Next Article in Journal
Polymer Conjugates of Antimicrobial Peptides (AMPs) with d-Amino Acids (d-aa): State of the Art and Future Opportunities
Previous Article in Journal
Focused Ultrasound-Induced Blood–Brain Barrier Opening Enhanced α-Synuclein Expression in Mice for Modeling Parkinson’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Prevalence and Risk Factors of Augmented Renal Clearance: A Systematic Review and Meta-Analysis

1
Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada
2
John W. Scott Health Sciences Library, University of Alberta, Edmonton, AB T6G 2R7, Canada
*
Author to whom correspondence should be addressed.
Pharmaceutics 2022, 14(2), 445; https://doi.org/10.3390/pharmaceutics14020445
Submission received: 30 December 2021 / Revised: 14 February 2022 / Accepted: 15 February 2022 / Published: 19 February 2022

Abstract

:
Kidney function assessment in the critically ill overlooks the possibility for hyperfunctioning kidneys, known as augmented renal clearance (ARC), which could contribute to therapeutic failures in the intensive care unit (ICU). The aim of this research is to conduct a systematic review and meta-analysis of prevalence and risk factors of ARC in the critically ill. MEDLINE, Embase, Cochrane Library, CINAHL, Scopus, ProQuest Dissertations and Theses Global databases were searched on 27 October 2020. We included studies conducted in critically ill adults who reported the prevalence and/or risk factors of ARC. We evaluated study quality using the Joanna Briggs Institute appraisal tool. Case reports, reviews, editorials and commentaries were excluded. We generated a random-effects meta-analytic model using the inverse variance method and visualized the pooled estimates using forest plots. Seventy studies were included. The pooled prevalence (95% CI) was 39% (34.9–43.3). Prevalence for neuro, trauma, mixed and sepsis ICUs were 74 (55–87), 58 (48–67), 36 (31–41) and 33 (21–48), respectively. Age, male sex and trauma were associated with ARC with pooled OR (95% CI) of 0.95 (0.93–0.96), 2.36 (1.28–4.36), 2.60 (1.21–5.58), respectively. Limitations included variations in ARC definition, inclusion and exclusion criteria and studies design. In conclusion, ARC is prevalent in critically ill patients, especially those in the neurocritical care and trauma ICU population. Young age, male sex and trauma are risk factors for ARC in those with apparently normal renal function. Further research on optimal dosing of drugs in the setting of ARC is warranted. (Prospero registration: CRD42021246417).

1. Introduction

Critical illness is unique for its complex nature, which very often requires a range of professional expertise to provide the most comprehensive care possible, hence the need for a multidisciplinary approach. When assessing a patient’s kidney function, particularly in a critical care setting, clinicians typically consider one of two possibilities: either normal renal function, or renal impairment, with most of the attention paid towards dosing adjustments in the presence of impaired renal function and/or the use of renal replacement therapy. This conventional view might in fact be overlooking a third category of patients who may exhibit hyperfunctioning kidneys or what is known as augmented renal clearance (ARC). This phenomenon, while not yet fully understood, may potentially be the rationale behind a range of therapeutic failures for renally-eliminated drugs [1,2,3]. This is mainly due to the fact that ARC is typically undetected unless clinicians proactively monitor for its presence and the lack of solid evidence on the dosing of renally-eliminated medications subject to an accelerated elimination, leading to subtherapeutic levels and sub-optimal outcomes. The pathophysiology of ARC is largely unknown, but it is thought to be closely tied to the vigorous sympathetic response associated with severe critical illness, alterations in vascular tone, cardiac output and major organs blood flow, resulting in a hyperdynamic state and augmented glomerular filtration rate [4,5]. This is in addition to the effects of administration of fluids and vasopressors aimed at maintaining organ perfusion [5,6]. ARC has most commonly been defined as a creatinine clearance (CrCl) higher than 130 mL/min/1.73 m2 [7,8,9]. However, there is not yet an agreed-upon cut-off for the CrCl above which a patient is diagnosed with ARC, nor a staging system for patients exhibiting CrCl more than 150 mL/min/1.73 m2 or even 200 mL/min/1.73 m2, analogous to renal impairment stages.
In recent years, there has been a growing number of reports recognizing the significance of ARC [4,10]. ARC prevalence has been reported to range from 18 to 80% in the general critically ill population [4,11,12,13,14,15,16,17,18]. However, reported studies varied in their patient population, sample sizes, inclusion and exclusion criteria and ARC definition, thus, impeding accurate identification of ARC prevalence and risk factors among intensive care unit (ICU) patients. Therefore, the aim of this research is to conduct a systematic review and meta-analysis of the available literature on ARC and to attempt to provide pooled estimates of its prevalence and contributing risk factors in various critically ill populations. To our knowledge, this is the first combined systematic review and meta-analysis of ARC in the critically ill. Our work represents a step towards defining the prevalence and risk factors of ARC, facilitating early identification of those at risk for ARC allowing timely medication optimization.

2. Materials and Methods

This review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) checklist [19]. This review was registered in international prospective register of systematic reviews (PROSPERO). Registration number CRD42021246417 and protocol can be accessed in the following link: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021246417.

2.1. Database Search Method

The medical librarian (JYK) developed comprehensive searches on 27 October 2020 in the following databases: MEDLINE (via Ovid), Embase (Ovid), Cochrane Library (Wiley), CINAHL, Scopus, and ProQuest Dissertations and Theses Global. Search strategies included keywords and controlled vocabulary related to augmented renal clearance in critical care (Supplementary Table S1). There were no date or language limits applied. To better facilitate the screening process, the research team used Covidence, a web-based systematic review screening tool (www.covidence.org). In addition to subscription databases, the first 200 results from Google Scholar were evaluated for inclusion. Bibliographies from included studies were also reviewed.

2.2. Inclusion and Exclusion Criteria

We included human studies conducted in critically ill adult populations that reported ARC prevalence and/or risk factors in our analysis. Studies also needed to have a clearly defined criteria for ARC and reported what method was used to measure or calculate CrCl. We excluded studies that focused on pediatric patients or patients with renal dysfunction (e.g., acute kidney injury), as well as studies conducted in populations that would have altered renal elimination (e.g., cystic fibrosis, burn patients). Case reports, reviews, editorials and commentaries were also excluded.

2.3. Study Screening

Study screening and selection were conducted independently by SHM and AS using Covidence. This was completed in two steps: (1) An initial title and abstract screening was performed. (2) The relevant abstracts were then introduced to a full-text review. The authors used discussion to come to a consensus about any arising conflicts during the screening process. Non-English language studies were translated using the Google Translate web-based document translator, when possible.

2.4. Data Extraction

The data were extracted independently by AS and FH from each of the included studies and then cross-checked to verify the integrity and completeness of the information. Any inconsistencies were resolved by discussion with SHM. The extracted data included: study design, exclusion and inclusion criteria, intensive care unit (ICU) type, ARC definition, diagnoses, patient demographics and ARC prevalence and risk factors contributing to ARC along with their measures of association. For studies that did not specify a cut-off for ARC but reported individual CrCl values, a value of >130 mL/min/1.73 m2 was applied to determine ARC prevalence.

2.5. Risk of Bias Assessment

All the included studies were individually assessed for their risk of bias by employing the “Joanna Briggs Institute Critical Appraisal Instrument for Studies Reporting Prevalence Data” (https://jbi.global/sites/default/files/2020-08/Checklist_for_Prevalence_Studies.pdf). This critical appraisal tool assessed nine aspects to assess the quality of each study: (1) Was the sample frame appropriate to address the target population? (2) Were study participants sampled in an appropriate way? (3) Was the sample size adequate? (4) Were the study subjects and the setting described in detail? (5) Was the data analysis conducted with sufficient coverage of the identified sample? (6) Were valid methods used for the identification of the condition? (7) Was the condition measured in a standard, reliable way for all participants? (8) Was there appropriate statistical analysis? (9) Was the response rate adequate, and if not, was the low response rate managed appropriately?

2.6. Data Analysis

The statistical analysis was performed by FH in consultation with a biostatistician using the package in R Statistical Software (Version 4.0.3, R Foundation for Statistical Computing, Vienna, Austria) and RStudio Interface (Version 1.3.1093, RStudio, Boston, MA, USA) [20,21,22]. For the meta-analysis of prevalence, the function metaprop was used to pool the meta-analytic estimate of prevalence of ARC using the reported number of cases and the total number of subjects in each included trial. We generated a random-effects meta-analytic model using the inverse variance method for weights, DerSimonian-Laird estimator [23,24] for Ƭ2 as the measure of true between-study variance, the Jackson method for confidence interval of Ƭ2 [25] and a Logit transformation to the calculated individual studies prevalence. Additionally, we examined the I2 statistic (the estimate of residual heterogeneity that is not due to sampling variation alone) and Cochrane Q statistic (describes the total heterogeneity not stemming from random error). The analyses were then visualized graphically using forest plots. To assess the risk of publication bias, Egger’s test [26] was conducted and tested for significance; a funnel plot was used to visualize the individual studies’ effect sizes against their estimate of precision. For studies reporting data for more than one distinct patient population, each population was entered separately in the meta-analysis. For the meta-analysis of risk factors, the function “metagen” from the package “meta” in R was utilized. It was used to synthesize the meta-analytic odds ratio size of the commonly reported risk factors: age, male sex, trauma, sequential organ failure assessment (SOFA) score, acute physiology and chronic health evaluation (APACHE II), and diabetes on ARC from their reported odds ratios of multivariate logistic regression.

3. Results

As depicted in Figure 1, comprehensive searches identified 3455 records across all databases. A total of 1761 records remained for screening after the removal of duplicate records. After the title and abstract screening, 384 records were subject to a full-text screening ending with a total of 70 included records Observational studies constituted the majority of collected evidence at 68 studies, along with 1 randomized controlled trial [27] and 1 prospective non-randomized interventional study [28]. Table 1 depicts a summary of the studies included in this systematic review and meta-analysis of prevalence and risk factors. Table 2 depicts a summary of the studies reporting other risk factors not included in the meta-analysis. Supplementary Table S2 depicts the risk of bias assessment of the included studies using the Joanna Briggs Institute critical appraisal tool for studies reporting prevalence data. The average score of all studies was 94.4%.

3.1. ARC Definition

Of the 70 included studies, 68 studies reported prevalence data. Studies varied in their definition of ARC in terms of CrCl cut-off. Most studies (52 records (76.5%)) defined ARC as CrCl ≥ 130 mL/min/1.73 m2; other definitions used were CrCl ≥ 120 mL/min/1.73 m2 (9 records (13.2%)), CrCl ≥ 150 mL/min/1.73 m2 (3 records (4.4%)), CrCl ≥ 140 mL/min/1.73 m2 (1 record (1.5%)), CrCl ≥ 155 mL/min/1.73 m2 (1 record (1.3%)), CrCl ≥ 160 mL/min/1.73 m2 (1 record (1.5%)), and CrCl ≥ 108 mL/min/1.73 m2 (1 record (1.5%)).

3.2. ARC Prevalence

Reports on the prevalence of ARC in this meta-analysis ranged between 4% and 100% in various critically ill populations, with an interquartile range of 25.9–55.8%, which suggests that ARC occurs very commonly. Our meta-analysis of prevalence included 68 studies representing 76 samples: 29 (38.2%) from mixed ICUs, 14 (18.4%) from sepsis ICUs, 9 (11.8%) from neuro ICUs, 9 (11.8%) from trauma ICUs, and 15 (19.7%) including patients from surgical, oncology, and other critically ill and non-critically ill hospitalized patients (Table 1). CrCl determination methods varied among studies, where 52 (68.4%) studies measured CrCl utilizing a 6–24 h urine collection method and 24 (31.6%) studies calculated CrCl using various equations. Among the studies that calculated CrCl, the majority used Cockcroft and Gault’s formula (n = 15).
The meta-analysis of prevalence of all included studies yielded a pooled prevalence (95% CI) of 39% (34.9–43.3) including patients from mixed (Figure 2), neuro, sepsis, trauma, surgical, and oncology critical care units, as well as non-ICU patients. The highest ARC occurrence was detected in neurocritical care patients with a 74% pooled prevalence across the 9 studies (Figure 3A), followed by 58% in trauma ICUs across 9 studies (Figure 3B), 36% in mixed ICUs across 29 studies (Figure 2), 33% in sepsis ICUs (Figure 4A), and 27% in the other patient populations collectively (Figure 4B). A meta-analysis of ARC prevalence in studies that only measured CrCl yielded a prevalence of 41% (35–46), while, in studies that calculated mathematical estimates of CrCl, the pooled prevalence was 23% (11–43), showing a stark underestimation in the case of calculated CrCl (Supplementary Figure S1). To assess the risk of publication bias, a funnel plot was used to visualize the individual studies’ effect sizes against their estimate of precision (Figure 5). Egger’s test [26] was conducted to test for funnel plot’s asymmetry; the result was insignificant (p-value > 0.05), suggesting no publication bias.

3.3. ARC Risk Factors

Reported risk factors included in the meta-analysis were age (as a continuous variable), male sex, trauma, SOFA and APACHEII disease severity scores, and diabetes. Among the reported risk factors, age, male sex and trauma were significantly associated with ARC with pooled odds ratio (95% CI) estimates of 0.95 (0.93–0.96), 2.36 (1.28–4.36), and 2.60 (1.21–5.58), respectively (Figure 6). SOFA, APACHEII and diabetes were not significantly associated with ARC, with pooled odds ratio (95% CI) estimates of 0.86 (0.73–1.01), 1.00 (0.95–1.06) and 1.21 (0.46–3.17), respectively (Supplementary Figure S2).

4. Discussion

ARC is a phenomenon wherein renal clearance is accelerated beyond normal range; it has also been referred to as glomerular hyperfiltration or enhanced renal clearance. ARC bears the risk of causing therapeutic failure of predominantly renally cleared drugs, which could be especially detrimental in critically ill populations. Numerous studies have described the association between ARC and higher rates of failure to attain therapeutic levels and compromised effectiveness of various drugs and the need for a more frequent administration and/or higher dosages. Standard doses of renally-eliminated medications are typically used in patients with “normal” renal function. However, pharmacodynamic targets that are consistently obtained in other populations with typical dosing are not met in the presence of ARC. Studies have suggested that ARC might be associated with subtherapeutic concentrations of antimicrobials and AEDs, [33,77,78,91] antimicrobial therapy failure, [40] increased odds of recurrent infections, [18] and poor seizure control [92]. Our systematic review and meta-analysis demonstrated the common occurrence of ARC in critical care settings, with higher prevalence among neurocritical care and trauma patients compared to mixed ICU population. In addition, risk factors consistently found to be associated with ARC includes age, male sex, and trauma. The differences in the pooled ARC prevalence demonstrated that different critically ill populations were not at an equivalent risk for ARC and highlighted the importance of screening for ARC in select patient populations, as well as the need to develop new screening tools that account for these risk differences. To our knowledge, this is the first combined systematic review and meta-analysis of the prevalence and risk factors of ARC.
In our random effects meta-analysis for ARC prevalence, patients in the neurocritical care population demonstrated the highest prevalence of ARC (74%). ARC incidence has been reported to range much higher in neurocritical care patients compared to the general critically ill population [4,11,12,13,14,15,16,17,18]. To illustrate, in a study of 20 traumatic brain injury (TBI) patients, 85% showed ARC [14]. In a study of patients with hemorrhagic stroke, ARC was reported in 50% of intracerebral hemorrhage (ICH) (n = 30) and 94 % of subarachnoid hemorrhage (n = 50) patients [16]. In addition, ICH was found to predict ARC in a retrospective study of heterogenous ICU patients, supporting the notion that neurological injury poses additional ARC risk [88]. This could be attributed to the possibility that patients with neurological injuries might have additional ARC risks. Neurocritical care patients tend to be relatively younger patients with single comorbidities and otherwise unimpaired organ systems, as well as a lower incidence of renal impairment. Furthermore, neurological injury could play an additional role in the pathophysiology of ARC; however, further studies are needed to confirm such association [47,61].
The employment of an accurate determination method for glomerular filtration rate is essential for ARC screening and diagnosis. Although using serum creatinine to assess kidney function carries limitations, CrCl measurement using 8-24h urine collection is the most agreed upon accurate method for the measurement of renal function in the clinical setting without the need of administrating an exogenous substance such as inulin. Moreover, due to the impracticality of routine and frequent measurement of CrCl in clinical settings, calculating CrCl using mathematical estimations derived from population parameters is often employed to allow for a more rapid determination. Commonly used formulae used to draw mathematical estimates of CrCl include the Cockcroft–Gault equation (CG), modification of diet in renal diseases (MDRD), and chronic kidney disease-epidemiology (CKD-EPI). Each of those methods possess their own merits and downfalls. Several studies assessed the relative accuracy of different mathematical estimates of CrCl in patients exhibiting ARC. It has been found that all mathematical estimations of CrCl grossly underestimate the actual CrCl when compared with their respective measured CrCl in patients with ARC [31,38,41,44,45,49,54,93,94,95]. Similarly, we found that the mathematical estimations of CrCl grossly underestimated the prevalence in ARC when compared to measured CrCl. To illustrate, the meta-analysis of prevalence of ARC in the same population (mixed ICU patients) was 23% in studies using mathematical estimates, whereas studies using measured CrCl showed a 41% prevalence. Therefore, we recommend obtaining a patient’s measured CrCl at least once on admission for a more judicious assessment if they are at risk for ARC. Special consideration must also be taken in immobile patients, children, burn patients or patients with conditions causing lower muscle mass or amputations to account for the reduced production of creatinine in these cases which could result in falsely low serum creatinine levels leading to incorrect diagnosis of augmented renal clearance.
It has been consistently shown in studies reporting risk factors of ARC that ARC patients tend to be younger males (<50 years old) with lower critical illness severity scores. These patients also tend to suffer from single organ impairment with unimpaired kidney function and a history of recent trauma. In our analysis, among the reported risk factors, age, male sex, and trauma were significantly associated with ARC with pooled odds ratio (95% CI) estimates of 0.95 (0.93–0.96), 2.36 (1.28–4.36), and 2.60 (1.21–5.58), respectively. The aforementioned risk factors have been utilized to develop clinical prediction tools needed for the early identification of patients at a higher risk for developing ARC. An ARC scoring system with 60% sensitivity and 95% specificity was introduced by Baptista et al. [96], where urinary creatinine higher than 45 mg/mL, age less than 65 years, and blood urea nitrogen (BUN) less than 7 mmol/L serve as predictors of ARC. Moreover, Udy et al. developed a scoring system that is based on age less than 50 years old, history of recent trauma, and SOFA score ≤ 4 [36]. This tool demonstrated 100% sensitivity and 71% specificity when validated by Akers et al. [97]. Furthermore, Barletta et al. [60] developed the augmented renal clearance in trauma intensive care (ARCTIC) scoring system, which eliminated the need to calculate a SOFA score in order to assess the patients’ risk for developing ARC, which can be impractical in some patient settings. The risk factors employed in the assessment tool were serum creatinine, sex and age; it stratified patients into high risk (ARCTIC score ≥ 6) and low risk (ARCTIC score < 6). Employing predictive tools such as ARC or ARCTIC in routine screening of critically ill patients could be valuable in the way of early recognition and timely management of ARC patients. However, the developed scoring tools were generated based on the general critically ill/trauma population rather than patients with severe neurological illnesses, potentially not capturing neurocritical care patients with additional risks for ARC.
Our systematic review was limited by the characteristics of the included studies. The main body of evidence comes from retrospective observational studies, which require caution in the interpretation of results. In addition, heterogeneity of the included studies was high secondary to variations in study populations, ARC definitions, the method of determining CrCl, studies inclusion and exclusion criteria may impede accurate comparisons among studies. For example, 65% of the studies in the meta-analysis excluded patients with existing acute and/or chronic renal impairment with various stages, impeding the possibility of extrapolating their results outside of the sampling context, as well as overestimating ARC occurrence in these samples compared to others where patients with renal impairment were included [9,18,59,88]. This highlights the need for unified assessment of ARC in future research. However, in our analysis, we took into consideration the heterogeneity of the included studies; our pooled estimates are a reasonable representation of the body of literature.

5. Conclusions

ARC is a prevalent phenomenon in critically ill patients especially neurocritical care and trauma ICU population. Young age, male sex, and trauma are risk factors for ARC in those with apparently normal renal function. The estimation of CrCl using mathematical estimates of GFR grossly underestimates the prevalence of ARC in the critical care setting; therefore measured CrCl through urine collections is prudent. Further research on optimal dosing of drugs in the setting of ARC is warranted.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/pharmaceutics14020445/s1, Table S1: Full search strategy; Table S2: Appraisal of individual studies included in this review; Figure S1: Forest plot of the prevalence of ARC in mixed intensive care unit (ICU) population. A, studies reported measured creatinine clearance (m); B, studies reported calculated creatinine clearance (c). Figure S2: Forest plot of risk factors of augmented renal clearance. A, diabetes; B, Sequential Organ Failure Assessment (SOFA) score; C, Acute Physiology and Chronic Health Evaluation (APACHE II).

Author Contributions

Conceptualization, S.H.M.; methodology, S.H.M. and J.Y.K.; study screening and data collection, F.H., S.H.M. and A.S.; data analysis, F.H.; writing—original draft preparation, F.H. and A.S.; writing—review and editing, All authors; supervision, S.H.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data is contained within this article and the associated supplementary materials.

Acknowledgments

We would like to acknowledge Nouran Hamza, BSc, PhD Candidate Biostatistics, for her valuable contribution to statistical analysis methodology proofing.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bakke, V.; Sporsem, H.; Von der Lippe, E.; Nordoy, I.; Lao, Y.; Nyrerod, H.C.; Sandvik, L.; Harvig, K.R.; Bugge, J.F.; Helset, E. Vancomycin levels are frequently subtherapeutic in critically ill patients: A prospective observational study. Acta Anaesthesiol. Scand. 2017, 61, 627–635. [Google Scholar] [CrossRef] [PubMed]
  2. Carrie, C.; Petit, L.; d’Houdain, N.; Sauvage, N.; Cottenceau, V.; Lafitte, M.; Foumenteze, C.; Hisz, Q.; Menu, D.; Legeron, R.; et al. Association between augmented renal clearance, antibiotic exposure and clinical outcome in critically ill septic patients receiving high doses of beta-lactams administered by continuous infusion: A prospective observational study. Int. J. Antimicrob. Agents 2018, 51, 443–449. [Google Scholar] [CrossRef] [PubMed]
  3. Hirai, K.; Ishii, H.; Shimoshikiryo, T.; Shimomura, T.; Tsuji, D.; Inoue, K.; Kadoiri, T.; Itoh, K. Augmented Renal Clearance in Patients with Febrile Neutropenia is Associated with Increased Risk for Subtherapeutic Concentrations of Vancomycin. Ther. Drug Monit. 2016, 38, 706–710. [Google Scholar] [CrossRef]
  4. Mahmoud, S.H.; Shen, C. Augmented Renal Clearance in Critical Illness: An Important Consideration in Drug Dosing. Pharmaceutics 2017, 9, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Udy, A.A.; Roberts, J.A.; Lipman, J. Implications of augmented renal clearance in critically ill patients. Nat. Rev. Nephrol. 2011, 7, 539–543. [Google Scholar] [CrossRef]
  6. Sime, F.B.; Udy, A.A.; Roberts, J.A. Augmented renal clearance in critically ill patients: Etiology, definition and implications for beta-lactam dose optimization. Curr. Opin. Pharmacol. 2015, 24, 1–6. [Google Scholar] [CrossRef]
  7. Gijsen, M.; Huang, C.Y.; Flechet, M.; Van Daele, R.; Declercq, P.; Debaveye, Y.; Meersseman, P.; Meyfroidt, G.; Wauters, J.; Spriet, I. Development and External Validation of an Online Clinical Prediction Model for Augmented Renal Clearance in Adult Mixed Critically Ill Patients: The Augmented Renal Clearance Predictor. Crit. Care Med. 2020, 48, e1260–e1268. [Google Scholar] [CrossRef]
  8. Gijsen, M.; Wilmer, A.; Meyfroidt, G.; Wauters, J.; Spriet, I. Can augmented renal clearance be detected using estimators of glomerular filtration rate? Crit. Care 2020, 24, 359. [Google Scholar] [CrossRef]
  9. Baptista, J.P. Augmented renal clearance. In Antibiotic Pharmacokinetic/Pharmacodynamic Considerations in the Critically Ill; Springer: Singapore, 2017; pp. 125–150. [Google Scholar]
  10. Bilbao-Meseguer, I.; Rodriguez-Gascon, A.; Barrasa, H.; Isla, A.; Solinis, M.A. Augmented Renal Clearance in Critically Ill Patients: A Systematic Review. Clin. Pharmacokinet. 2018, 57, 1107–1121. [Google Scholar] [CrossRef]
  11. Udy, A.A.; Roberts, J.A.; Boots, R.J.; Paterson, D.L.; Lipman, J. Augmented renal clearance: Implications for antibacterial dosing in the critically ill. Clin. Pharmacokinet. 2010, 49, 1–16. [Google Scholar] [CrossRef]
  12. Udy, A.A.; Putt, M.T.; Boots, R.J.; Lipman, J. ARC—Augmented renal clearance. Curr. Pharm. Biotechnol. 2011, 12, 2020–2029. [Google Scholar] [CrossRef] [PubMed]
  13. Hobbs, A.L.; Shea, K.M.; Roberts, K.M.; Daley, M.J. Implications of Augmented Renal Clearance on Drug Dosing in Critically Ill Patients: A Focus on Antibiotics. Pharmacotherapy 2015, 35, 1063–1075. [Google Scholar] [CrossRef] [PubMed]
  14. Udy, A.; Boots, R.; Senthuran, S.; Stuart, J.; Deans, R.; Lassig-Smith, M.; Lipman, J. Augmented Creatinine Clearance in Traumatic Brain Injury. Anesth. Analg. 2010, 111, 1505–1510. [Google Scholar] [CrossRef] [PubMed]
  15. May, C.C.; Arora, S.; Parli, S.E.; Fraser, J.F.; Bastin, M.T.; Cook, A.M. Augmented Renal Clearance in Patients with Subarachnoid Hemorrhage. Neurocrit. Care 2015, 23, 374–379. [Google Scholar] [CrossRef] [PubMed]
  16. Morbitzer, K.A.; Jordan, J.D.; Dehne, K.A.; Durr, E.A.; Olm-Shipman, C.M.; Rhoney, D.H. Enhanced Renal Clearance in Patients With Hemorrhagic Stroke. Crit. Care Med. 2019, 47, 800–808. [Google Scholar] [CrossRef]
  17. Lautrette, A.; Phan, T.-N.; Ouchchane, L.; AitHssain, A.; Tixier, V.; Heng, A.-E.; Souweine, B. High creatinine clearance in critically ill patients with community-acquired acute infectious meningitis. BMC Nephrol. 2012, 13, 124. [Google Scholar] [CrossRef] [Green Version]
  18. Carrie, C.; Bentejac, M.; Cottenceau, V.; Masson, F.; Petit, L.; Cochard, J.F.; Sztark, F. Association between augmented renal clearance and clinical failure of antibiotic treatment in brain-injured patients with ventilator-acquired pneumonia: A preliminary study. Anaesth. Crit. Care Pain Med. 2018, 37, 35–41. [Google Scholar] [CrossRef]
  19. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. J. Clin. Epidemiol. 2021, 88, 105906. [Google Scholar] [CrossRef]
  20. Balduzzi, S.; Rucker, G.; Schwarzer, G. How to perform a meta-analysis with R: A practical tutorial. Evid. Based Ment. Health 2019, 22, 153–160. [Google Scholar] [CrossRef] [Green Version]
  21. R Development Core Team. R: A Language and Environment for Statistical Computing; 4.0.3 (2020-10-10); R Foundation for Statistical Computing: Vienna, Austria, 2020. [Google Scholar]
  22. RStudio Team. RStudio: Integrated Development for R; RStudio: Boston, MA, USA, 2020. [Google Scholar]
  23. DerSimonian, R.; Laird, N. Meta-analysis in clinical trials revisited. Contemp. Clin. Trials 2015, 45, 139–145. [Google Scholar] [CrossRef] [Green Version]
  24. DerSimonian, R.; Laird, N. Meta-analysis in clinical trials. Control Clin. Trials 1986, 7, 177–188. [Google Scholar] [CrossRef]
  25. Jackson, D.; White, I.R.; Thompson, S.G. Extending DerSimonian and Laird’s methodology to perform multivariate random effects meta-analyses. Stat. Med. 2010, 29, 1282–1297. [Google Scholar] [CrossRef] [PubMed]
  26. Egger, M.; Davey Smith, G.; Schneider, M.; Minder, C. Bias in meta-analysis detected by a simple, graphical test. BMJ 1997, 315, 629–634. [Google Scholar] [CrossRef] [Green Version]
  27. Udy, A.A.; Dulhunty, J.M.; Roberts, J.A.; Davis, J.S.; Webb, S.A.R.; Bellomo, R.; Gomersall, C.; Shirwadkar, C.; Eastwood, G.M.; Myburgh, J.; et al. Association between augmented renal clearance and clinical outcomes in patients receiving beta-lactam antibiotic therapy by continuous or intermittent infusion: A nested cohort study of the BLING-II randomised, placebo-controlled, clinical trial. Int. J. Antimicrob. Agents 2017, 49, 624–630. [Google Scholar] [CrossRef]
  28. Cojutti, P.G.; Lazzarotto, D.; Candoni, A.; Dubbini, M.V.; Zannier, M.E.; Fanin, R.; Pea, F. Real-time TDM-based optimization of continuous-infusion meropenem for improving treatment outcome of febrile neutropenia in oncohaematological patients: Results from a prospective, monocentric, interventional study. J. Antimicrob. Chemother. 2020, 75, 3029–3037. [Google Scholar] [CrossRef]
  29. Joynt, G.M.; Lipman, J.; Gomersall, C.D.; Young, R.J.; Wong, E.L.; Gin, T. The pharmacokinetics of once-daily dosing of ceftriaxone in critically ill patients. J. Antimicrob. Chemother. 2001, 47, 421–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Fuster-Lluch, O.; Geronimo-Pardo, M.; Peyro-Garcia, R.; Lizan-Garcia, M. Glomerular hyperfiltration and albuminuria in critically ill patients. Anaesth. Intensive Care 2008, 36, 674–680. [Google Scholar] [CrossRef] [Green Version]
  31. Baptista, J.P.; Udy, A.A.; Sousa, E.; Pimentel, J.; Wang, L.; Roberts, J.A.; Lipman, J. A comparison of estimates of glomerular filtration in critically ill patients with augmented renal clearance. Crit. Care 2011, 15, R139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Minville, V.; Asehnoune, K.; Ruiz, S.; Breden, A.; Georges, B.; Seguin, T.; Tack, I.; Jaafar, A.; Saivin, S.; Fourcade, O.; et al. Increased creatinine clearance in polytrauma patients with normal serum creatinine: A retrospective observational study. Crit. Care 2011, 15, R49. [Google Scholar] [CrossRef] [Green Version]
  33. Baptista, J.P.; Sousa, E.; Martins, P.J.; Pimentel, J.M. Augmented renal clearance in septic patients and implications for vancomycin optimisation. Int. J. Antimicrob. Agents 2012, 39, 420–423. [Google Scholar] [CrossRef]
  34. Grootaert, V.; Spriet, I.; Decoutere, L.; Debaveye, Y.; Meyfroidt, G.; Willems, L. Augmented renal clearance in the critically ill: Fiction or fact? Int. J. Clin. Pharm. 2012, 34, 143. [Google Scholar] [CrossRef]
  35. Carlier, M.; Carrette, S.; Roberts, J.A.; Stove, V.; Verstraete, A.; Hoste, E.; Depuydt, P.; Decruyenaere, J.; Lipman, J.; Wallis, S.C.; et al. Meropenem and piperacillin/tazobactam prescribing in critically ill patients: Does augmented renal clearance affect pharmacokinetic/pharmacodynamic target attainment when extended infusions are used? Crit. Care 2013, 17, R84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Udy, A.A.; Roberts, J.A.; Shorr, A.F.; Boots, R.J.; Lipman, J. Augmented renal clearance in septic and traumatized patients with normal plasma creatinine concentrations: Identifying at-risk patients. Crit. Care 2013, 17, R35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Minkute, R.; Briedis, V.; Steponaviciute, R.; Vitkauskiene, A.; Maciulaitis, R. Augmented renal clearance—An evolving risk factor to consider during the treatment with vancomycin. J. Clin. Pharm. Ther. 2013, 38, 462–467. [Google Scholar] [CrossRef] [PubMed]
  38. Udy, A.A.; Morton, F.J.A.; Nguyen-Pham, S.; Jarrett, P.; Lassig-Smith, M.; Stuart, J.; Dunlop, R.; Starr, T.; Boots, R.J.; Lipman, J. A comparison of CKD-EPI estimated glomerular filtration rate and measured creatinine clearance in recently admitted critically ill patients with normal plasma creatinine concentrations. BMC Nephrol. 2013, 14, 250. [Google Scholar] [CrossRef] [Green Version]
  39. Claus, B.O.M.; Hoste, E.A.; Colpaert, K.; Robays, H.; Decruyenaere, J.; De Waele, J.J. Augmented renal clearance is a common finding with worse clinical outcome in critically ill patients receiving antimicrobial therapy. J. Crit. Care 2013, 28, 695–700. [Google Scholar] [CrossRef]
  40. Baptista, J.P.; Roberts, J.A.; Sousa, E.; Freitas, R.; Deveza, N.; Pimentel, J. Decreasing the time to achieve therapeutic vancomycin concentrations in critically ill patients: Developing and testing of a dosing nomogram. Crit. Care 2014, 18, 654. [Google Scholar] [CrossRef] [Green Version]
  41. Baptista, J.P.; Neves, M.; Rodrigues, L.; Teixeira, L.; Pinho, J.; Pimentel, J. Accuracy of the estimation of glomerular filtration rate within a population of critically ill patients. J. Nephrol. 2014, 27, 403–410. [Google Scholar] [CrossRef]
  42. Campassi, M.L.; Gonzalez, M.C.; Masevicius, F.D.; Vazquez, A.R.; Moseinco, M.; Navarro, N.C.; Previgliano, L.; Rubatto, N.P.; Benites, M.H.; Estenssoro, E.; et al. Augmented renal clearance in critically ill patients: Incidence, associated factors and effects on vancomycin treatment. Rev. Bras. Ter. Intensiva 2014, 26, 13–20. [Google Scholar] [CrossRef]
  43. Udy, A.A.; Baptista, J.P.; Lim, N.L.; Joynt, G.M.; Jarrett, P.; Wockner, L.; Boots, R.J.; Lipman, J. Augmented Renal Clearance in the ICU: Results of a Multicenter Observational Study of Renal Function in Critically Ill Patients with Normal Plasma Creatinine Concentrations. Crit. Care Med. 2014, 42, 520–527. [Google Scholar] [CrossRef]
  44. Adnan, S.; Ratnam, S.; Kumar, S.; Paterson, D.; Lipman, J.; Roberts, J.; Udy, A.A. Select critically ill patients at risk of augmented renal clearance: Experience in a Malaysian intensive care unit. Anaesth. Intensive Care 2014, 42, 715–722. [Google Scholar] [CrossRef]
  45. Ruiz, S.; Minville, V.; Asehnoune, K.; Virtos, M.; Georges, B.; Fourcade, O.; Conil, J.M. Screening of patients with augmented renal clearance in ICU: Taking into account the CKD-EPI equation, the age, and the cause of admission. Ann. Intensive Care 2015, 5, 49. [Google Scholar] [CrossRef] [Green Version]
  46. Huttner, A.; Von Dach, E.; Renzoni, A.; Huttner, B.D.; Affaticati, M.; Pagani, L.; Daali, Y.; Pugin, J.; Karmime, A.; Fathi, M.; et al. Augmented renal clearance, low beta-lactam concentrations and clinical outcomes in the critically ill: An observational prospective cohort study. Int. J. Antimicrob. Agents 2015, 45, 385–392. [Google Scholar] [CrossRef]
  47. Dias, C.; Gaio, A.R.; Monteiro, E.; Barbosa, S.; Cerejo, A.; Donnelly, J.; Felgueiras, O.; Smielewski, P.; Paiva, J.A.; Czosnyka, M. Kidney-brain link in traumatic brain injury patients? A preliminary report. Neurocrit. Care 2015, 22, 192–201. [Google Scholar] [CrossRef] [PubMed]
  48. De Waele, J.J.; Dumoulin, A.; Janssen, A.; Hoste, E.A. Epidemiology of augmented renal clearance in mixed ICU patients. Minerva Anestesiol. 2015, 81, 1079–1085. [Google Scholar]
  49. Steinke, T.; Moritz, S.; Beck, S.; Gnewuch, C.; Kees, M.G. Estimation of creatinine clearance using plasma creatinine or cystatin C: A secondary analysis of two pharmacokinetic studies in surgical ICU patients. BMC Anesthesiol. 2015, 15, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Chu, Y.; Luo, Y.; Qu, L.; Zhao, C.; Jiang, M. Application of vancomycin in patients with varying renal function, especially those with augmented renal clearance. Pharmaceut. Biol. 2016, 54, 2802–2806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Kawano, Y.; Morimoto, S.; Izutani, Y.; Muranishi, K.; Kaneyama, H.; Hoshino, K.; Nishida, T.; Ishikura, H. Augmented renal clearance in Japanese intensive care unit patients: A prospective study. J. Intensive Care 2016, 4, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Saour, M.; Klouche, K.; Deras, P.; Damou, A.; Capdevila, X.; Charbit, J. Assessment of modification of diet in renal disease equation to predict reference serum creatinine value in severe trauma patients: Lessons from an obser vational study of 775 cases. Ann. Surg. 2016, 263, 814–820. [Google Scholar] [CrossRef]
  53. Abdel El Naeem, H.E.M.; Abdelhamid, M.H.E.; Atteya, D.A.M. Impact of augmented renal clearance on enoxaparin therapy in critically ill patients. Egypt. J. Anaesth. 2017, 33, 113–117. [Google Scholar] [CrossRef] [Green Version]
  54. Barletta, J.F.; Mangram, A.J.; Byrne, M.; Hollingworth, A.K.; Sucher, J.F.; Ali-Osman, F.R.; Shirah, G.R.; Dzandu, J.K. The importance of empiric antibiotic dosing in critically ill trauma patients: Are we under-dosing based on augmented renal clearance and inaccurate renal clearance estimates? J. Trauma Acute Care Surg. 2016, 81, 1115–1121. [Google Scholar] [CrossRef] [Green Version]
  55. Declercq, P.; Nijs, S.; D’Hoore, A.; Van Wijngaerden, E.; Wolthuis, A.; de Buck van Overstraeten, A.; Wauters, J.; Spriet, I. Augmented renal clearance in non-critically ill abdominal and trauma surgery patients is an underestimated phenomenon: A point prevalence study. J. Trauma Acute Care Surg. 2016, 81, 468–477. [Google Scholar] [CrossRef] [PubMed]
  56. Ehmann, L.; Zoller, M.; Minichmayr, I.K.; Scharf, C.; Maier, B.; Schmitt, M.V.; Hartung, N.; Huisinga, W.; Vogeser, M.; Frey, L.; et al. Role of renal function in risk assessment of target non-attainment after standard dosing of meropenem in critically ill patients: A prospective observational study. Crit. Care 2017, 21, 263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Burnham, J.P.; Micek, S.T.; Kollef, M.H. Augmented renal clearance is not a risk factor for mortality in Enterobacteriaceae bloodstream infections treated with appropriate empiric antimicrobials. PLoS ONE 2017, 12, e0180247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Carrie, C.; Lannou, A.; Rubin, S.; De Courson, H.; Petit, L.; Biais, M. Augmented renal clearance in critically ill trauma patients: A pathophysiologic approach using renal vascular index. Anaesth. Crit. Care Pain Med. 2019, 38, 371–375. [Google Scholar] [CrossRef] [PubMed]
  59. Udy, A.A.; Jarrett, P.; Lassig-Smith, M.; Stuart, J.; Starr, T.; Dunlop, R.; Deans, R.; Roberts, J.A.; Senthuran, S.; Boots, R.; et al. Augmented Renal Clearance in Traumatic Brain Injury: A Single-Center Observational Study of Atrial Natriuretic Peptide, Cardiac Output, and Creatinine Clearance. J. Neurotrauma 2017, 34, 137–144. [Google Scholar] [CrossRef]
  60. Barletta, J.F.; Mangram, A.J.; Byrne, M.; Sucher, J.F.; Hollingworth, A.K.; Ali-Osman, F.R.; Shirah, G.R.; Haley, M.; Dzandu, J.K. Identifying augmented renal clearance in trauma patients: Validation of the Augmented Renal Clearance in Trauma Intensive Care scoring system. J. Trauma Acute Care Surg. 2017, 82, 665–671. [Google Scholar] [CrossRef]
  61. Dhaese, S.A.M.; Roberts, J.A.; Carlier, M.; Verstraete, A.G.; Stove, V.; De Waele, J.J. Population pharmacokinetics of continuous infusion of piperacillin in critically ill patients. Int. J. Antimicrob. Agents 2018, 51, 594–600. [Google Scholar] [CrossRef] [Green Version]
  62. Tamatsukuri, T.; Ohbayashi, M.; Kohyama, N.; Kobayashi, Y.; Yamamoto, T.; Fukuda, K.; Nakamura, S.; Miyake, Y.; Dohi, K.; Kogo, M. The exploration of population pharmacokinetic model for meropenem in augmented renal clearance and investigation of optimum setting of dose. J. Infect. Chemother. Off. J. Jpn. Soc. Chemother. 2018, 24, 834–840. [Google Scholar] [CrossRef]
  63. Carrie, C.; Legeron, R.; Petit, L.; Ollivier, J.; Cottenceau, V.; d’Houdain, N.; Boyer, P.; Lafitte, M.; Xuereb, F.; Sztark, F.; et al. Higher than standard dosing regimen are needed to achieve optimal antibiotic exposure in critically ill patients with augmented renal clearance receiving piperacillin-tazobactam administered by continuous infusion. J. Crit. Care 2018, 48, 66–71. [Google Scholar] [CrossRef]
  64. Kawano, Y.; Maruyama, J.; Hokama, R.; Koie, M.; Nagashima, R.; Hoshino, K.; Muranishi, K.; Nakashio, M.; Nishida, T.; Ishikura, H. Outcomes in patients with infections and augmented renal clearance: A multicenter retrospective study. PLoS ONE 2018, 13, e0208742. [Google Scholar] [CrossRef] [Green Version]
  65. Tsai, D.; Udy, A.A.; Stewart, P.C.; Gourley, S.; Morick, N.M.; Lipman, J.; Roberts, J.A. Prevalence of augmented renal clearance and performance of glomerular filtration estimates in Indigenous Australian patients requiring intensive care admission. Anaesth. Intensive Care 2018, 46, 42–50. [Google Scholar] [CrossRef] [Green Version]
  66. Wong, G.; Briscoe, S.; McWhinney, B.; Ally, M.; Ungerer, J.; Lipman, J.; Roberts, J.A. Therapeutic drug monitoring of beta-lactam antibiotics in the critically ill: Direct measurement of unbound drug concentrations to achieve appropriate drug exposures. J. Antimicrob. Chemother. 2018, 73, 3087–3094. [Google Scholar] [CrossRef] [Green Version]
  67. Ishii, H.; Hirai, K.; Sugiyama, K.; Nakatani, E.; Kimura, M.; Itoh, K. Validation of a Nomogram for Achieving Target trough Concentration of Vancomycin: Accuracy in Patients with Augmented Renal Function. Ther. Drug Monit. 2018, 40, 693–698. [Google Scholar] [CrossRef]
  68. Ollivier, J.; Carrie, C.; d’Houdain, N.; Djabarouti, S.; Petit, L.; Xuereb, F.; Legeron, R.; Biais, M.; Breilh, D. Are Standard Dosing Regimens of Ceftriaxone Adapted for Critically Ill Patients with Augmented Creatinine Clearance? Antimicrob. Agents Chemother. 2019, 63, e02134-18. [Google Scholar] [CrossRef] [Green Version]
  69. Wu, C.C.; Tai, C.H.; Liao, W.Y.; Wang, C.C.; Kuo, C.H.; Lin, S.W.; Ku, S.C. Augmented renal clearance is associated with inadequate antibiotic pharmacokinetic/pharmacodynamic target in Asian ICU population: A prospective observational study. Infect. Drug Resist. 2019, 12, 2531–2541. [Google Scholar] [CrossRef] [Green Version]
  70. Aitullina, A.; Krumina, A.; Purvina, S. Augmented clearance in patients with colistin therapy in intensive care units. Int. J. Clin. Pharm. 2019, 41, 310. [Google Scholar] [CrossRef]
  71. Weber, N.; Jackson, K.; McWhinney, B.; Ungerer, J.; Kennedy, G.; Lipman, J.; Roberts, J.A. Evaluation of pharmacokinetic/pharmacodynamic and clinical outcomes with 6-hourly empiric piperacillin-tazobactam dosing in hematological malignancy patients with febrile neutropenia. J. Infect. Chemother. Off. J. Jpn. Soc. Chemother. 2019, 25, 503–508. [Google Scholar] [CrossRef]
  72. Izumisawa, T.; Kaneko, T.; Soma, M.; Imai, M.; Wakui, N.; Hasegawa, H.; Horino, T.; Takahashi, N. Augmented Renal Clearance of Vancomycin in Hematologic Malignancy Patients. Biol. Pharm. Bull. 2019, 42, 2089–2094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Chu, Y.; Luo, Y.; Jiang, M.; Zhou, B. Application of vancomycin in patients with augmented renal clearance. Eur. J. Hosp. Pharm. Sci. Pract. 2020, 27, 276–279. [Google Scholar] [CrossRef] [PubMed]
  74. Villanueva, R.D.; Talledo, O.; Neely, S.; White, B.; Celii, A.; Cross, A.; Kennedy, R. Vancomycin dosing in critically ill trauma patients: The VANCTIC Study. J. Trauma Acute Care Surg. 2019, 87, 1164–1171. [Google Scholar] [CrossRef] [PubMed]
  75. Morbitzer, K.A.; Rhoney, D.H.; Dehne, K.A.; Jordan, J.D. Enhanced renal clearance and impact on vancomycin pharmacokinetic parameters in patients with hemorrhagic stroke. J. Intensive Care 2019, 7, 51. [Google Scholar] [CrossRef] [PubMed]
  76. Mulder, M.B.; Eidelson, S.A.; Sussman, M.S.; Schulman, C.I.; Lineen, E.B.; Iyenger, R.S.; Namias, N.; Proctor, K.G. Risk Factors and Clinical Outcomes Associated with Augmented Renal Clearance in Trauma Patients. J. Surg. Res. 2019, 244, 477–483. [Google Scholar] [CrossRef] [PubMed]
  77. Bricheux, A.; Lenggenhager, L.; Hughes, S.; Karmime, A.; Lescuyer, P.; Huttner, A. Therapeutic drug monitoring of imipenem and the incidence of toxicity and failure in hospitalized patients: A retrospective cohort study. Clin. Microbiol. Infect. Off. Publ. Eur. Soc. Clin. Microbiol. Infect. Dis. 2019, 25, 383.e381–383.e384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Helset, E.; Nordøy, I.; Sporsem, H.; Bakke, V.D.; Bugge, J.F.; Gammelsrud, K.W.; Zucknick, M.; Lippe, E.; von der Lippe, E. Factors increasing the risk of inappropriate vancomycin therapy in ICU patients: A prospective observational study. Acta Anaesthesiol. Scand. 2020, 64, 1295–1304. [Google Scholar] [CrossRef]
  79. Barrasa, H.; Soraluce, A.; Uson, E.; Sainz, J.; Martin, A.; Sanchez-Izquierdo, J.A.; Maynar, J.; Rodriguez-Gascon, A.; Isla, A. Impact of augmented renal clearance on the pharmacokinetics of linezolid: Advantages of continuous infusion from a pharmacokinetic/pharmacodynamic perspective. Int. J. Infect. Dis. Off. Publ. Int. Soc. Infect. Dis. 2020, 93, 329–338. [Google Scholar] [CrossRef]
  80. Lannou, A.; Carrie, C.; Rubin, S.; Cane, G.; Cottenceau, V.; Petit, L.; Biais, M. Salt wasting syndrome in brain trauma patients: A pathophysiologic approach using sodium balance and urinary biochemical analysis. BMC Neurol. 2020, 20, 190. [Google Scholar] [CrossRef]
  81. Arechiga-Alvarado, N.A.; Medellin-Garibay, S.E.; Milan-Segovia, R.D.C.; Ortiz-Alvarez, A.; Magana-Aquino, M.; Romano-Moreno, S. Population Pharmacokinetics of Amikacin Administered Once Daily in Patients with Different Renal Functions. Antimicrob. Agents Chemother. 2020, 64, e02178-19. [Google Scholar] [CrossRef]
  82. Carrie, C.; Delzor, F.; Roure, S.; Dubuisson, V.; Petit, L.; Molimard, M.; Breilh, D.; Biais, M. Population Pharmacokinetic Study of the Suitability of Standard Dosing Regimens of Amikacin in Critically Ill Patients with Open-Abdomen and Negative-Pressure Wound Therapy. Antimicrob. Agents Chemother. 2020, 64, e02098-19. [Google Scholar] [CrossRef]
  83. Saito, K.; Kamio, S.; Ito, K.; Suzuki, N.; Abe, K.; Goto, T. A simple scoring method to predict augmented renal clearance in haematologic malignancies. J. Clin. Pharm. Ther. 2020, 45, 1120–1126. [Google Scholar] [CrossRef]
  84. Lannou, A.; Carrie, C.; Rubin, S.; De Courson, H.; Biais, M. Renal response after traumatic brain injury: A pathophysiological relationship between augmented renal clearance and salt wasting syndrome? Anaesth. Crit. Care Pain Med. 2020, 39, 239–241. [Google Scholar] [CrossRef]
  85. Brown, A.; Lavelle, R.; Gerlach, A. Discordance of renal drug dosing using estimated creatinine clearance and measured urine creatinine clearance in hospitalized adults: A retrospective cohort study. Int. J. Crit. Illn. Inj. Sci. 2020, 10, S1–S5. [Google Scholar] [CrossRef]
  86. Chen, Y.; Liu, L.; Zhu, M. Effect of augmented renal clearance on the therapeutic drug monitoring of vancomycin in patients after neurosurgery. J. Int. Med. Res. 2020, 48, 300060520949076. [Google Scholar] [CrossRef] [PubMed]
  87. Baptista, J.P.; Martins, P.J.; Marques, M.; Pimentel, J.M. Prevalence and Risk Factors for Augmented Renal Clearance in a Population of Critically Ill Patients. J. Intensive Care Med. 2020, 35, 1044–1052. [Google Scholar] [CrossRef] [PubMed]
  88. Nei, A.M.; Kashani, K.B.; Dierkhising, R.; Barreto, E.F. Predictors of Augmented Renal Clearance in a Heterogeneous ICU Population as Defined by Creatinine and Cystatin C. Nephron 2020, 144, 313–320. [Google Scholar] [CrossRef]
  89. Ramos, A.; Acharta, F.; Perezlindo, M.; Lovesio, L.; Gauna Antonelli, P.; Dogliotti, A.; Lovesio, C. Factors that predict supranormal glomerular filtration in critical diseases. In Proceedings of the 37th International Symposium on Intensive Care and Emergency Medicine, Brussels, Belgium, 21–24 March 2017; Volume 21. [Google Scholar] [CrossRef] [Green Version]
  90. Eidelson, S.A.; Mulder, M.B.; Rattan, R.; Karcutskie, C.A.; Meizoso, J.P.; Madiraju, S.K.; Lineen, E.B.; Schulman, C.I.; Namias, N. Incidence and Functional Significance of Augmented Renal Clearance in Trauma Patients at High Risk for Venous Thromboembolism. J. Am. Coll. Surg. 2018, 227, S80–S81. [Google Scholar] [CrossRef]
  91. Udy, A.A.; Varghese, J.M.; Altukroni, M.; Briscoe, S.; McWhinney, B.C.; Ungerer, J.P.; Lipman, J.; Roberts, J.A. Subtherapeutic initial beta-lactam concentrations in select critically ill patients: Association between augmented renal clearance and low trough drug concentrations. Chest 2012, 142, 30–39. [Google Scholar] [CrossRef]
  92. Drust, A.; Luchtmann, M.; Firsching, R.; Troger, U.; Martens-Lobenhoffer, J.; Bode-Boger, S.M. Recurrent seizures in a levetiracetam-treated patient after subarachnoid hemorrhage: A matter of enhanced renal function? Epilepsy Behav. 2012, 23, 394–395. [Google Scholar] [CrossRef]
  93. Grootaert, V.; Willems, L.; Debaveye, Y.; Meyfroidt, G.; Spriet, I. Augmented renal clearance in the critically ill: How to assess kidney function. Ann. Pharmacother. 2012, 46, 952–959. [Google Scholar] [CrossRef]
  94. Morbitzer, K.; Jordan, D.; Sullivan, K.; Durr, E.; Olm-Shipman, C.; Rhoney, D. Enhanced renal clearance and impact on vancomycin trough concentration in patients with hemorrhagic stroke. Pharmacotherapy 2016, 36, e218. [Google Scholar] [CrossRef]
  95. Neves, M.; Baptista, J.P.; Rodrigues, L.; Pinho, J.; Teixeira, L.; Pimentel, J. Correlation between estimated glomerular filtration rate and measured renal creatinine clearance in critically ill patients with normal serum creatinine. Nephrol. Dial. Transplant. 2013, 28, 345. [Google Scholar] [CrossRef] [Green Version]
  96. Baptista, J.P.; Silva, N.; Costa, E.; Fontes, F.; Marques, M.; Ribeiro, G.; Pimentel, J. Identification of the critically ill patient with augmented renal clearance: Make do with what you have! Intensive Care Med. 2014, 40, S110. [Google Scholar] [CrossRef]
  97. Akers, K.S.; Niece, K.L.; Chung, K.K.; Cannon, J.W.; Cota, J.M.; Murray, C.K. Modified Augmented Renal Clearance score predicts rapid piperacillin and tazobactam clearance in critically ill surgery and trauma patients. J. Trauma Acute Care Surg. 2014, 77, S163–S170. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Flow chart of the study search and screening.
Figure 1. Flow chart of the study search and screening.
Pharmaceutics 14 00445 g001
Figure 2. Forest plot of the prevalence of ARC in mixed intensive care unit (ICU) population. Clearance Determination method: m = measured, c = calculated; CI, confidence interval; N, study size.
Figure 2. Forest plot of the prevalence of ARC in mixed intensive care unit (ICU) population. Clearance Determination method: m = measured, c = calculated; CI, confidence interval; N, study size.
Pharmaceutics 14 00445 g002
Figure 3. Forest plot of the prevalence of ARC in neurocritical care (A) and trauma intensive care unit (ICU) population (B). Clearance Determination method: m = measured, c = calculated; CI, confidence interval; N, study size.
Figure 3. Forest plot of the prevalence of ARC in neurocritical care (A) and trauma intensive care unit (ICU) population (B). Clearance Determination method: m = measured, c = calculated; CI, confidence interval; N, study size.
Pharmaceutics 14 00445 g003
Figure 4. Forest plot of the prevalence of ARC in sepsis intensive care unit (ICU) (A) and other population (B). Clearance Determination method: m = measured, c = calculated; CI, confidence interval; N, study size.
Figure 4. Forest plot of the prevalence of ARC in sepsis intensive care unit (ICU) (A) and other population (B). Clearance Determination method: m = measured, c = calculated; CI, confidence interval; N, study size.
Pharmaceutics 14 00445 g004
Figure 5. Funnel plot of studies reporting prevalence.
Figure 5. Funnel plot of studies reporting prevalence.
Pharmaceutics 14 00445 g005
Figure 6. Forest plot of risk factors for augmented renal clearance. (A), age (as continuous variable); (B), male sex; (C), trauma. Clearance Determination method: m = measured, c = calculated; CI, confidence interval; OR, odds ratio; SE, standard error.
Figure 6. Forest plot of risk factors for augmented renal clearance. (A), age (as continuous variable); (B), male sex; (C), trauma. Clearance Determination method: m = measured, c = calculated; CI, confidence interval; OR, odds ratio; SE, standard error.
Pharmaceutics 14 00445 g006
Table 1. Summary characteristics of studies included in ARC systematic review and meta-analysis of prevalence and risk factors.
Table 1. Summary characteristics of studies included in ARC systematic review and meta-analysis of prevalence and risk factors.
AuthorYearPopulationStudy DesignClearance DeterminationARC DefinitionNPrevalence (%)Male n (%)Age *Main DiagnosesIdentifiable Risk FactorsRenal Impairment
Joynt et al. [29]2001Sepsis ICUprospective observationalm24 h Urine1301136.47 (63.6)45 ± 16Sepsisnot reportedExcluded
Fuster-Lluch et al. [30]2008Mixed ICUprospective observationalcNKF1208918.067 (75.3)60.5 (18–86)Severalnot reportedExcluded
Baptista et al. Portugal [31]2011Mixed ICUprospective observationalm24 h Urine13012035.887 (72.5)55.9 ± 21.1Sepsis, Traumanot reportedExcluded
Baptista et al. Australia [31]2011Mixed ICUprospective observationalm8 h Urine1308948.364 (71.9)40 ± 18.9Sepsis, Traumanot reportedExcluded
Minville et al. PolyTrauma [32]2011Trauma ICUretrospective observationalm24 h Urine12014454.9108 (75)42 ± 18Poly trauma ICUAge
Trauma
Excluded
Minville et al. Non-PolyTrauma [32]2011Trauma ICUretrospective observationalm24 h Urine12014019.388 (62.8)58 ± 17Non trauma ICUAge
Trauma
Excluded
Lautrette et al. [17]2012Sepsis ICUretrospective observationalm24 h Urine1403225.015 (46.8)54 ± 16Infectious meningitisnot reportedIncluded
Baptista et al. [33]2012Sepsis ICUprospective observationalm24 h Urine1309339.869 (74.2)58 (34–75)Trauma, Sepsis,
Other.
not reported Excluded
Grootaert et al. [34]2012Mixed ICUretrospective observationalm24 h Urine120131729.6247 (18.8)59 (48–67)Severalnot reportedUnclear
Carlier et al. [35]2013Mixed ICUprospective observationalm24 h Urine1306131.151 (85)56 (48–67)Infectionsnot reportedExcluded
Udy et al. Sepsis [36]2013Sepsis ICUprospective observationalm6 h Urine1304339.522 (51.2)46.3 ± 17.1SepsisAge, Trauma, mod. SOFA Included
Udy et al. Trauma [36]2013Trauma ICUprospective observationalm6 h Urine1302885.723 (82.1)36.4 ± 13.9TraumaAge, Trauma, mod. SOFA Included
Minkute et al. [37]2013Mixed ICUretrospective observationalcC&G1303650.029 (80.5)49.75 (21)Several not reportedExcluded
Udy et al. [38]2013Mixed ICUprospective observationalm8 h Urine12011053.670 (63.6)50.9 ± 16.9Severalnot reported Excluded
Claus et al. [39]2013Mixed ICUprospective observationalm24 h Urine13012851.686 (67.2)59 (49–67.8)SeveralAge, APACHEII, Male sexExcluded
Baptista et al. group 2 [40]2014Sepsis ICUprospective observationalm8 h Urine1302540.017 (68)59.9 ± 17.2Severalnot reported Excluded
Baptista et al. group 1 [40]2014Sepsis ICUretrospective observationalm8 h Urine1307936.752 (66)57.8 ± 15.5Severalnot reportedExcluded
Baptista et al. [41]2014Mixed ICUprospective observationalm8 h Urine1305455.639 (72.2)54.2 ± 16.9Severalnot reportedExcluded
Campassi et al. [42]2014Mixed ICUprospective observationalm24 h Urine12036328.4103 (28.4)56.5 ± 16SeveralAge, DMExcluded
Udy et al. Multicenter [43]2014Mixed ICUprospective observationalm8 h Urine13028165.1178 (63.3)54.4 (52.5–56.4)Severalnot reportedExcluded
Adnan et al. [44]2014Mixed ICUprospective observationalm24 h Urine1304938.837 (75.5)34 (24–47)Trauma, others not reportedExcluded
Ruiz et al. [45]2015Mixed ICUprospective observationalm24 h Urine13036033.3246 (68.3)50 ± 19Polytrauma, Non-polytrauma Age, PolytraumaExcluded
Huttner et al. [46]2015Sepsis ICUprospective observationalcC&G13010064.075 (73.5)46 ± 10.55Severalnot reportedExcluded
Dias et al. [47]2015Neuro ICUretrospective observationalcC&G1301888.916 (89)41 ± 15.6TBI, Polytraumanot reportedIncluded
May et al. [15]2015Neuro ICUprospective observationalm24 h Urine13020100.08 (40)52.14 ± 10.36SAHnot reportedExcluded
De Waele et al. [48]2015Mixed ICUretrospective observationalm24 h Urine130108155.9687 (63.6)62 (20.5)Severalnot reportedExcluded
Steinke et al. [49]2015Surgical ICUretrospective observationalm18 h Urine13010016.061 (61)66 (57–74)Infection, others not reportedIncluded
Chu et al. [50]2016Sepsis ICUretrospective observationalcC&G13014847.397 (65.5)55.3 ± 14.9Infectionnot reported Excluded
Kawano et al. [51]2016Mixed ICUprospective observationalm8 h Urine13011138.762 (55.9)67 (53–770)SeveralAge, DM, Weight, APACHEII, othersExcluded
Saour et al. [52]2016Trauma ICUretrospective observationalcMDRD12077561.3581 (75)37.7 ± 17Severalnot reported Excluded
Abd El Naeem et al. [53]2017Mixed ICUprospective observationalm24 h Urine1305040.032 (64)71 ± 15Sepsis, othersnot reported Excluded
Barletta et al. [54]2016Trauma ICUretrospective observationalm12 h Urine1306569.248 (74)48 ± 18TBI, other traumasnot reportedUnclear
Declercq et al. Trauma Surgery [55]2016Surgical non-ICUprospective observationalm8 h Urine13012934.975 (58)62 (46–75)Trauma surgeryAge, SexExcluded
Declercq et al. Abdominal Surgery [55]2016Surgical non-ICUprospective observationalm8 h Urine13010330.176 (74)63 (51–71)Abdominal surgeryAge Excluded
Hirai et al. [3]2016Mixed ICUretrospective observationalcC&G13029216.4185 (63.4)72 (62.8–82)SeveralAge, Brain injury, othersExcluded
Ehmann et al. [56]2017Mixed ICUprospective observationalcC&G1304810.427 (56.3)55.5 (32–69.9)Sepsis, othersnot reportedIncluded
Burnham et al. [57]2017Sepsis ICUretrospective observationalcMDRD1304945.5260 (52.6)59.9 ± 15.8SepsisAge, sepsis severity, othersIncluded
Carrie et al. RVI [58]2018Trauma ICUretrospective observationalm24 h Urine1303066.727 (90)48 (32–67)Polytrauma, TBInot reportedExcluded
Udy et al. TBI [59]2017Neuro ICUprospective observationalm8 h Urine15011100.09 (81.8)37 (24–49)TBInot reportedIncluded
Barletta et al. ARCTIC [60]2017Trauma ICUprospective observationalm12 h Urine13013366.9101 (76)48 ± 19TBI, fractures, othersAge, SexExcluded
Dhaese et al. [61]2018Surgical ICUprospective observationalm8 h Urine13011031.875 (68.2)60 ± 14.4Severalnot reportedExcluded
Tamatsukuri et al. [62]2018Sepsis ICUprospective observationalm8 h Urine1301735.311 (64.7)60 (19.5)Sepsisnot reported Excluded
Carrie et al. main study [2]2018Sepsis ICUprospective observationalm24 h Urine1507955.762 (78)52 (33–68)Sepsisnot reportedExcluded
Carrie et al. PIP/TAZO [63]2018Sepsis ICUprospective observationalm24 h Urine1305961.047 (80)53 ± 21Polytrauma, non-trauma surgerynot reportedExcluded
Carrie et al. TBI [18]2018Neuro ICUprospective observationalm24 h Urine13022373.1184 (83)36 (23–57)TBI, VAPnot reported Included
Kawano et al. [64]2018Sepsis ICUretrospective observationalcJapanese equation1302806.8145 (51.8)74 (64–83)InfectionAge, Sex, DM, othersExcluded
Tsai et al. [65]2018Mixed ICUprospective observationalm8 h Urine1309732.060 (46)50 ± 18Sepsis, Trauma, others not reportedExcluded
Wong et al. [66]2018Mixed ICUprospective observationalcC&G13033058.2198 (60)53.4 ± 17.7Infectionnot reported Included
Ishii et al. [67]2018Mixed ICU—Non-ICUretrospective observationalcJapanese equation12017726.0109 (62)73 (63–80)Tumors, Brain injurynot reported Excluded
Udy et al. BLINGII [27]2018Sepsis ICUrandomized controlled trialm8 h Urine13025417.7151 (59.4)63 (52–71)Infectionnot reported Included
Ollivier et al. [68]2019Mixed ICUprospective observationalm24 h Urine1502185.717 (81)36 (27–60)Trauma, Surgerynot reportedIncluded
Wu et al. [69]2019Mixed ICUprospective observationalm24 h Urine13010046.066 (66)60 (47–71)SeveralAge, SOFA, Weight, othersExcluded
Aitullina et al. [70]2019Mixed ICUretrospective observationalcnot reported1089716.565 (67)63 (51–73.5)Severalnot reportedIncluded
Weber et al. [71]2019Oncology ICUprospective observationalm24 h Urine1202437.514 (58.3)59 (39.8–63.5)Febrile neutropenianot reported Excluded
Izumisawa et al. Hematomalignancy [72]2019Oncology Non-ICU & ICUretrospective observationalcC&G1202618.4146 (55.9)65.6 ± 13.6Hematologic malignancynot reported Excluded
Izumisawa et al. Non-Malignancy [72]2019Oncology Non-ICU & ICUretrospective observationalcC&G12026111.1175 (67)67.2 ± 16.9Non malignancynot reported Excluded
Chu et al. [73]2019Mixed ICU—Non-ICUretrospective observationalcC&G13031559.0213 (67.6)56.3 (19)Infectionnot reportedExcluded
Villanueva et al. [74]2019Trauma ICUretrospective observationalcC&G1607050.057 (81.4)47.5 (31–61)TBI, Spinal injurynot reported Excluded
Morbitzer et al. aSAH [75]2019Neuro ICUprospective observationalm8 h Urine1305094.016 (32)57.2 ± 10.7SAHnot reported Excluded
Morbitzer et al. ICH [75]2019Neuro ICUprospective observationalm8 h Urine1303050.018 (60)70 ± 13.7ICHnot reported Excluded
Mulder et al. [76]2019Trauma ICUretrospective observationalm24 h Urine13020757.0141 (68)45 ± 20TraumaAge, Sex, othersExcluded
Bricheux et al. [77].2019Hospitalized retrospective observationalcC&G13030026.7203 (68)59 ± 17Abdominal infection, Pneumonianot reportedUnclear
Helset et al. [78]2020Mixed ICUprospective observationalm24 h Urine1308325.361 (73.5)54.5 (38–63)Severalnot reported Unclear
Gijsen et al. [7]2020Mixed ICUretrospective observationalm24 h Urine130426735.22669 (62.5)65 (54–74)Severalnot reportedExcluded
Barrasa et al. [79]2020Mixed ICUprospective observationalm10 h Urine1301723.512 (70.6)61.7Severalnot reported Included
Lannou et al. [80]2020Neuro ICUprospective observationalm24 h Urine1306053.353 (88)48 (32–60)TBI, Multiple traumanot reported Excluded
Aréchiga-Alvarado et al. [81]2020Mixed ICUprospective observationalcC&G1306350.856 (88.9)33.25 (47.5)Infectionnot reportedUnclear
Carrie et al. Amikacin [82]2020Surgical ICUretrospective observationalcC&G1307020.053 (76)65 (51–73)Infectionnot reported Unclear
Saito et al. [83]2020Oncology ICUretrospective observationalcown predictive model13013341.480 (60.2)64 (25–86)Haematologic malignanciesAge, Sex, Scr, othersIncluded
Lannou et al. Editorial Letter [84]2020Neuro ICUretrospective observationalm24 h Urine1553076.7not reported33 (47–57)Brain traumanot reportedIncluded
Cojutti et al. [28]2020Oncology ICUprospective interventionalcMDRD1307536.047 (62.7)58 (51–66)Febrile neutropenianot reportedIncluded
Brown et al. [85]2020Hospitalized retrospective observationalm8 h Urine1308525.943 (50.6)55 (41–70)Severalnot reportedExcluded
Chen et al. [86]2020Neuro ICUretrospective observationalcC&G13010425.071 (68.3)44.5 (18.5)Cerebral tumor, Stroke, TBInot reported Excluded
Baptista et al. [87]2020Mixed ICUretrospective observationalm8 h Urine13045424.9293 (64.5)66 (52–76)SeveralAge, Sex, Trauma, othersIncluded
Nei et al. [88]2020Mixed ICUretrospective observationalcCKD-EPI1303684.1208 (56.5)66.8 (55.7–76.6)TBI, Trauma, Sepsis, othersAge, ICH, SOFA, Trauma, othersIncluded
APACHE II = Acute Physiology and Chronic Health Evaluation; ARC = Augmented Renal Clearance; aSAH = aneurysmal subarachnoid hemorrhage; CG = Cockcroft Gault equation; CKD-EPI = Chronic Kidney Disease Epidemiology; CrCl = creatinine clearance; ICH = intracranial hemorrhage; ICU = intensive care unit; MDRD = modification of diet in renal disease method; SAH = subarachnoid hemorrhage; SAPS II = Simplified Acute Physiology Score; SCr = serum creatinine; SOFA = sequential organ failure assessment score; TBI = traumatic brain injury. * Age reported in median (IQR) or mean ± SD, ARC cut-off reported in mL/min/1.73 m2, Clearance Determination method: m = measured, c = calculated.
Table 2. Summary characteristics of individual studies reporting other risk factors.
Table 2. Summary characteristics of individual studies reporting other risk factors.
AuthorYearPopulationSample SizeClearance DeterminationIdentified Risk Factor (s)Odds Ratio (95% CI)Study Inclusion in Prevalence Meta-Analysis
Hirai et al. [3]2016Mixed Hospital292CalculatedFebrile Neutropenia2.76 (1.11–6.67)
Fluid Infusion ≥ 1500 mL/day2.53 (1.27–5.16)
Traumatic Brain Injury5.11 (1.49–17.57)
Nei et al. [88]2020Mixed ICU368CalculatedCharlson Comorbidity Index0.80 (0.16–1.00)
Intracerebral Hemorrhage2.82 (1–69.1)
Kawano et al. [51]2016Mixed ICU111MeasuredPost-Operative Without Sepsis0.28 (0.07–1.04)
Wu et al. [69]2019Mixed ICU100MeasuredLoop Diuretics0.32 (0.11–0.93)
Age < 504.02 (1.54–10.51)
Udy et al. [36]2013Mixed ICU71MeasuredAge </= 5028.6 (4.4–187.2)
Ramos et al. [89]2017Mixed ICU36Measured24h Sodium Excretion0.99 (0.98–1.00)
Saito et al. [83]2020Oncology Hospital133CalculatedSerum Creatinine0.89 (0.83–0.94)
Leukemia9.4 (2.4–36.8)
Fever2.4 (0.78–7.1)
Burnham et al. [57]2017Sepsis ICU494CalculatedAfrican American Ethnicity3.45 (1.40–8.50)
Sepsis Severity0.54 (0.30–0.97)
Mulder et al. [76]2019Trauma ICU207MeasuredPacked RBC Transfusion0.31 (0.15–0.66)
Eidelson et al. [90]2018Trauma ICU154MeasuredAdmission Hematocrit1.18 (1.04–1.33)
Barletta et al. [60]2017Trauma ICU133MeasuredSerum Creatinine < 0.7 mg/dL12.5 (3–52.6)
Age < 5658.3 (5.2–658.9)
Age 56–7513.5 (1.2–151.7)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hefny, F.; Stuart, A.; Kung, J.Y.; Mahmoud, S.H. Prevalence and Risk Factors of Augmented Renal Clearance: A Systematic Review and Meta-Analysis. Pharmaceutics 2022, 14, 445. https://doi.org/10.3390/pharmaceutics14020445

AMA Style

Hefny F, Stuart A, Kung JY, Mahmoud SH. Prevalence and Risk Factors of Augmented Renal Clearance: A Systematic Review and Meta-Analysis. Pharmaceutics. 2022; 14(2):445. https://doi.org/10.3390/pharmaceutics14020445

Chicago/Turabian Style

Hefny, Fatma, Anna Stuart, Janice Y. Kung, and Sherif Hanafy Mahmoud. 2022. "Prevalence and Risk Factors of Augmented Renal Clearance: A Systematic Review and Meta-Analysis" Pharmaceutics 14, no. 2: 445. https://doi.org/10.3390/pharmaceutics14020445

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