, M.J. Effects of targeting higher vancomycin trough levels on clinical outcomes and costs in a matched patient cohort. Pharmacotherapy 2012, 32, 195–201, Erratum in Pharmacotherapy 2012, 32, 869. [Google Scholar] [CrossRef] [PubMed]
  114. Ley, E.J.; Liou, D.Z.; Singer, M.B.; Mirocha, J.; Srour, M.; Bukur, M.; Margulies, D.R.; Salim, A. Supratherapeutic vancomycin levels after trauma predict acute kidney injury and mortality. J. Surg. Res. 2013, 184, 501–506. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, Y.; Yin, Y.; Liu, X.Z.; Yao, H.J.; Li, L.X.; Chen, J.H.; Chen, T.; Lu, X.T.; Bu, S.H.; Zhang, J. Retrospective Analysis of Vancomycin Nephrotoxicity in Elderly Chinese Patients. Pharmacology 2015, 95, 279–284. [Google Scholar] [CrossRef] [PubMed]
  116. Masuda, N.; Maiguma, T.; Komoto, A.; Haruki, Y.; Sugiyama, T.; Kondo, S.; Teshima, D. Impact of pharmacist intervention on preventing nephrotoxicity from vancomycin. Int. J. Clin. Pharmacol. Ther. 2015, 53, 284–291. [Google Scholar] [CrossRef] [PubMed]
  117. Meng, L.; Wong, T.; Huang, S.; Mui, E.; Nguyen, V.; Espinosa, G.; Desai, J.; Holubar, M.; Deresinski, S. Conversion from Vancomycin Trough Concentration-Guided Dosing to Area under the Curve-Guided Dosing Using Two Sample Measurements in Adults: Implementation at an Academic Medical Center. Pharmacotherapy 2019, 39, 433–442. [Google Scholar] [CrossRef] [PubMed]
  118. Muklewicz, J.D.; Steuber, T.D.; Edwards, J.D. Evaluation of area under the concentration-time curve-guided vancomycin dosing with or without piperacillin-tazobactam on the incidence of acute kidney injury. Int. J. Antimicrob. Agents 2021, 57, 106234. [Google Scholar] [CrossRef] [PubMed]
  119. Park, S.J.; Lim, N.R.; Park, H.J.; Yang, J.W.; Kim, M.J.; Kim, K.; In, Y.W.; Lee, Y.M. Evaluation of risk factors for vancomycin-induced nephrotoxicity. Int. J. Clin. Pharm. 2018, 40, 1328–1334. [Google Scholar] [CrossRef] [PubMed]
  120. Prabaker, K.K.; Tran, T.P.; Pratummas, T.; Goetz, M.B.; Graber, C.J. Elevated vancomycin trough is not associated with nephrotoxicity among inpatient veterans. J. Hosp. Med. 2012, 7, 91–97. [Google Scholar] [CrossRef] [PubMed]
  121. Pritchard, L.; Baker, C.; Leggett, J.; Sehdev, P.; Brown, A.; Bayley, K.B. Increasing vancomycin serum trough concentrations and incidence of nephrotoxicity. Am. J. Med. 2010, 123, 1143–1149. [Google Scholar] [CrossRef] [PubMed]
  122. Qian, X.; Du, G.; Weng, C.; Zhou, H.; Zhou, X. Evaluation of the variability and safety of serum trough concentrations of vancomycin in patients admitted to the intensive care unit. Int. J. Infect. Dis. 2017, 60, 17–22. [Google Scholar] [CrossRef] [PubMed]
  123. Reynolds, D.C.; Waite, L.H.; Alexander, D.P.; DeRyke, C.A. Performance of a vancomycin dosage regimen developed for obese patients. Am. J. Health Syst. Pharm. 2012, 69, 944–950. [Google Scholar] [CrossRef] [PubMed]
  124. Robertson, A.D.; Li, C.; Hammond, D.A.; Dickey, T.A. Incidence of Acute Kidney Injury Among Patients Receiving the Combination of Vancomycin with Piperacillin-Tazobactam or Meropenem. Pharmacotherapy 2018, 38, 1184–1193. [Google Scholar] [CrossRef] [PubMed]
  125. Rybak, M.J.; Albrecht, L.M.; Boike, S.C.; Chandrasekar, P.H. Nephrotoxicity of vancomycin, alone and with an aminoglycoside. J. Antimicrob. Chemother. 1990, 25, 679–687. [Google Scholar] [CrossRef] [PubMed]
  126. Sazanami, K.; Inose, R.; Dote, S.; Horiuchi, N.; Kobayashi, Y.; Muraki, Y. Combination therapy of vancomycin and piperacillin/tazobactam in adult febrile neutropenia patients with haematopoietic malignancies increases the risk of acute kidney injury regardless of vancomycin trough concentration. J. Chemother. 2021, 33, 440–442. [Google Scholar] [CrossRef] [PubMed]
  127. Alosaimy, S.; Murray, K.P.; Zasowski, E.J.; Morrisette, T.; Lagnf, A.M.; Lodise, T.P.; Rybak, M.J. Vancomycin Area under the Curve to Predict Timely Clinical Response in the Treatment of Methicillin-resistant Staphylococcus aureus Complicated Skin and Soft Tissue Infections. Clin. Infect. Dis. 2021, 73, e4560–e4567. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  128. Fan, Y.X.; Chen, M.T.; Li, N.Y.; Liu, X.F.; Yang, M.J.; Chen, Y.C.; Liang, X.Y.; Wu, J.F.; Guo, B.N.; Song, S.C.; et al. Sequence Type 5 (ST5) as a Possible Predictor of Bacterial Persistence in Adult Patients with Methicillin-Resistant Staphylococcus aureus Pneumonia Treated with Vancomycin. Microbiol. Spectr. 2022, 10, e0134822. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  129. Ren, J.; Hou, Y.; Li, J.; Gao, Y.; Li, R.; Jin, X.; Zhang, J.; Wang, X.; Wang, G. An evaluation on the association of vancomycin trough concentration with mortality in critically ill patients: A multicenter retrospective study. Clin. Transl. Sci. 2021, 14, 1780–1790. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  130. Lines, J.; Burchette, J.; Kullab, S.M.; Lewis, P. Evaluation of a trough-only extrapolated area under the curve vancomycin dosing method on clinical outcomes. Int. J. Clin. Pharm. 2021, 43, 263–269. [Google Scholar] [CrossRef] [PubMed]
  131. Hou, Y.; Ren, J.; Li, J.; Jin, X.; Gao, Y.; Li, R.; Zhang, J.; Wang, X.; Li, X.; Wang, G. Relationship Between Mean Vancomycin Trough Concentration and Mortality in Critically Ill Patients: A Multicenter Retrospective Study. Front Pharmacol. 2021, 12, 690157. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  132. Lodise, T.P.; Rosenkranz, S.L.; Finnemeyer, M.; Evans, S.; Sims, M.; Zervos, M.J.; Creech, C.B.; Patel, P.C.; Keefer, M.; Riska, P.; 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 Methicillin-resistant Staphylococcus aureus Bloodstream Infections (PROVIDE). Clin. Infect. Dis. 2020, 70, 1536–1545. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  133. Chattaweelarp, T.; Changpradub, D.; Punyawudho, B.; Thunyaharn, S.; Santimaleeworagun, W. Is Early Monitoring Better? Impact of Early Vancomycin Exposure on Treatment Outcomes and Nephrotoxicity in Patients with Methicillin-Resistant Staphylococcus aureus Infections. Antibiotics 2020, 9, 672. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  134. Makmor-Bakry, M.; Ahmat, A.; Shamsuddin, A.; Lau, C.L.; Ramli, R. Association between single trough-based area under the curve estimation of vancomycin and treatment outcome among methicillin-resistant Staphylococcus aureus bacteremia patients. Anaesthesiol. Intensive Ther. 2019, 51, 218–223. [Google Scholar] [CrossRef] [PubMed]
  135. Shen, K.; Yang, M.; Fan, Y.; Liang, X.; Chen, Y.; Wu, J.; Yu, J.; Zhang, H.; Wang, R.; Zhang, F.; et al. Model-based Evaluation of the Clinical and Microbiological Efficacy of Vancomycin: A Prospective Study of Chinese Adult In-house Patients. Clin. Infect. Dis. 2018, 67 (Suppl. S2), S256–S262. [Google Scholar] [CrossRef] [PubMed]
  136. Komoto, A.; Maiguma, T.; Teshima, D.; Sugiyama, T.; Haruki, Y. Effects of pharmacist intervention in Vancomycin treatment for patients with bacteremia due to Methicillin-resistant Staphylococcus aureus. PLoS ONE 2018, 13, e0203453. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  137. Fu, C.F.; Huang, J.D.; Wang, J.T.; Lin, S.W.; Wu, C.C. The ratio of pre-dialysis vancomycin trough serum concentration to minimum inhibitory concentration is associated with treatment outcomes in methicillin-resistant Staphylococcus aureus bacteremia. PLoS ONE 2018, 13, e0193585. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  138. Moise, P.A.; Culshaw, D.L.; Wong-Beringer, A.; Bensman, J.; Lamp, K.C.; Smith, W.J.; Bauer, K.; Goff, D.A.; Adamson, R.; Leuthner, K.; et al. Comparative Effectiveness of Vancomycin Versus Daptomycin for MRSA Bacteremia With Vancomycin MIC > 1 mg/L: A Multicenter Evaluation. Clin. Ther. 2016, 38, 16–30. [Google Scholar] [CrossRef] [PubMed]
  139. Ji, M.; Kim, H.K.; Kim, S.K.; Lee, W.; Sung, H.; Chun, S.; Kim, M.N.; Min, W.K. Vancomycin AUC24 /MIC Ratio in Patients with Methicillin-Resistant Staphylococcus aureus Pneumonia. J. Clin. Lab. Anal. 2016, 30, 485–489. [Google Scholar] [CrossRef] [PubMed]
  140. Stevenson, S.; Tang, W.; Cho, Y.; Mudge, D.W.; Hawley, C.M.; Badve, S.V.; Johnson, D.W. The role of monitoring vancomycin levels in patients with peritoneal dialysis-associated peritonitis. Perit. Dial. Int. 2015, 35, 222–228. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  141. Song, K.H.; Kim, H.B.; Kim, H.S.; Lee, M.J.; Jung, Y.; Kim, G.; Hwang, J.H.; Kim, N.H.; Kim, M.; Kim, C.J.; et al. Impact of area under the concentration-time curve to minimum inhibitory concentration ratio on vancomycin treatment outcomes in methicillin-resistant Staphylococcus aureus bacteraemia. Int. J. Antimicrob. Agents 2015, 46, 689–695. [Google Scholar] [CrossRef] [PubMed]
  142. Casapao, A.M.; Lodise, T.P.; Davis, S.L.; Claeys, K.C.; Kullar, R.; Levine, D.P.; Rybak, M.J. Association between vancomycin day 1 exposure profile and outcomes among patients with methicillin-resistant Staphylococcus aureus infective endocarditis. Antimicrob. Agents Chemother. 2015, 59, 2978–2985. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  143. Cao, G.; Liang, X.; Zhang, J.; Zhou, Y.; Wu, J.; Zhang, Y.; Chen, Y.; Huang, J.; Liu, X.; Yu, J. Vancomycin serum trough concentration vs. clinical outcome in patients with gram-positive infection: A retrospective analysis. J. Clin. Pharm. Ther. 2015, 40, 640–644. [Google Scholar] [CrossRef] [PubMed]
  144. Lodise, T.P.; Drusano, G.L.; Zasowski, E.; Dihmess, A.; Lazariu, V.; Cosler, L.; McNutt, L.A. Vancomycin exposure in patients with methicillin-resistant Staphylococcus aureus bloodstream infections: How much is enough? Clin. Infect. Dis. 2014, 59, 666–675. [Google Scholar] [CrossRef] [PubMed]
  145. Lin, Z.; Jiang, Z.; Chen, J.; Ouyang, B.; Chen, M.; Guan, X. Clinical research for trough value of serum vancomycin in critical patients. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 2014, 26, 473–477. (In Chinese) [Google Scholar] [CrossRef] [PubMed]
  146. Jung, Y.; Song, K.H.; Cho, J.; Kim, H.S.; Kim, N.H.; Kim, T.S.; Choe, P.G.; Chung, J.Y.; Park, W.B.; Bang, J.H.; et al. Area under the concentration-time curve to minimum inhibitory concentration ratio as a predictor of vancomycin treatment outcome in methicillin-resistant Staphylococcus aureus bacteraemia. Int. J. Antimicrob. Agents 2014, 43, 179–183. [Google Scholar] [CrossRef] [PubMed]
  147. Ghosh, N.; Chavada, R.; Maley, M.; van Hal, S.J. Impact of source of infection and vancomycin AUC0-24/MICBMD targets on treatment failure in patients with methicillin-resistant Staphylococcus aureus bacteraemia. Clin. Microbiol. Infect. 2014, 20, O1098–O1105. [Google Scholar] [CrossRef] [PubMed]
  148. Zelenitsky, S.; Rubinstein, E.; Ariano, R.; Iacovides, H.; Dodek, P.; Mirzanejad, Y.; Kumar, A. Cooperative Antimicrobial Therapy of Septic Shock-CATSS Database Research Group. Vancomycin pharmacodynamics and survival in patients with methicillin-resistant Staphylococcus aureus-associated septic shock. Int. J. Antimicrob. Agents 2013, 41, 255–260. [Google Scholar] [CrossRef] [PubMed]
  149. Holmes, N.E.; Turnidge, J.D.; Munckhof, W.J.; Robinson, J.O.; Korman, T.M.; O’Sullivan, M.V.; Anderson, T.L.; Roberts, S.A.; Warren, S.J.; Gao, W.; et al. Vancomycin AUC/MIC ratio and 30-day mortality in patients with Staphylococcus aureus bacteremia. Antimicrob. Agents Chemother. 2013, 57, 1654–1663. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  150. Moore, C.L.; Osaki-Kiyan, P.; Haque, N.Z.; Perri, M.B.; Donabedian, S.; Zervos, M.J. Daptomycin versus vancomycin for bloodstream infections due to methicillin-resistant Staphylococcus aureus with a high vancomycin minimum inhibitory concentration: A case-control study. Clin. Infect. Dis. 2012, 54, 51–58. [Google Scholar] [CrossRef] [PubMed]
  151. Cheong, J.Y.; Makmor-Bakry, M.; Lau, C.L.; Abdul Rahman, R. The relationship between trough concentration of vancomycin and effect on methicillin-resistant Staphylococcus aureus in critically ill patients. S. Afr. Med. J. 2012, 102, 616–619. [Google Scholar] [CrossRef] [PubMed]
  152. Brown, J.; Brown, K.; Forrest, A. Vancomycin AUC24/MIC ratio in patients with complicated bacteremia and infective endocarditis due to methicillin-resistant Staphylococcus aureus and its association with attributable mortality during hospitalization. Antimicrob. Agents Chemother. 2012, 56, 634–638. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  153. Clemens, E.C.; Chan, J.D.; Lynch, J.B.; Dellit, T.H. Relationships between vancomycin minimum inhibitory concentration, dosing strategies, and outcomes in methicillin-resistant Staphylococcus aureus bacteremia. Diagn Microbiol. Infect. Dis. 2011, 71, 408–414. [Google Scholar] [CrossRef] [PubMed]
  154. Chung, J.; Oh, J.M.; Cho, E.M.; Jang, H.J.; Hong, S.B.; Lim, C.M.; Koh, Y.S. Optimal dose of vancomycin for treating methicillin-resistant Staphylococcus aureus pneumonia in critically ill patients. Anaesth Intensive Care 2011, 39, 1030–1037. [Google Scholar] [CrossRef] [PubMed]
  155. Chan, J.D.; Pham, T.N.; Wong, J.; Hessel, M.; Cuschieri, J.; Neff, M.; Dellit, T.H. Clinical outcomes of linezolid vs vancomycin in methicillin-resistant Staphylococcus aureus ventilator-associated pneumonia: Retrospective analysis. J. Intensive Care Med. 2011, 26, 385–391. [Google Scholar] [CrossRef] [PubMed]
  156. Kullar, R.; Davis, S.L.; Levine, D.P.; Rybak, M.J. Impact of vancomycin exposure on outcomes in patients with methicillin-resistant Staphylococcus aureus bacteremia: Support for consensus guidelines suggested targets. Clin. Infect. Dis. 2011, 52, 975–981. [Google Scholar] [CrossRef] [PubMed]
  157. Mohammedi, I.; Descloux, E.; Argaud, L.; Le Scanff, J.; Robert, D. Loading dose of vancomycin in critically ill patients: 15 mg/kg is a better choice than 500 mg. Int. J. Antimicrob. Agents 2006, 27, 259–262. [Google Scholar] [CrossRef] [PubMed]
  158. Jeffres, M.N.; Isakow, W.; Doherty, J.A.; McKinnon, P.S.; Ritchie, D.J.; Micek, S.T.; Kollef, M.H. Predictors of mortality for methicillin-resistant Staphylococcus aureus health-care-associated pneumonia: Specific evaluation of vancomycin pharmacokinetic indices. Chest 2006, 130, 947–955. [Google Scholar] [CrossRef] [PubMed]
  159. Moise-Broder, P.A.; Forrest, A.; Birmingham, M.C.; Schentag, J.J. Pharmacodynamics of vancomycin and other antimicrobials in patients with Staphylococcus aureus lower respiratory tract infections. Clin. Pharmacokinet 2004, 43, 925–942. [Google Scholar] [CrossRef] [PubMed]
  160. Vuagnat, A.; Stern, R.; Lotthe, A.; Schuhmacher, H.; Duong, M.; Hoffmeyer, P.; Bernard, L. High dose vancomycin for osteomyelitis: Continuous vs. intermittent infusion. J. Clin. Pharm. Ther. 2004, 29, 351–357. [Google Scholar] [CrossRef] [PubMed]
  161. Moise, P.A.; Forrest, A.; Bhavnani, S.M.; Birmingham, M.C.; Schentag, J.J. Area under the inhibitory curve and a pneumonia scoring system for predicting outcomes of vancomycin therapy for respiratory infections by Staphylococcus aureus. Am. J. Health Syst. Pharm. 2000, 57 (Suppl. S2), S4–S9, Erratum in Am. J. Health Syst. Pharm. 2001, 58, 78. [Google Scholar] [CrossRef] [PubMed]
  162. Karam, C.M.; McKinnon, P.S.; Neuhauser, M.M.; Rybak, M.J. Outcome assessment of minimizing vancomycin monitoring and dosing adjustments. Pharmacotherapy 1999, 19, 257–266, Erratum in Pharmacotherapy 1999, 19, 674. [Google Scholar] [CrossRef] [PubMed]
  163. Rojas, L.; Bunsow, E.; Muñoz, P.; Cercenado, E.; Rodríguez-Créixems, M.; Bouza, E. Vancomycin MICs do not predict the outcome of methicillin-resistant Staphylococcus aureus bloodstream infections in correctly treated patients. J. Antimicrob. Chemother. 2012, 67, 1760–1768. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow chart for study identification and selection for nephrotoxicity (A) and efficacy (B).
Figure 1. Flow chart for study identification and selection for nephrotoxicity (A) and efficacy (B).
Antibiotics 13 00497 g001
Figure 2. Correlation between nephrotoxicity and trough concentrations. Black circles represent the observed vancomycin Ctrough values from each study. The solid line and the shaded area represent the estimated Emax model curve with 95% credible intervals of parameters. The grey shade represents the interval between Ctrough 10 mg/L and 20 mg/L.
Figure 2. Correlation between nephrotoxicity and trough concentrations. Black circles represent the observed vancomycin Ctrough values from each study. The solid line and the shaded area represent the estimated Emax model curve with 95% credible intervals of parameters. The grey shade represents the interval between Ctrough 10 mg/L and 20 mg/L.
Antibiotics 13 00497 g002
Figure 3. Logistic regression illustrating the association of the probability of experiencing nephrotoxicity and as a function of vancomycin trough concentrations. The upper and lower circles represent the presence or absence of a given nephrotoxicity across the range of vancomycin trough concentrations, respectively. The dots depict the observed incidence for the quartiles of exposure, whereas the corresponding vertical bars represent the exact 95% CI calculated using Wilson’s method. Finally, the middle line and its corresponding shaded area represent the model-based exposure–safety relationship and the 95% CI, respectively. Vertical dashed lines represent min, 25%, median, 75% and max percentile of trough concentrations, respectively.
Figure 3. Logistic regression illustrating the association of the probability of experiencing nephrotoxicity and as a function of vancomycin trough concentrations. The upper and lower circles represent the presence or absence of a given nephrotoxicity across the range of vancomycin trough concentrations, respectively. The dots depict the observed incidence for the quartiles of exposure, whereas the corresponding vertical bars represent the exact 95% CI calculated using Wilson’s method. Finally, the middle line and its corresponding shaded area represent the model-based exposure–safety relationship and the 95% CI, respectively. Vertical dashed lines represent min, 25%, median, 75% and max percentile of trough concentrations, respectively.
Antibiotics 13 00497 g003
Figure 4. Logistic regression illustrating the association of the probability of experiencing nephrotoxicity and as a function of vancomycin AUC24. The upper and lower circles represent the presence or absence of a given nephrotoxicity across the range of vancomycin AUC24, respectively. The dots depict the observed incidence for the quartiles of exposure, whereas the corresponding vertical bars represent the exact 95% CI calculated using Wilson’s method. Finally, the middle line and its corresponding shaded area represent the model-based exposure–safety relationship and the 95% CI, respectively. Vertical dashed lines represent min, 25%, median, 75% and max percentile of AUC24, respectively.
Figure 4. Logistic regression illustrating the association of the probability of experiencing nephrotoxicity and as a function of vancomycin AUC24. The upper and lower circles represent the presence or absence of a given nephrotoxicity across the range of vancomycin AUC24, respectively. The dots depict the observed incidence for the quartiles of exposure, whereas the corresponding vertical bars represent the exact 95% CI calculated using Wilson’s method. Finally, the middle line and its corresponding shaded area represent the model-based exposure–safety relationship and the 95% CI, respectively. Vertical dashed lines represent min, 25%, median, 75% and max percentile of AUC24, respectively.
Antibiotics 13 00497 g004
Figure 5. Correlation between outcomes and continuous AUC24/MIC stratified by MIC method. The solid black line and the shaded area represent the estimated Emax model curve with 95% credible intervals of parameters to reflect the correlation between clinical outcomes and AUC24/MIC. The black dashed transverse line represents the 60% and 80% treatment success rate for each clinical outcome. The black circles represent the observed incidence of success of each clinical outcome. The shaded area represents the AUC24/MIC interval between 400 and 600.
Figure 5. Correlation between outcomes and continuous AUC24/MIC stratified by MIC method. The solid black line and the shaded area represent the estimated Emax model curve with 95% credible intervals of parameters to reflect the correlation between clinical outcomes and AUC24/MIC. The black dashed transverse line represents the 60% and 80% treatment success rate for each clinical outcome. The black circles represent the observed incidence of success of each clinical outcome. The shaded area represents the AUC24/MIC interval between 400 and 600.
Antibiotics 13 00497 g005
Figure 6. Correlation between exposure metrics (Steady-state AUC24 and trough concentrations). The chord diagram presents the difference of the mean VTC with mean AUC24 for each subgroup of studies. (A) The correlation between AUC24 and trough concentration from studies included in the nephrotoxicity analysis; (B) the correlation between AUC24 and trough concentration from studies reporting efficacy. VTC: vancomycin trough concentration.
Figure 6. Correlation between exposure metrics (Steady-state AUC24 and trough concentrations). The chord diagram presents the difference of the mean VTC with mean AUC24 for each subgroup of studies. (A) The correlation between AUC24 and trough concentration from studies included in the nephrotoxicity analysis; (B) the correlation between AUC24 and trough concentration from studies reporting efficacy. VTC: vancomycin trough concentration.
Antibiotics 13 00497 g006
Table 1. Estimated parameters of Emax model for nephrotoxicity and efficacy endpoints.
Table 1. Estimated parameters of Emax model for nephrotoxicity and efficacy endpoints.
OutcomeEndpointPK/PD ParameterEmax (%)
(CV%)
EC50 (mg/L)
(CV%)
γ
(CV%)
Nephrotoxicity2009 ConsensusCtrough (mg/L) (n = 90)32.5 (43.7%)18.8 (78.4)1.0 (FIX)
AKINCtrough (mg/L) (n = 22)42.7 (37.6)21.4 (86.2)1.51 (148)
KDIGOCtrough (mg/L) (n = 24)100 (FIX)22.7 (19.8)4.15 (45.1)
RIFLECtrough (mg/L) (n = 29)100 (FIX)51.1 (25.1)1.47 (23.1)
EfficacyTreatment failureAUC24/MICBMD (n = 11)100 (FIX)367 (20.0)1.0 FIX
AUC24/MICEtest (n = 6)100 (FIX)335 (36.6)2.65 (59.2)
30- or 28-day all-cause mortalityAUC24/MICBMD (n = 8)100 (FIX)123 (19.4)1.0 FIX
AUC24/MICEtest (n = 9)100 (FIX)96.7 (62.3)1.03 (54.4)
Microbiologic failureAUC24/MICBMD (n = 9)100 (FIX)296 (36.0)2.39 (64.9)
AUC24/MICEtest (n = 11)100 (FIX)99.8 (60.8)1.01 (79.2)
AKIN = Acute Kidney Injury Network; KDIGO = Kidney Disease Improving Global Outcomes; RIFLE = Risk, Injury, Failure, Loss of kidney function and End-stage kidney disease; BMD = broth microdilution; CV = coefficient of variation; PK/PD = pharmacokinetic/pharmacodynamics; Emax = maximum effect; EC50 = the indicators required to achieve half the Emax; γ = slope factor (also known as Hill factor).
Table 2. The overall distribution of mean trough concentration and AUC24.
Table 2. The overall distribution of mean trough concentration and AUC24.
Analysis, n (%/%)Ctrough (mg/L)
Nephrotoxicity (n = 61)AUC24 (mg·h/L)≤10 (n = 9)10–15 (n = 31)15–20 (n = 16)>20 (n = 5)
≤200 (n = 0)0 (0/0)0 (0/0)0 (0/0)0 (0/0)
200–400 (n = 10)7 (70.0/77.8)2 (20.0/6.5)1 (10.0/6.2)0 (0/0)
400–600 (n = 39)1 (2.6/11.1)24 (61.5/77.4)12 (30.8/75.0)2 (5.1/40.0)
>600 (n = 12)1 (8.3/11.1)5 (41.7/16.1)3 (25.0/18.8)3 (25.0/60.0)
Efficacy (n = 83)AUC24 (mg·h/L)≤10 (n = 28)10–15 (n = 45)15–20 (n = 7)>20 (n = 3)
≤200 (n = 0)0 (0/0)0 (0/0)0 (0/0)0 (0/0)
200–400 (n = 24)19 (79.2/67.9)5 (20.8/11.1)0 (0/0)0 (0/0)
400–600 (n = 54)9 (16.7/32.1)38 (70.4/84.4)5 (9.3/71.4)2 (3.7/66.7)
>600 (n = 5)0 (0/0)2 (40.0/4.4)2 (40.0/28.6)1 (20.0/33.3)
n (%/%) represents the numbers of groups in each paired group with the percentage (the first %) of groups in each of the AUC24 categories and the percentage (the second %) of groups in each of the Ctrough categories.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yang, W.; Zhang, K.; Chen, Y.; Fan, Y.; Zhang, J. Is It Still Beneficial to Monitor the Trough Concentration of Vancomycin? A Quantitative Meta-Analysis of Nephrotoxicity and Efficacy. Antibiotics 2024, 13, 497. https://doi.org/10.3390/antibiotics13060497

AMA Style

Yang W, Zhang K, Chen Y, Fan Y, Zhang J. Is It Still Beneficial to Monitor the Trough Concentration of Vancomycin? A Quantitative Meta-Analysis of Nephrotoxicity and Efficacy. Antibiotics. 2024; 13(6):497. https://doi.org/10.3390/antibiotics13060497

Chicago/Turabian Style

Yang, Wanqiu, Kaiting Zhang, Yuancheng Chen, Yaxin Fan, and Jing Zhang. 2024. "Is It Still Beneficial to Monitor the Trough Concentration of Vancomycin? A Quantitative Meta-Analysis of Nephrotoxicity and Efficacy" Antibiotics 13, no. 6: 497. https://doi.org/10.3390/antibiotics13060497

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop