Skip to main content
Journal of Clinical Medicine logoLink to Journal of Clinical Medicine
. 2020 May 10;9(5):1413. doi: 10.3390/jcm9051413

Timing of Initiation of Renal Replacement Therapy in Sepsis-Associated Acute Kidney Injury

José Agapito Fonseca 1,*, Joana Gameiro 1, Filipe Marques 1, José António Lopes 1
PMCID: PMC7290350  PMID: 32397637

Abstract

Sepsis-associated acute kidney injury (SA-AKI) is a major issue in medical, surgical and intensive care settings and is an independent risk factor for increased mortality, as well as hospital length of stay and cost. SA-AKI encompasses a proper pathophysiology where renal and systemic inflammation play an essential role, surpassing the classic concept of acute tubular necrosis. No specific treatment has been defined yet, and renal replacement therapy (RRT) remains the cornerstone supportive therapy for the most severe cases. The timing to start RRT, however, remains controversial, with early and late strategies providing conflicting results. This article provides a comprehensive review on the available evidence on the timing to start RRT in patients with SA-AKI.

Keywords: acute kidney injury, prevention, renal replacement therapy, sepsis, treatment

1. Introduction

Acute kidney injury (AKI) is a clinical syndrome characterized by a rapid decline in kidney function, encompassing numerous etiologies, and different and complex pathophysiological processes [1,2]. AKI is particularly important since it is strongly associated with increased hospital length of stay (LOS), costs, in-hospital mortality, development of chronic kidney disease (CKD) and long-term mortality risk [3,4,5,6]. The global burden of AKI accounts for around 13.3 million cases a year, and in the United States alone hospitalizations for AKI are on the rise [7,8,9].

The definition of AKI has undergone serious modifications over the last twenty years. Since the first consensual definition provided in 2004 by the Risk, Injury, Failure, Loss of kidney function and End-stage renal disease (RIFLE) classification [10], the initial term of acute renal failure (ARF) evolved to AKI as small increases in serum creatinine (SCr) were associated with increased mortality [1,11], widening the spectrum of the definition which now comprises from minor changes in markers of renal function to requirement for renal replacement therapy (RRT), as adopted by the Acute Kidney Injury Network (AKIN), in 2007 [12]. More recently, in 2012, the Kidney Disease: Improving Global Outcomes (KDIGO) combined both RIFLE and AKIN classifications, allowing for a definition that could be applied in clinical medicine, research and for public health purposes (Table 1) [13]. These definitions are not immutable and new definitions, chiefly concerning the duration of AKI, have recently been addressed, as observed by the proposed definitions for transient and persistent acute kidney disease (AKD) [14].

Table 1.

Acute kidney injury according to the Kidney Disease: Improving Global Outcomes (KDIGO) classification. SCr—serum creatinine, UO—urinary output.

Acute Kidney Injury Staging According to Kidney Disease Improving Global Outcomes (KDIGO) Classification
Stage SCr UO
1 ↑ SCr ≥ 26.5 μmol/l (≥ 0.3 mg/dl) or ↑ SCr ≥ 150–200% (1.5–1.9×) <0.5 mL/kg/h (>12 h)
2 ↑ SCr > 200–300% (> 2–2.9×) <0.5 mL/kg/h (>12 h)
3 ↑ SCr > 300% (≥3×) or ↑ SCr to ≥ 353.6 μmol/l (≥4 mg/dl) or initiation of renal replacement therapy <0.3 mL/kg/h (24 h) or anuria (12 h)

Acute complications related to AKI are diverse and depend on the severity of the insult [15]. Prompt recognition and early intervention directed to the cause of AKI and to the related complications, may soften both the duration and severity of this clinical syndrome, improving outcomes [16]. Special attention must be paid to identifying and eliminating potential insults, namely drugs, hypotensive states and exposure to iodinated contrast agents. Adequate volume assessment, as well as acid-based and electrolyte disturbances management must be addressed as well [17,18,19,20]. Nutritional support, surveillance of uremic symptoms and uremic bleeding associated with platelet dysfunction are also of critical importance in the management of AKI and the respective outcomes [21,22]. However, to date, no specific pharmacological therapy has proved to be effective in the prevention or treatment of AKI, partly owing to the various insults and different pathophysiological pathways associated with AKI [23].

In recent years, retrospective and prospective studies, as well as randomized controlled trials (RCTs), have focused on the timing of initiation of renal replacement therapy (RRT), yielding inconsistent results [24,25,26,27]. Most studies are considerably heterogenic concerning the definitions of early and late strategies. The clinical setting of the population is also highly variable between studies [28,29]. Whether an early, compared to a delayed strategy, for RRT initiation can improve outcomes in AKI is still a matter of debate.

Sepsis is the leading cause of AKI in critically ill patients, and sepsis-associated AKI (SA-AKI) is associated with higher severity scores, an increased need for RRT, and increases the risk of death and prolonged LOS compared to nonseptic AKI [30,31]. SA-AKI has a proper pathophysiology that exceeds the previously admitted acute tubular necrosis resulting from kidney ischemic injury. It is now understood that a set of different mechanisms including intra-renal and systemic inflammation, oxidative stress, microvascular and endothelial dysfunction are detrimental in the pathophysiology of SA-AKI [32,33,34].

Data regarding the impact of the timing of RRT initiation in SA-AKI are scarce and somewhat contradictory and inconclusive. In fact, most of the studies on this topic are small, retrospective, single-center and observational, and in studies addressing the impact of the timing of RRT initiation in the general ICU patients, data on SA-AKI patients are almost inexistent.

The aim of this review is to focus on the different studies comparing early versus delayed strategies in initiation of RRT in SA-AKI.

2. Timing of Initiation of RRT

Established and widely accepted indications for starting RRT include refractory fluid overload, severe hyperkalemia and metabolic acidosis refractory to medical therapy, signs of uremia (namely pericarditis and encephalopathy) and intoxications from dialyzable drugs or poisons [35].

There is no established universal definition for early initiation of RRT. Studies reporting different populations allocate to the “early” cohorts of patients with different serum urea levels, according to the RIFLE, AKIN or KDIGO classifications, with different urinary outputs and starting RRT within different times after AKI detection [28,36]. The potential benefits of earlier RRT initiation include premature acid-base and electrolyte equilibrium, avoidance of hypervolemia, which can be deleterious and provokes higher mortality according to different studies, elimination of uremic toxins and reducing both renal and systemic inflammation [37,38]. Retrospective and prospective cohorts, as well as RCTs, have proven the benefits of this approach, translated into earlier recovery of renal function, lesser hospital LOS and improved survival [36,39,40,41].

“Late” or “delayed“ initiation RRT is also loosely defined. Most studies also report more elevated serum urea or creatinine, a higher grade of AKI classified by the RIFLE, AKIN or KDIGO classifications, lower urinary output or even only formal indications for RRT initiation as criteria for the “late” group [28,36]. A delayed start of RRT may allow for patient stabilization, ensuring mainly hemodynamic and ventilatory conditions, reducing potential complications associated with prompt RRT start (catheter misplacement, catheter-related bloodstream infections, bleeding or thrombotic events), and even permitting time to renal recovery averting the need for RRT [42,43,44]. Despite great enthusiasm for an early start of RRT, more recent RCTs and meta-analysis have questioned this approach, reporting no survival advantage [24,25,28,45].

Whether these principles also apply exclusively to SA-AKI is also unknown. Table 2 summarizes the different studies that addressed early vs. late initiation of RRT in SA-AKI cohorts.

Table 2.

Trial summary characteristics reporting data on early vs. late renal replacement therapy initiation.

Study Design N RRT Modality Early RRT Start Late RRT Start Follow-Up Mortality
Early vs. Late RRT Start
AUROC
Baek, 2017 Retrospective, single-center, cohort study 177 CRRT initiation within 24 h of vasopressor treatment
(Tvaso-CRRT less than 24 h)
initiation beyond 24 h of vasopressor treatment
(Tvaso-CRRT over 24 h)
28 days, 90 days 28 days - 33.6% vs. 61.5% (p = 0.001)
adjusted OR 0.449 (95% CI 0.211–0.956), p = 0.038
90 days - 44.0% vs. 75.0% (p < 0.001)
adjusted OR 0.369 (95% CI 0.165–0.825), p = 0.015
Tvaso-CRRT >24 h, AUC, 0.634; 95% CI, 0.559–0.705, p = 0.001;
Barbar, 2018 Multicenter, RCT 488 RRT RRT within 12 h after documentation of failure-stage AKI RRT after 48 h if renal recovery had not occurred 28 days, 90 days, 180 days 28 days - 45% vs. 42% (p = 0.48)
90 days - 58% vs. 54% (p = 0.38)
180 days - 61% vs. 57% (p = 0.37)
-
Carl, 2010 Retrospective, single-center, cohort study 147 RRT BUN < 100 mg/dL + AKIN stage ≥ 2 BUN ≥ 100 mg/dL + AKIN stage ≥ 2 14 days, 28 days, 365 days 14 days - 33% vs. 53.3% (p = 0.01)
adjusted OR 3.6 (95% CI 1.7–7.6), p = 0.001
28 days - 52.3% vs. 68.3% (p < 0.05)
adjusted OR 2.6 (95% CI 1.2–5.7), p = 0.01
365 days - 69.3% vs. 86.7% (p < 0.05)
adjusted OR 3.5 (95% CI 1.2–10), p = 0.02
-
Chon, 2012 Retrospective, single-center, cohort study 55 CRRT ≤24 h (mean time to
RRT = 12.5 h)
RIFLE-I and RIFLE-F
>24 h (mean time to
RRT = 42.2 h)
RIFLE-I and RIFLE-F
28 days, 90 days 28 days - 19.4% vs. 47.4% (p = 0.030)
adjusted HR 3.378 (95% CI 1.174–9.722), p = 0.024
90 days - 38.2% vs. 61.1% (p = 0.115)
-
Chou, 2011 Retrospective, single-center, cohort study 370 RRT sRIFLE-0 or -Risk sRIFLE-Injury or -Failure during ICU stay 70.8% vs. 69.7%, p = 0.98 -
Oh, 2016 Retrospective, single-center cohort study 60 CRRT ≤26.4 h
mean time between EGDT and CRRT initiation 7.9 h (1.0–25.1 h)
>26.4 h
mean time between EGDT and CRRT initiation
61.5 h (32.3–137.6 h)
28 days 30.0% vs. 56.7%, p = 0.037
Late CRRT treatment (vs. early CRRT treatment)
adjusted HR 2.461 (95% CI 1.044–5.800), p = 0.040
-
Payen, 2009 Prospective, randomized, multicenter study 76 CRRT RRT for at least 96 h
within 24 h of randomization,
No RRT
unless metabolic
renal failure and classic
indications for RRT
present
28 days CRRT vs. control (54% vs. 44%; p < 0.49) -
Shum, 2013 Retrospective, single center, cohort study 120 CRRT simplified RIFLE-Risk
(Mean time from
ICU admission to RRT = 20.7 h)
simplified
RIFLE-Injury or Failure
(Mean time from ICU admission
to RRT = 10.8 h)
28 days, 3 months and 6 months 28 days - 48.4% vs. 48.3% (p = 0.994)
3 months - 58.1% vs. 55.1% (p = 0.771)
6 months - 61.3% vs. 56.2% (p = 0.62)
-
Tian, 2014 Retrospective, single center, cohort study 160 CRRT CRRT group control group 28 days AKIN 1 - 21.7% vs. 42.3% (NS)
AKIN 2 - 38.7% vs. 66.7% (p = 0.048)
AKIN 3 - 67.4% vs. 84.6% (NS)
adjusted OR 0.254 (95% CI 0.072–0.897), p = 0.033
Yoon, 2018 Retrospective, single center, cohort study 158 CRRT <16.5 h ≥16.5 h 28 days, 60 days, 90 days 28 days - 40.7% vs. 70.8%
HR 2.118 (95% CI 1.375–3.261), p < 0.001
60 days - HR 2.244 (95% CI 1.497–3.363), p < 0.001
90 days - HR 2.115 (95% CI 1.424–3.141), p < 0.001
Interval time from AKI to CRRT initiation
Adjusted HR 1.016 (95% CI 1.008–1.025; p < 0.001)
interval time from AKI to CRRT initiation for ICU mortality AUC 0.786 (95% CI, 0.716–0.856; p < 0.001)

AKI—acute kidney injury, AKIN—Acute Kidney Injury Network, AUC—area under curve, AUROC—area under receiver operating characteristic, BUN—blood urea nitrogen, CI—confidence interval, CRRT—continuous renal replacement therapy, EGDT—early goal directed therapy, HR—hazard ratio, ICU—intensive care unit, RIFLE—Risk, Injury, Failure, Loss of kidney function and End-stage renal disease, OR—odds ratio, RCT—randomized controlled trial, RRT—renal replacement therapy, Tvaso—time from vasopressor.

A Korean retrospective single-center study with 60 patients assessed whether a shorter interval between the start of early goal-directed therapy (EGDT) and CRRT initiation was an independent predictor of mortality in patients with SA-AKI. Patients were divided into an early (≤26.4 h) CRRT initiation group and a late (>26.4 h) CRRT initiation group. All-cause mortality at 28 days was significantly higher in the late CRRT group (30.0% in the early group vs. 56.7% in the late group), even after adjusting for diabetes mellitus, liver failure, and Acute Physiology and Chronic Health Evaluation II scores, conferring a potential benefit for early initiation of CRRT [46].

Earlier, an observational retrospective single-center study with 55 SA-AKI patients at the injury or failure stages of the RIFLE classification compared outcomes between inception of early (≤24 h) and late (>24 h) continuous renal replacement therapy (CRRT). The primary outcome, 28-day mortality, was lower in the early group (19.4% vs. 47.4%) but no differences were observed between the injury or failure stages [47].

A retrospective analysis of time from AKI onset to CRRT initiation was performed according to ICU mortality in 158 septic shock patients with AKI. Mortality rate was high at ICU discharge (50.6%), and non-survivors initiated CRRT later than survivors with a cut-off time from AKI onset to CRRT initiation for ICU mortality of 16.5 h. The cumulative mortality rate was significantly higher in patients in whom CRRT was initiated beyond 16.5 h after AKI onset than in those in whom CCRT was initiated within 16.5 h, pointing to earlier initiation as a predictor of survival [48].

The definitions of “early” and “late” may also encompass other features than solely time from detection of AKI. A retrospective cohort study investigated 3-time interval parameters on the morbidity and mortality of 177 patients with septic shock–induced acute kidney injury. Time from vasopressor initiation to CRRT initiation < 24 h, but not time from ICU admission to CRRT initiation, nor time from endotracheal intubation to CRRT initiation was associated with survival and acted as an independent factor associated with 28-day and 90-day mortality. Therefore, time from vasopressor initiation to CRRT initiation < 24 h could be considered as a bundle for the definition of early CRRT initiation [49].

A retrospective cohort study of 130 ICU patients with sepsis and acute renal failure requiring RRT assessed whether early implementation of RRT (defined as a BUN < 100 mg/dL) compared to a late RRT initiation (after BUN ≥ 100 mg/dL) had an effect on the 28-day mortality. Survival rates were 67%, 47.7% and 30.7% at 14, 28 and 365 days, respectively, in the early group. Survival rates were 46.7%, 31.7% and 13.3%, respectively, in the late group. After a logistic regression analysis, initiating dialysis with a BUN > 100 mg/dL predicted mortality at 14 days and 365 days, thus early initiation of dialysis was associated with improved survival rates up to one year [50].

A retrospective analysis of 160 critically-ill patients with SA-AKI treated with or without CRRT was performed to investigate which AKI stage might be the optimal timing for starting CRRT. Starting CRRT at AKI stage 2 was associated with reduction in the 28-day mortality, increase in the 28-day survival rate, lesser ICU LOS and a reduction in ventilation time, compared with those in the control group. This hypothesizes that AKI stage 2 may be the ideal time to initiate RRT [51].

The first RCT goes back to 2009, when twelve French ICUs conducted a RCT with 80 patients enrolled within 24 h of developing the first organ failure related to a new septic insult. Patients were assigned to an early intervention group, who received hemofiltration (25 mL/kg/h) for a 96-h period, or to the conventional group, who received standard sepsis management. The primary end point (number, severity, and duration of organ failures during the 14 days) was significantly higher in the early intervention group, suggesting that early application of CRRT may be deleterious in severe sepsis and septic shock [52].

A retrospective single-center cohort study with 120 patients with septic shock and AKI evaluated the impact of early versus late initiation of CRRT on organ dysfunction. Patients were dichotomized into “early” (stage Risk of the RIFLE classification) or “late” (stages Injury or Failure of the RIFLE classification) CRRT initiation. Organ dysfunction at 48 h, evaluated through the SOFA/non-renal SOFA, dialysis requirement and mortality at 28 days, 3 months and 6 months were similar between groups and no clinical benefits of early CRRT initiation were identified [53].

Chou and colleagues evaluated 370 septic patients presenting with AKI requiring RRT in a surgical ICU setting. Patients were divided into early (RIFLE-0 or -Risk) or late (RIFLE-Injury or -Failure) initiation of RRT by RIFLE criteria. Mortality rates in early (70.8%) and late (69.7%) RRT groups were similar and an early strategy was not associated to a decrease in hospital mortality. Therefore, the RIFLE classification was not able to predict the benefits of early RRT initiation in the context of septic AKI [54].

Finally, the Initiation of Dialysis Early versus Late in the Intensive Care Unit (IDEAL-ICU) was a multicenter RCT of 488 patients with septic shock and severe AKI at the failure stage of the RIFLE classification. The early group was defined as RRT start within 12 h of achieving the Failure stage without life-threatening AKI complications, and the delayed group started RRT after a delay of 48 h of achieving the Failure stage, if renal function recovery did not ensue. This trial demonstrated no significant difference in mortality between groups at 90 days [55].

3. Limitations

Several limitations have been pointed to the potential benefit of an early versus late strategy for RRT initiation. Results either showing advantage of an early strategy [27] or benefit from a more conservative approach [24,25,26] must be carefully interpreted since the ICU setting of these cohorts may be predominantly surgical [27,56]. Most studies include different population cohorts with clinical heterogeneities and mixed causes of AKI, in which the extrapolation of the results for SA-AKI is discouraged [24,25,26,27,57]. In addition, previous studies mostly combined early start with more-intensive dialysis and late start with less-intensive dialysis, creating a bias [13].

A recent and comprehensive meta-analysis concerning time of initiation of RRT in AKI described 18 RCTs, of which only three addressed exclusively SA-AKI cohorts and only one of these included more than 100 patients [58]. Study design, including small sample sizes, single-center and retrospective studies, paucity of double-blinded studies, heterogeneity in eligibility criteria, lack of high-quality studies as well as scarcity of RCTs pose major challenges in ascertaining the benefits from early versus delayed strategies [46,47,48,49,51,53].

RRT modalities (intermittent hemodialysis or CRRT) and dialysis dose have shown great disparity between studies, and were mostly left to clinicians’ decision, which creates a bias concerning patients’ outcomes [46,48,49,54].

AKI was also subject of different definitions, and not every study included urinary output as criteria to define AKI, thus leading to exclusion of potential patients with SA-AKI [53,54]. Most studies also report the RIFLE classification for AKI, whose sensitivity for detection AKI is known to be inferior compared to contemporary KDIGO classification [53,55,59,60].

Furthermore, the definitions of “early” and “late” are not standardized and remain to be defined according to clinicians’ criteria. These criteria may include time, hemodynamic parameters, biochemical characteristics, and what one study may define as “early” can be defined as “late” in other studies [28,58,61]. Additionally, criticism in the choice of delaying only 48 h to allow for renal recovery and including these patients in the “late” or “delayed” strategy has been stated. Kidney function may ensue much later than 48 h, allowing for latter renal recovery and avert the need for RRT [55].

Follow-up length is also inconsistent and despite some studies reporting evidence of the 28-day survival benefit for the early RRT group, the effect of this strategy on long-term survival remains unclear [47,49,61]. Moreover, the benefit of RRT might be attributable not only to the early nonspecific removal of inflammatory mediators but also to an early stabilization of the hemodynamic, respiratory, and biological status [46].

4. Future Perspectives

The optimal timing for initiation of RRT is not clarified on the basis of research evaluated to date both in SA-AKI and other etiologies of AKI [28,58,61]. The results of the studies included in our analysis are contradictory. Large, multicenter prospective observational studies are required to make sure the impact of CRRT timing on septic AKI [62].

To our knowledge, no specific RCT is underway to ascertain the optimal time for RRT initiation in SA-AKI. The Standard Versus Accelerated Initiation of Dialysis in Acute Kidney Injury (STARRT-AKI) is an ongoing multicenter trial that plans to enroll at least 2866 critically ill patients in order to detect a 6% difference in mortality in favor of an early RRT start strategy. A specific subgroup analysis for patients with sepsis and septic shock, as defined by the Sepsis-3 criteria based on the rationale that earlier RRT, due to more aggressive removal of inflammatory mediators, might have a more prominent effect among patients with SA-AKI, is awaited in order to define whether an early strategy could be beneficial for this specific population [63].

All in all, timing to start RRT should be individualized to patients’ clinical status and laboratory progression. Nonetheless, an early RRT start in septic patients could potentially improve outcomes by limiting systemic inflammation, fluid overload and organ injury, though consistent evidence is still lacking. Moreover, SA-AKI is associated with lower SCr due to reduced production of creatinine and hemodilution associated with a considerably positive fluid balance, and a more pronounced oliguria, thus a less severe KDIGO stage defined these criteria might underestimate the severity of AKI and create a bias in defining RRT timing. Novel biomarkers, such as NGAL and TIMP-2 x IGFBP-7, have been reported as potential predictors of AKI severity after admission, which might prove useful in deciding the timing to start RRT in this setting, though further studies are still required to validate the routine use of these biomarkers in clinical practice.

5. Conclusions

In conclusion, the available evidence demonstrates contradictory results concerning the benefits of early RRT initiation in SA-AKI. Moreover, postponing RRT initiation may allow for renal recovery from AKI, averting the need for RRT. Evidence results essentially from small, retrospective, non-double blinded studies and although evidence in mortality improvement is lacking, especially with RCTs, other outcomes such as ICU and hospital LOS, development of CKD and late mortality were rarely assessed. Definitions for “early” and “late” start need to be standardized and the optimal time to initiate RRT remains undefined. Large multicenter randomized trials with better design are needed to answer these questions.

Author Contributions

J.A.F. and J.G. made substantial contributions to the study concept and design, analysis and interpretation of data. J.A.F., J.G. and F.M. were involved in drafting the manuscript and revising it critically for important intellectual content. J.A.F. and F.M. participated in the acquisition of data. J.G. and J.A.L. were involved in the critical revision of the manuscript and approval of the final version to be submitted. All authors have read and agreed to the published version of the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  • 1.Chertow G., Burdick E., Honour M., Bonventre J., Bates D. Acute kidney injury, mortality, length of stay, and costs in hospitalized patients. J. Am. Soc. Nephrol. 2005;16:3365–3370. doi: 10.1681/ASN.2004090740. [DOI] [PubMed] [Google Scholar]
  • 2.Kellum J.A., Prowle J.R. Paradigms of acute kidney injury in the intensive care setting. Nat. Rev. Nephrol. 2018;14:217–230. doi: 10.1038/nrneph.2017.184. [DOI] [PubMed] [Google Scholar]
  • 3.Uchino S., Kellum J.A., Bellomo R., Doig G.S., Morimatsu H., Morgera S. Acute renal failure in critically ill patients: A multinational, multicenter study. JAMA. 2005;294:813–818. doi: 10.1001/jama.294.7.813. [DOI] [PubMed] [Google Scholar]
  • 4.Hoste E.A., Bagshaw S.M., Bellomo R., Cely C.M., Colman R., Cruz D.N., Honoré P. Epidemiology of acute kidney injury in critically ill patients: The multinational AKI-EPI study. Intensive Care Med. 2015;41:1411–1423. doi: 10.1007/s00134-015-3934-7. [DOI] [PubMed] [Google Scholar]
  • 5.Bellomo R. The epidemiology of acute renal failure: 1975 versus 2005. Curr. Opin. Crit. Care. 2006;12:557–560. doi: 10.1097/01.ccx.0000247443.86628.68. [DOI] [PubMed] [Google Scholar]
  • 6.Lafrance J.P., Miller D.R. Acute kidney injury associates with increased long-term mortality. J. Am. Soc. Nephrol. 2010;21:345–352. doi: 10.1681/ASN.2009060636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Thongprayoon C., Kaewput W., Thamcharoen N., Bathini T., Watthanasuntorn K., Lertjitbanjong P., Kröner P.T. Incidence and Impact of Acute Kidney Injury after Liver Transplantation: A Meta-Analysis. J. Clin. Med. 2019;8:372. doi: 10.3390/jcm8030372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lertjitbanjong P., Thongprayoon C., Cheungpasitporn W., O’Corragain O.A., Srivali N., Bathini T., Watthanasuntorn K., Aeddula N.R., Salim S.A., Ungprasert P., et al. Acute Kidney Injury after Lung Transplantation: A Systematic Review and Meta-Analysis. J. Clin. Med. 2019;8:1713. doi: 10.3390/jcm8101713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Thongprayoon C., Kaewput W., Thamcharoen N., Bathini T., Watthanasuntorn K., Salim S.A., Ungprasert P., Lertjitbanjong P., Aeddula N.R., Torres-Ortiz A., et al. Acute Kidney Injury in Patients Undergoing Total Hip Arthroplasty: A Systematic Review and Meta-Analysis. J. Clin. Med. 2019;8:66. doi: 10.3390/jcm8010066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bellomo R., Ronco C., Kellum J.A., Mehta R.L., Palevsky P. 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. doi: 10.1186/cc2872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lassnigg A., Schmidlin D., Mouhieddine M., Bachmann L.M., Druml W., Bauer P., Hiesmayr M. Minimal changes of serum creatinine predict prognosis in patients after cardiothoracic surgery: A prospective cohort study. J. Am. Soc. Nephrol. 2004;15:1597–1605. doi: 10.1097/01.ASN.0000130340.93930.DD. [DOI] [PubMed] [Google Scholar]
  • 12.Mehta R.L., Kellum J.A., Shah S.V., Molitoris B.A., Ronco C., Warnock D.G., Levin A. Acute Kidney Injury Network: Report of an initiative to improve outcomes in acute kidney injury. Crit. Care. 2007;11:R31. doi: 10.1186/cc5713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kidney Disease: Improving Global Outcomes (KDIGO) Acute Kidney Injury Work Group KDIGO clinical practice guideline for acute kidney injury. Kidney Int. Suppl. 2012;2:S1–S138. [Google Scholar]
  • 14.Chawla L.S., Bellomo R., Bihorac A., Goldstein S.L., Siew E.D., Bagshaw S.M., Bittleman D., Cruz D., Endre Z., Fitzgerald S.L., et al. Acute kidney disease and renal recovery: Consensus report of the Acute Disease Quality Initiative (ADQI) 16 Workgroup. Nat. Rev. Nephrol. Nat. Res. 2017;13:241–257. doi: 10.1038/nrneph.2017.2. [DOI] [PubMed] [Google Scholar]
  • 15.Murugan R., Kellum J.A. Acute kidney injury: What’s the prognosis? Nat. Rev. Nephrol. 2011;7:209–217. doi: 10.1038/nrneph.2011.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Doyle J.F., Forni L.G. Acute kidney injury: Short-term and long-term effects. Crit. Care. 2016;20:188. doi: 10.1186/s13054-016-1353-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bellomo R., Cass A., Cole L., Finfer S., Gallagher M., Lee J., Lo S., McArthur C., McGuiness S., Norton R., et al. An observational study fluid balance and patient outcomes in the Randomized Evaluation of Normal vs. Augmented Level of Replacement Therapy trial. Crit. Care Med. 2012;40:1753–1760. doi: 10.1097/CCM.0b013e318246b9c6. [DOI] [PubMed] [Google Scholar]
  • 18.Payen D., de Pont A.C., Sakr Y., Spies C., Reinhart K., Vincent J.L. A positive fluid balance is associated with a worse outcome in patients with acute renal failure. Crit. Care. 2008;12:R74. doi: 10.1186/cc6916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bouchard J., Soroko S.B., Chertow G.M., Himmelfarb J., Ikizler T.A., Paganini E.P., Mehta R.L. Fluid accumulation, survival and recovery of kidney function in critically ill patients with AKI. Kidney Int. 2009;76:422–427. doi: 10.1038/ki.2009.159. [DOI] [PubMed] [Google Scholar]
  • 20.Davison D., Junker C. Advances in critical care for the nephrologist: Hemodynamic monitoring and volume management. Clin. J. Am. Soc. Nephrol. 2008;3:554–561. doi: 10.2215/CJN.01440307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Finfer S., Chittock D.R., Su S.Y., Blair D., Foster D., Dhingra V., Bellomo R., Cook D., Dodek P., Henderson W.R., et al. Intensive versus conventional glucose control in critically ill patients. N. Engl. J. Med. 2009;360:1283–1297. doi: 10.1056/NEJMoa0810625. [DOI] [PubMed] [Google Scholar]
  • 22.Boccardo P., Remuzzi G., Galbusera M. Platelet dysfunction in renal failure. Semin. Thromb. Hemost. 2004;30:579–589. doi: 10.1055/s-2004-835678. [DOI] [PubMed] [Google Scholar]
  • 23.Vanmassenhove J., Kielstein J., Joerres A., Biesen W.V. Management of patients at risk of AKI. Lancet. 2017;389:2139–2151. doi: 10.1016/S0140-6736(17)31329-6. [DOI] [PubMed] [Google Scholar]
  • 24.Wald R., Adhikari N.K., Smith O.M., Weir M.A., Pope K., Cohen A., Thorpe K., McIntyre L., Lamontagne F., Soth M., et al. Comparison of standard and accelerated initiation of renal replacement therapy in acute kidney injury. Kidney Int. 2015;88:897–904. doi: 10.1038/ki.2015.184. [DOI] [PubMed] [Google Scholar]
  • 25.Jamale T.E., Hase N.K., Kulkarni M., Pradeep K.J., Keskar V., Jawale S., Mahajan D. Earlier-start versus usual-start dialysis in patients with community-acquired acute kidney injury: A randomized controlled trial. Am. J. Kidney Dis. 2013;62:1116–1121. doi: 10.1053/j.ajkd.2013.06.012. [DOI] [PubMed] [Google Scholar]
  • 26.Gaudry S., Hajage D., Schortgen F., Martin-Lefevre L., Pons B., Boulet E., Boyer A., Chevrel G., Lerolle N., Carpentier D., et al. Initiation Strategies for Renal-Replacement Therapy in the Intensive Care Unit. N. Engl. J. Med. 2016;375:122. doi: 10.1056/NEJMoa1603017. [DOI] [PubMed] [Google Scholar]
  • 27.Zarbock A., Kellum J.A., Schmidt C., Van Aken H., Wempe C., Pavenstädt H., Boanta A., Gerß J., Meersch M. Effect of Early vs. Delayed Initiation of Renal Replacement Therapy on Mortality in Critically Ill Patients With Acute Kidney Injury: The ELAIN Randomized Clinical Trial. JAMA. 2016;315:2190. doi: 10.1001/jama.2016.5828. [DOI] [PubMed] [Google Scholar]
  • 28.Wierstra B.T., Kadri S., Alomar S., Burbano X., Barrisford G.W., Kao R.L.C. The impact of “early” versus “late” initiation of renal replacement therapy in critical care patients with acute kidney injury: A systematic review and evidence synthesis. Crit. Care. 2016;20:122. doi: 10.1186/s13054-016-1291-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Besen B.A.M.P., Romano T.G., Mendes P.V., Gallo C.A., Zampieri F.G., Nassar A.P., Jr., Park M. Early Versus Late Initiation of Renal Replacement Therapy in Critically Ill Patients: Systematic Review and Meta-Analysis. J. Intensive Care Med. 2019;34:714. doi: 10.1177/0885066617710914. [DOI] [PubMed] [Google Scholar]
  • 30.Bagshaw S.M., Uchino S., Bellomo R., Morimatsu H., Morgera S., Schetz M., Tan I., Bouman C., Macedo E., Gibney M., et al. Septic acute kidney injury in critically ill patients: Clinical characteristics and outcomes. Clin. J. Am. Soc. Nephrol. 2007;2:431–439. doi: 10.2215/CJN.03681106. [DOI] [PubMed] [Google Scholar]
  • 31.Cruz M.G., Dantas J.G., Levi T.M., Rocha M., de Souza S.P., Boa-Sorte N., de Moura C.G., Cruz C.M. Septic versus non-septic acute kidney injury in critically ill patients: Characteristics and clinical outcomes. Rev. Bras Ter. Intensiva. 2014;26:384–391. doi: 10.5935/0103-507X.20140059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Prowle J.R., Bellomo R. Sepsis-associated acute kidney injury: Macrohemodynamic and microhemodynamic alterations in the renal circulation. Semin. Nephrol. 2015;35:64–74. doi: 10.1016/j.semnephrol.2015.01.007. [DOI] [PubMed] [Google Scholar]
  • 33.Poston J.T., Koyner J.L. Sepsis associated acute kidney injury. BMJ. 2019;364:k4891. doi: 10.1136/bmj.k4891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Gomez H., Ince C., De Backer D., Pickkers P., Payen D., Hotchkiss J., Kellum J.A. A unified theory of sepsis- induced acute kidney injury: Inflammation, microcirculatory dysfunction, bioenergetics, and the tubular cell adaptation to injury. Shock. 2014;41:3–11. doi: 10.1097/SHK.0000000000000052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Pannu N., Gibney R.N. Renal replacement therapy in the intensive care unit. Ther. Clin. Risk Manag. 2005;1:141–150. doi: 10.2147/tcrm.1.2.141.62908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Karvellas C.J., Farhat M.R., Sajjad I., Mogensen S.S., Leung A.A., Wald R., Bagshaw S.M. A comparison of early versus late initiation of renal replacement therapy in critically ill patients with acute kidney injury: A systematic review and meta-analysis. Crit. Care. 2011;15:R72. doi: 10.1186/cc10061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Matson J., Zydney A., Honore P.M. Blood filtration: New opportunities and the implications of systems biology. Crit. Care Resusc. 2004;6:209–217. [PubMed] [Google Scholar]
  • 38.Sugahara S., Suzuki H. Early start on continuous hemodialysis therapy improves survival rate in patients with acute renal failure following coronary bypass surgery. Hemodial Int. 2004;8:320–325. doi: 10.1111/j.1492-7535.2004.80404.x. [DOI] [PubMed] [Google Scholar]
  • 39.Bagshaw S.M., Uchino S., Bellomo R., Morimatsu H., Morgera S., Schetz M., Tan I., Bouman C., Macedo E., Gibney M., et al. Timing of renal replacement therapy and clinical outcomes in critically ill patients with severe acute kidney injury. J. Crit. Care. 2009;24:129–140. doi: 10.1016/j.jcrc.2007.12.017. [DOI] [PubMed] [Google Scholar]
  • 40.Liu K.D., Himmelfarb J., Paganini E., Ikizler T.A., Soroko S.H., Mehta R.L., Chertow G.M. Timing of initiation of dialysis in critically ill patients with acute kidney injury. Clin. J. Am. Soc. Nephrol. 2006;1:915–919. doi: 10.2215/CJN.01430406. [DOI] [PubMed] [Google Scholar]
  • 41.Leite T.T., Macedo E., Pereira S.M., Bandeira S.R., Pontes P.H., Garcia A.S., Militão F.R., Sobrinho I.M., Assunção L.M., Libório A.B. Timing of renal replacement therapy initiation by AKIN classification system. Crit. Care. 2013;17:R62. doi: 10.1186/cc12593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Shingarev R., Wille K., Tolwani A. Management of complications in renal replacement therapy. Semin. Dial. 2011;24:164–168. doi: 10.1111/j.1525-139X.2011.00828.x. [DOI] [PubMed] [Google Scholar]
  • 43.Moreira F.T., Palomba H., Chaves R.C.F., Bouman C., Schultz M.J., Serpa Neto A. Early versus delayed initiation of renal replacement therapy for acute kidney injury: An updated systematic review, meta-analysis, meta-regression and trial sequential analysis of randomized controlled trials. Rev. Bras Ter. Intensiva. 2018;30:376–384. doi: 10.5935/0103-507X.20180054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Elseviers M.M., Lins R.L., Van der Niepen P., Hoste E., Malbrain M.L., Damas P., Devriendt J. Renal replacement therapy is an independent risk factor for mortality in critically ill patients with acute kidney injury. Crit. Care. 2010;14:R221. doi: 10.1186/cc9355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Combes A., Brechot N., Amour J., Cozic N., Lebreton G., Guidon C., Zogheib E., Thiranos J.C., Rigal J.C., Bastien O., et al. Early high-volume hemofiltration versus standard care for post-cardiac surgery shock. The HEROICS study. Am. J. Respir. Crit. Care Med. 2015;192:1179–1190. doi: 10.1164/rccm.201503-0516OC. [DOI] [PubMed] [Google Scholar]
  • 46.Oh H.J., Kim M.H., Ahn J.Y., Ku N.S., Park J.T., Han S.H., Choi J.Y., Han S.H., Yoo T.H., Song Y.G., et al. Can early initiation of continuous renal replacement therapy improve patient survival with septic acute kidney injury when enrolled in early goal-directed therapy? J. Crit. Care. 2016;35:51–56. doi: 10.1016/j.jcrc.2016.04.032. [DOI] [PubMed] [Google Scholar]
  • 47.Chon G.R., Chang J.W., Huh J.W., Lim C.M., Koh Y., Park S.K., Park J.S., Hong S.B. A comparison of the time from sepsis to inception of continuous renal replacement therapy versus RIFLE criteria in patients with septic acute kidney injury. Shock. 2012;38:30–36. doi: 10.1097/SHK.0b013e31825adcda. [DOI] [PubMed] [Google Scholar]
  • 48.Yoon B.R., Leem A.Y., Park M.S., Kim Y.S., Chung K.S. Optimal timing of initiating continuous renal replacement therapy in septic shock patients with acute kidney injury. Sci. Rep. 2019;9:11981. doi: 10.1038/s41598-019-48418-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Baek S.D., Yu H., Shin S., Park H.S., Kim M.S., Kim S.M., Lee E.K., Chang J.W. Early continuous renal replacement therapy in septic acute kidney injury could be defined by its initiation within 24 h of vasopressor infusion. J. Crit. Care. 2017;39:108–114. doi: 10.1016/j.jcrc.2016.12.014. [DOI] [PubMed] [Google Scholar]
  • 50.Carl D.E., Grossman C., Behnke M., Sessler C.N., Gehr T.W. Effect of timing of dialysis on mortality in critically ill, septic patients with acute renal failure. Hemodial Int. 2010;14:11–17. doi: 10.1111/j.1542-4758.2009.00407.x. [DOI] [PubMed] [Google Scholar]
  • 51.Tian H., Sun T., Hao D., Wang T., Li Z., Han S., Qi Z., Dong Z., Lv C., Wang X. The optimal timing of continuous renal replacement therapy for patients with sepsis-induced acute kidney injury. Int. Urol. Nephrol. 2014;46:2009–2014. doi: 10.1007/s11255-014-0747-5. [DOI] [PubMed] [Google Scholar]
  • 52.Payen D., Mateo J., Cavaillon J.M., Fraisse F., Floriot C., Vicaut E. Impact of continuous venovenous hemofiltration on organ failure during the early phase of severe sepsis: A randomized controlled trial. Crit. Care Med. 2009;37:803–810. doi: 10.1097/CCM.0b013e3181962316. [DOI] [PubMed] [Google Scholar]
  • 53.Shum H.P., Chan K.C., Kwan M.C., Yeung AW T., Cheung EW S., Yan W.W. Timing for initiation of continuous renal replacement therapy in patients with septic shock and acute kidney injury. Ther. Apher. Dial. 2013;17:305–310. doi: 10.1111/j.1744-9987.2012.01147.x. [DOI] [PubMed] [Google Scholar]
  • 54.Chou Y.H., Huang T.M., Wu V.C., Wang C.Y., Shiao C.C., Lai C.F., Tsai H.B., Chao C.T., Young G.H., Wang W.J., et al. Impact of timing of renal replacement therapy initiation on outcome of septic acute kidney injury. Crit. Care. 2011;15:R134. doi: 10.1186/cc10252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Barbar S.D., Clere-Jehl R., Bourredjem A., Hernu R., Montini F., Bruyère R., Lebert C., Bohé J., Badie J., Eraldi J.P., et al. Investigators I-IT, the CTN. Timing of renal-replacement therapy in patients with acute kidney injury and sepsis. N. Engl. J. Med. 2018;379:1431–1442. doi: 10.1056/NEJMoa1803213. [DOI] [PubMed] [Google Scholar]
  • 56.Bouman C.S., Oudemans-Van Straaten H.M., Tijssen J.G., Zandstra D.F., Kesecioglu J. Effects of early high-volume continuous venovenous hemofiltration on survival and recovery of renal function in intensive care patients with acute renal failure: A prospective, randomized trial. Crit. Care Med. 2002;30:2205–2211. doi: 10.1097/00003246-200210000-00005. [DOI] [PubMed] [Google Scholar]
  • 57.Lim C.C., Tan C.S., Kaushik M., Tan H.K. Initiating acute dialysis at earlier Acute Kidney Injury Network stage in critically ill patients without traditional indications does not improve outcome: A prospective cohort study. Nephrology (Carlton) 2015;20:148–154. doi: 10.1111/nep.12364. [DOI] [PubMed] [Google Scholar]
  • 58.Zhang L., Chen D., Tang X., Li P., Zhang Y., Tao Y. Timing of initiation of renal replacement therapy in acute kidney injury: An updated meta-analysis of randomized controlled trials. Ren Fail. 2020;42:77–88. doi: 10.1080/0886022X.2019.1705337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Lopes J.A., Jorge S. The RIFLE and AKIN classifications for acute kidney injury: A critical and comprehensive review. Clin. Kidney J. 2013;6:8–14. doi: 10.1093/ckj/sfs160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Luo X., Jiang L., Du B., Wen Y., Wang M., Xi X. A comparison of different diagnostic criteria of acute kidney injury in critically ill patients. Crit. Care. 2014;18:R144. doi: 10.1186/cc13977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Xiao L., Jia L., Li R., Zhang Y., Ji H., Faramand A. Early versus late initiation of renal replacement therapy for acute kidney injury in critically ill patients: A systematic review and meta-analysis. PLoS ONE. 2019;14:e0223493. doi: 10.1371/journal.pone.0223493. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Li Y., Li H., Zhang D. Timing of continuous renal replacement therapy in patients with septic AKI: A systematic review and meta-analysis. Medicine (Baltimore) 2019;98:e16800. doi: 10.1097/MD.0000000000016800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.STARRT-AKI Investigators STandard versus Accelerated initiation of Renal Replacement Therapy in Acute Kidney Injury: Study Protocol for a Multi-National, Multi-Center, Randomized Controlled Trial. Can. J. Kidney Health Dis. 2019;6:2054358119852937. doi: 10.1177/2054358119852937. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Clinical Medicine are provided here courtesy of Multidisciplinary Digital Publishing Institute (MDPI)

RESOURCES