Next Article in Journal
Evaluation of the Photoactivation Effect of 3% Hydrogen Peroxide in the Disinfection of Dental Implants: In Vitro Study
Previous Article in Journal
Epigallocatechin-3-Gallate Prevents the Acquisition of a Cancer Stem Cell Phenotype in Ovarian Cancer Tumorspheres through the Inhibition of Src/JAK/STAT3 Signaling
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Epidemiology and Outcome of Early-Onset Acute Kidney Injury and Recovery in Critically Ill COVID-19 Patients: A Retrospective Analysis

1
CHU Clermont-Ferrand, Service de Réanimation Médicale, F-63000 Clermont-Ferrand, France
2
CHU Clermont-Ferrand, Service de Néphrologie, F-63000 Clermont-Ferrand, France
3
CHU Clermont-Ferrand, Laboratoire de Biochimie, F-63000 Clermont-Ferrand, France
4
CHU Clermont-Ferrand, Laboratoire d’Immunologie, F-63000 Clermont-Ferrand, France
5
ECREIN, UMR1019 UNH, UFR Médecine de Clermont-Ferrand, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
6
Laboratoire Microorganismes: Génome et Environnement, UMR CNRS 6023, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
7
Unité de Nutrition Humaine, INRAe, CRNH Auvergne, Université Clermont Auvergne, F-63000 Clermont-Ferrand, France
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(4), 1001; https://doi.org/10.3390/biomedicines11041001
Submission received: 9 February 2023 / Revised: 28 February 2023 / Accepted: 16 March 2023 / Published: 23 March 2023
(This article belongs to the Section Immunology and Immunotherapy)

Abstract

:
Background: The clinical significance of early-onset acute kidney injury (EO-AKI) and recovery in severe COVID-19 intensive care unit (ICU) patients is poorly documented. Objective: The aim of the study was to assess the epidemiology and outcome of EO-AKI and recovery in ICU patients admitted for SARS-CoV-2 pneumonia. Design: This was a retrospective single-centre study. Setting: The study was carried out at the medical ICU of the university hospital of Clermont-Ferrand, France. Patients: All consecutive adult patients aged ≥18 years admitted between 20 March 2020 and 31 August 2021 for SARS-CoV-2 pneumonia were enrolled. Patients with chronic kidney disease, referred from another ICU, and with an ICU length of stay (LOS) ≤72 h were excluded. Interventions: EO-AKI was defined on the basis of serum creatinine levels according to the Kidney Disease Improving Global Outcomes criteria, developing ≤7 days. Depending on renal recovery, defined by the normalization of serum creatinine levels, EO-AKI was transient (recovery within 48 h), persistent (recovery between 3 and 7 days) or AKD (no recovery within 7 days after EO-AKI onset). Measurements: Uni- and multivariate analyses were performed to determine factors associated with EO-AKI and EO-AKI recovery. Main Results: EO-AKI occurred in 84/266 (31.5%) study patients, of whom 42 (50%), 17 (20.2%) and 25 (29.7%) had EO-AKI stages 1, 2 and 3, respectively. EO-AKI was classified as transient, persistent and AKD in 40 (47.6%), 15 (17.8%) and 29 (34.6%) patients, respectively. The 90-day mortality was 87/244 (35.6%) and increased with EO-AKI occurrence and severity: no EO-AKI, 38/168 (22.6%); EO-AKI stage 1, 22/39 (56.4%); stage 2, 9/15 (60%); and stage 3, 18/22 (81.8%) (p < 0.01). The 90-day mortality in patients with transient or persistent AKI and AKD was 20/36 (55.6%), 8/14 (57.1%) and 21/26 (80.8%), respectively (p < 0.01). MAKE-90 occurred in 42.6% of all patients. Conclusions: In ICU patients admitted for SARS-CoV-2 pneumonia, the development of EO-AKI and time to recovery beyond day 7 of onset were associated with poor outcome.

1. Introduction

Since the beginning of the outbreak of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), many studies have been published to better understand and characterize the global pandemic and its impact on the healthcare system especially in intensive care units (ICUs).
We now know that the virus does not only affect the respiratory system but also other organ systems, with acute kidney injury (AKI) probably being the second most frequent organ failure [1,2,3].
Although early reports from China showed a low AKI rate amongst patients [4,5,6] with SARS-CoV-2 pneumonia, recent studies suggest a higher average incidence, between 44% and 85% in critically ill patients, and a high mortality rate rising up to 51% [7,8,9,10,11,12,13,14,15] in cases of AKI (Table S1).
Several risk factors of AKI in Coronavirus Disease-19 (COVID-19) have been reported such as male gender, ethnicity, older age and pre-existing long-term conditions including diabetes, obesity and chronic kidney disease [1,7,16,17,18]. Several mechanisms could explain the occurrence of AKI in COVID-19 including acute systemic inflammation, coagulopathy, direct viral toxicity, renin angiotensin system activation or unspecific kidney injury factors, such as lung/heart dysfunction, hypovolemia, a high level of positive end-expiratory pressure (PEEP), hemodynamic instability or nephrotoxic drugs [19,20,21,22].
Studying renal recovery to characterize AKI has been suggested in the last few years [23,24,25] as a means to predict patient outcomes and thereby improve treatment and prognosis. For instance, Abdel-Nabey et al. [26] underlined the importance of assessing AKI and renal recovery in order to target a specific population at a higher risk of residual kidney dysfunction.
Until recently, very few studies focused on renal recovery including consequences and risk factors in patients with SARS-CoV-2 pneumonia [16,27].
Thus, the purpose of the study, called SarSCoV-AKI, was to assess the prevalence, risk factors and outcome of early-onset AKI (EO-AKI) and recovery in critically ill patients admitted for severe COVID-19.

2. Materials and Methods

2.1. Study Design

The SarSCoV-AKI study was a retrospective analysis of prospectively collected data. It was a single-centre observational study performed at the medical ICU of the university hospital of Clermont-Ferrand, France. All consecutive adult patients aged ≥18 years admitted between 20 March 2020 and 31 August 2021 with a positive SARS-CoV-2 polymerase chain reaction test result, pneumonia confirmed by computed tomography and hypoxemia requiring >6 L/min supplemental oxygen were enrolled. Exclusion criteria were patients referred from another ICU, patients with chronic kidney disease stages 4 and 5, kidney transplant recipients, patients who refused to participate and patients with a hospital stay shorter than 72 h since they could not be classified according to our definitions. If there were multiple admissions, only the first admission to the ICU was included.

2.2. Ethics

The SarSCoV-AKI study received approval from the ethics committee of the French Intensive Care Society in 2020 (CE-SRLF20-21—French acronym for Comité éthique de la Société de réanimation de langue française) in accordance with our local regulations. All patients or close relatives were informed that their data would be included in the SarSCoV-AKI cohort study and gave written informed consent for the storage and research use of residual blood from samples collected as part of routine care. All the procedures were followed in accordance with the ethical standards of the responsible committee on human experimentation (institutional or regional) and with the Helsinki Declaration of 1975.

2.3. Data Collection

All data were obtained from medical records and electronic patient charts. Baseline patient characteristics were collected, including demographics and comorbidities, before ICU admission. The variables recorded regarding ICU admission and treatments were relative to clinical presentation, reason for ICU admission, diagnosis, therapies implemented and outcomes. Blood sampling and routine biological testing were performed on the day of admission and daily according to standard laboratory protocols.

2.4. Definitions

AKI was defined according to the classification of Kidney Disease Improving Global Outcomes (KDIGO) [28] (Annex 1), using only the serum creatinine (SCr) component. “Baseline SCr” was the best outpatient SCr value between 7 and 365 days before ICU admission or, if unavailable, was estimated using the Modification of Diet in Renal Disease (MDRD) equation [28]. Early-onset AKI was defined as AKI occurring within 7 days after ICU admission: only the first AKI episode was taken into account in the study. Patients were stratified according to the highest AKI stage attained during this first episode.
EO-AKI recovery was based on a sustained (≥48 h) and complete reversal of AKI by KDIGO criteria, and therefore a minimum of 48 h of renal recovery was necessary to separate two distinct AKI episodes. EO-AKI was classified as transient, persistent and AKD according to the Acute Dialysis Quality Initiative (ADQI) [25]. “Transient” AKI was defined as renal recovery within 48 h of AKI onset, and “persistent” AKI as renal recovery occurring ≥3 days and <7 days of EO-AKI onset. Acute kidney disease (AKD) was characterized when AKI stage 1 or greater persisted ≥7 days after EO-AKI onset [25] (Annex 2). Chronic kidney disease (CKD) staging was defined according to the KDIGO definition [28].
The outcome of major adverse kidney event at 90 days (MAKE-90) was the composite outcome of death, dialysis dependence or a glomerular filtration rare (eGFR) <60 mL/min 90 days after ICU admission [25,29,30]. GFR was estimated using Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation (31). SCr at 90 days was retrieved from the medical health records and if unavailable, the patient’s general practitioner was contacted.
Pulmonary bacterial co-infection was defined by the presence of a community-acquired or hospital-acquired bacterial pneumonia associated with SARS-CoV2 pneumonia on ICU admission. The presence of pulmonary bacterial co-infection was defined by the presence of radiological and/or scanographic condensation, bacteriological documentation (a positive quantitative culture of lower respiratory tract samples collected as recommended: bronchoalveolar lavage, >104 CFU/mL, plugged telescoping catheter, >103 CFU/mL, endotracheal aspirate, >106 CFU/mL) and/or presence of positive antigenuria, as defined by the European Centre for Disease Control and Prevention [31]. If bacterial pneumonia occurred at least 2 days after intubation, it was classified as ventilator associated pneumoniae (VAP) [32]. The period at risk for VAP begins from 48 h after intubation, until removal of the tracheal tube and weaning from invasive ventilation, and thus ends with extubation.

2.5. Endpoint

The primary outcome was the incidence of AKI within 7 days after ICU admission, its staging, and the proportions of transient AKI, persistent AKI and AKD.
The secondary endpoints were the determination of the risk factors of transient and persistent AKI, and AKD, and the renal prognostic evaluation assessed with MAKE at the end of ICU and hospital stays and 90 days after ICU admission.

2.6. Statistical Analysis

Patient characteristics were expressed as number (percentage) for categorical variables and median (interquartile range (IQR)) for continuous variables. Comparisons were made with exact Fisher tests for categorical variables and Wilcoxon tests for continuous variables.
Univariate followed by multivariate logistic regression analyses were performed to identify risk factors of EO-AKI. We used conditional stepwise variable selection with 0.2 as the critical p-value for entry into the model and 0.05 as the p-value for removal. Interactions and correlations between the explanatory variables were carefully checked. Longitudinal data were categorized according to their median. Data are given as odds ratio (ORs) and their 95% confidence intervals (CI).
A competing risk analysis was performed to assess cumulative renal recovery. The concomitant competing risks taken into account were discharge alive from the ICU and ICU mortality. The Fine and Gray model [33] was used to assess risk factors of renal recovery. We also used a conditional stepwise variable selection with 0.2 as the critical p-value for entry into the model and 0.05 as the p-value for removal. It was pre-planned to force the “AKI stage” in the model if this variable was not selected as sensitivity analysis. Data are given as subdistribution hazard ratios (sHRs) and their 95% CI. Proportionality of the risks was also carefully checked.
All reported probability values are two-tailed, and p-value < 0.05 was considered to be statistically significant. Missing data were imputed linearly. All analyses were performed with SAS software version 9.4 (SAS Institute Inc., Cary, NC, USA) or R Statistical Software (v4.2.1; R Core Team 2021, Vienna, Austria).

3. Results

3.1. Initial Characteristics

Of the 336 patients screened, 266 were finally included in our study (Figure 1). The median time between symptom onset and ICU admission was 9 days (IQR, 6–11 d) and between hospital and ICU admission was 2 days (1–5). The median age of the patients was 69 years (61–75), of whom 186 (70%) were of male gender, 50 (18.8%) had a cardiovascular disease and 40 (15%) had diabetes mellitus. Initial severity according to simplified acute physiology score II (SAPS II) and sequential organ failure assessment (SOFA) score was 36 (29–45) and 4 (3–6), respectively; PaO2/FiO2 was 98 (70–210), and 31 (11.6%) patients required vasopressors and invasive mechanical ventilation. Patient characteristics are detailed in Table 1.

3.2. Prevalence, Illness Severity Associated with, and Risk Factors for, EO-AKI

The cumulative incidence of EO-AKI and that of EO-AKI recovery, taking into account competing events, are shown in Figure 2, panels A and B, respectively.
On ICU admission, EO-AKI was recorded in 84 patients (31.5%), of whom 42 (50%) had AKI stage 1, 17 (20.2%) had AKI stage 2 and 25 (29.7%) had AKI stage 3 (Figure 1). There was no significant difference in clinical characteristics between patients with and without EO-AKI, except for age (73.7 years (65.2; 76.9) vs. 67.2 years (59.8–73.3), p < 0.01), and time from first symptoms to ICU admission (10 days (8–12) vs. 8 days (4.5–10), p < 0.01).
On ICU admission, patients with EO-AKI had greater illness severity than patients without as evidenced by a higher SAPS II score (42 (33–50) vs. 34 (28–41), p < 0.01) and a greater need for vasopressors (25 (29.8%) vs. 6 (3.3%), p < 0.01) and invasive mechanical ventilation (22 (26.2%) vs. 8 (4.4%), p < 0.01). They had also received aminoglycosides more often before ICU admission (8 (9.5%) vs. 1 (0.6%), p < 0.01) and had higher SCr values (119 µmol/L (86–161) vs. 74 (61–89), p < 0.01) and higher serum levels of procalcitonin (0.5 g/L (0.2–2.2) vs. 0.2 g/L (0.1–0.4)).
Other biological inflammatory factors such as serum values of D-dimers, ferritin and C-reactive protein (CRP) were not associated with EO-AKI. The median length of ICU stay was significantly longer in patients with EO-AKI (10.5 days (7–16)) than without (6 days (4–10), p < 0.01) (Table 1).
Overall, EO-AKI patients developed greater illness severity than patients without, as shown by worse oxygenation and higher needs in invasive mechanical ventilation and vasopressors (all p < 0.01). Higher ICU and hospital mortality rates were observed in patients with EO-AKI (47 (56%) and 52 (61.9%)) than in patients without EO-AKI (32 (17.6%) and 38 (20.8%)), (all p < 0.01). The higher the EO-AKI stage, the greater the mortality (Table 2).
Of note, patients with EO-AKI who required RRT during ICU stay, compared to patients who required RRT during ICU but without EO-AKI (late-AKI) were quite similar in terms of age and comorbidities. However, patients with EO-AKI and RRT had more severe disease on ICU admission. The day-90 death rate in these two subgroups was high: 21 (87.5%) for EO-AKI and 9 (81.82%) for late-AKI (Table S2).
Multivariable analysis showed that risk factors associated with EO-AKI included age, impaired immune system, being under vasopressors on admission, higher serum procalcitonin levels and exposure to aminoglycosides prior to ICU admission. In contrast, we identified that time between symptom onset and ICU admission >10 days was a protective factor against EO-AKI (Figure 3 and Figure S1, Table S2).

3.3. EO-AKI Renal Recovery and Risk Factors

EO-AKI was classified as transient or persistent and AKD in 40 (47.6%), 15 (17.8%) and 29 (34.5%) patients, respectively (Figure 1). The cumulative incidence of EO-AKI recovery in patients with EO-AKI was 54.3% (53.7–54.9) at day 5, and 72.8% (72.3–73.3) at day 20.
Patients who developed AKD had higher ICU and hospital mortality than patients without AKI, 21 (72.4%) vs. 32 (17.6%), (p < 0.01) and 23 (79.3%) vs. 38 (20.9%), (p < 0.01), respectively. ICU mortality was higher in patients with AKD than in patients with transient and persistent AKI, 21 (72.4%) vs. 7 (46.7%) and 19 (47.5%), respectively, (p < 0.01). Patients who developed AKD more often had EO-AKI stage 3 (Table 3).
EO-AKI recovery was independently associated with the initial staging of EO-AKI (stage 3 vs. stage 1, sHR, 0.11; 95% CI, 0.05–0.22), and exposure to diuretics before ICU admission (sHR, 0.39; 95% CI, 0.26–0.6) (Figure 4 and Table S3).

3.4. Outcome of EO-AKI and of EO-AKI Recovery

Patients with EO-AKI had lower survival at day 90 than patients without, 49/76 (64.5%) vs. 38/168 (22.6%) (p < 0.01), respectively. The 90-day mortality increased with EO-AKI staging: 22/39 (56.4%), 9/15 (60%) and 18/22 (81.8%) in patients with EO-AKI stages 1, 2 or 3, respectively (p < 0.01) (Table 2).
During ICU stay, we identified 38 (14.2%) patients who needed renal replacement therapy, of whom 25 were EO-AKI patients. At day 90, no EO-AKI patients required RRT but 10.8% had a GFR < 60 mL/min.
Among EO-AKI patients, lack of renal recovery was more frequently observed in those with EO-AKI stage 3 (Table S4). The 90-day mortality was lower in patients with transient or persistent EO-AKI than in patients who developed AKD, 20/36 (55.6%), 8/14 (57.1%) and 21/26 (80.8%) (p < 0.01), respectively (Table 3). MAKE-90 occurred in 104/244 (42.6%) of all patients.

4. Discussion

To our knowledge, this study is one of the first to characterize EO-AKI in critically ill COVID-19 patients and to assess the relationship between time to recovery and outcome.
First, we confirmed that EO-AKI is a frequent complication of COVID-19 in critically ill patients [15]. The timing of studies according to the outbreak wave and WHO treatment recommendations, the diversity of the case-mix populations (restricted or not to patients on invasive oxygen therapy, for instance), the differences in the definitions of AKI (based on both diuresis and SCr criteria or on SCr criteria alone) and of AKI onset make comparisons between studies on this topic difficult.
In our study, we observed a −31% prevalence of AKI, which is lower than the rates reported in most recent European studies, such as those of Lumlertgul et al. in the United Kingdom [10], who found an AKI prevalence of 76.6%, or of Arrestier et al. [14] in France with an AKI prevalence of 64.3%. In the study by Geri et al. [34], half of the cohort of 379 patients developed AKI in the first 7 days of admission. These studies used a longer time window of up to 21 days, which could be an explanation for the differences in AKI rates. In contrast, in a cohort study performed by the COVID-19 Critical Care Consortium and using the maximum SCr on day 1–2 of invasive mechanical ventilation to define EO-AKI, the rate of EO-AKI was 20.9%.
It is noteworthy that most patients in the SarSCoV-AKI cohort were recruited after August 2020, hence after the first wave in France. As a consequence, they received the recommended treatment of steroids and reinforced anticoagulation, and in most cases the intubation strategy was performed later than during the first wave [35,36]. The patients in our cohort were also probably less severely ill at ICU admission than in cohorts reporting higher rates of AKI with a lesser need for vasopressors, 11.4% compared to 44.3% in Arrestier et al.’s study [15] or 44.2% in Lumlertgul et al.’s study [11]. We also used only SCr results whereas other studies used both SCr and urine output criteria to define AKI. Collectively, the results might have led to an underestimation of AKI prevalence in our study [37].
Older age, a higher procalcitonin level, a higher SOFA score and use of mechanical ventilation were independently associated with an increased risk of EO-AKI in our cohort. These results are consistent with those of other studies in both SARS-CoV-2 ICU populations [10,15,17] and general ICU patients [38,39], in which a higher severity score, hypertension and older age were frequent risk factors for developing AKI.
Since patients who developed AKI had more antimicrobial therapy, especially aminoglycoside use, more organ failure requiring vasopressors compared to patients without AKI, but also presented more bacteriemia on admission, EO-AKI might be caused by sepsis rather than direct renal effects of the SARS-CoV-2 virus in most cases.
Severe COVID-19 is classically associated with elevated values of systemic non-specific inflammatory biomarkers including CRP, ferritin and D-dimers [40,41]. Numerous studies suggest that in critically ill patients, systemic inflammation contributes to AKI occurrence [10,13,42,43]. In our study, only increased values of serum D-dimers were associated with EO-AKI in univariate analysis, but they were no longer found in multivariate models. This absence of an independent association between systemic inflammation and EO-AKI could result from the widespread use of corticosteroids during severe COVID [44]. Further investigations regarding the relationship between the inflammatory process and EO-AKI in severe COVID are therefore warranted.
Although our mortality rate was high at 90 days in EO-AKI patients (64.5%), only 10.8% of them developed chronic kidney disease, as defined by eGFR < 60 mL/min, and none were dialysis dependent. Other studies showed renal recovery rates between 17% and 84% and dialysis dependence rates between 8% and 56.5% [9,45,46,47]. Lumlertgul et al. [10] reported kidney recovery in 90.9% of survivors at 90 days. The absence of dialysis dependence observed in our study could have been due to the exclusion of patients with pre-existing kidney disease. However, these findings strongly suggest that developing AKD after EO-AKI is often a reversible condition in ICU survivors. In our cohort, most of the patients with AKI stages 2 or 3 and persistent AKI or AKD died, and hence, only less severely ill patients survived and were able to recover from EO-AKI.
In our study, which was performed mainly during waves 2 and 3, we observed a lower rate of transient EO-AKI (47.5%) and a higher rate of patients with non-renal recovery ≥7 days (34.5%) than those reported by Lumlertgul et al. during wave 2 (65% and 10%, respectively) [47]. However, patients in the latter study were more likely to have received mechanical ventilation. We observed a higher rate of patients with non-renal recovery ≥7 days in our cohort. We cannot exclude that these discrepancies were partly due to differences in the definition of renal recovery: we used normalization of SCr while Lumlertgul et al. used SCr < 1.5 times baseline value, a more sensitive definition [48]. Additionally, our study shows that patients who made a rapid renal recovery had a better prognosis since AKD was associated with increased mortality at 90 days and mortality was strongly associated with initial AKI stage. These findings are in line with studies in both the general ICU population [49] and COVID-19 ICU populations [15,50].
We also identified that use of diuretics on ICU admission was a risk factor for non-renal recovery. While diuretics can help prevent lung overload, they could have induced hypovolemia and pre-renal AKI, which might increase the risk of renal failure and delay renal adaptation to injury due to SARS-CoV2, and subsequent recovery. On the other hand, we found that an interval between symptom onset and ICU admission greater than 10 days protected against AKI development. This can be explained by the fact that peak infection usually occurs after 3–7 days [51] and ICU admission often occurs before 10 days of the first symptoms [36], and hence, patients admitted to the ICU after 10 days had probably gone through the most unstable period of the disease.
We found that, after adjusting for cofounders, all EO-AKI stages were associated with higher ICU mortality, which is in line with other reports [10,18,52].
Our study shows that EO-AKI patients had a longer length of stay, a higher rate of invasive mechanical ventilation and a greater need for vasopressors. EO-AKI was strongly associated with mortality, which is consistent with the results of other studies evaluating AKI in ICU-hospitalized patients [9,10,14,15,17].
Our study has several strengths. First, we admitted patients consecutively thus limiting selection bias, and had high quality data, prospectively collected, with almost none missing. In addition, we were able to establish the incidence and outcomes of transient AKI, persistent AKI and AKD. To our knowledge, data reporting renal recovery in patients with SARS-CoV2 pneumonia are scarce [10].
However, our work also has certain limitations. First, it was a single-centre observational study, which could impede the generalization of the results. Second, we might have over- or underestimated the prevalence of AKI. True baseline SCr was known for only 55% of patients and if the baseline value was unknown, SCr was estimated using the MDRD formula. In accordance with French legislation, we did not record patient ethnicity, and back-calculated SCr on the assumption that all patients were Caucasians. In addition, we did not define AKI according to the urine output criteria of the KDIGO definition, which could also have biased the true prevalence of AKI. Third, we might have either overestimated the prevalence of AKD, since it was defined by the absence of SCr normalization following EO-AKI and not by a decrease in SCr < 1.5-fold baseline value, or underestimated it since we did not take into account the markers of kidney damage to diagnose AKD. Similarly, since markers of kidney damage were not recorded, the MAKE criterion “persistence of renal failure” was defined by the functional criterion for kidney diseases and disorders: eGFR < 60 mL/min/1.73 m2 [28]. Finally, as previously mentioned, follow-up was only 3 months, which is a short period to evaluate renal prognosis, and further studies are needed to evaluate long-term kidney outcomes in COVID ICU survivors after EO-AKI [47,53]. Additional studies to assess long-term renal prognosis should also be undertaken.

5. Conclusions

To conclude, we found that one third of critically ill patients with severe COVID-19 developed EO-AKI. EO-AKI stage 3 or AKD were associated with fatal outcome in around two-thirds of the cases at 3 months. EO-AKI stage 1 or transient AKI were also associated with a higher death rate in half of the cases. Monitoring and preserving kidney function should be considered for all patients admitted to the ICU for SARS-CoV-2 pneumonia.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biomedicines11041001/s1, Figure S1: Correlation coefficient between covariates on admission; Table S1: AKI prevalence and mortality rate in the ICU; Table S2: Characteristics of patients depending on occurrence of early onset and late AKI and requirement for renal replacement therapy; Table S3: Factors associated with occurrence of Early-Onset Acute Kindey Injury, uni and then multivariate logistic regression analyses; Table S4: Factors associated with renal recovery after Early-Onset Acute Kidney Injury, uni and then sub distribution, survival analyses.

Author Contributions

Conceptualization, A.R., C.P., B.S. and C.D.; methodology, B.S. and C.D.; software, C.D.; validation, B.S. and C.D.; formal analysis, C.D.; investigation, B.S. and C.D.; resources, C.P., M.A. and B.S.; data curation, A.R, C.D., V.S., B.E., R.B., L.C., F.T., K.G., B.B. and M.A.; writing—original draft preparation, A.R., C.P., B.S. and C.D.; writing—review and editing, C.P., V.S. and B.E., R.B., L.C., F.T., K.G., B.B. and M.A.; visualization, C.D.; supervision, B.S.; project administration, M.A.; funding acquisition, B.S., M.A. and C.P. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by a grant from the Michelin Corporate Foundation. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Institutional Review Board Statement

The SarSCoV-AKI study received approval from the ethics committee of the French Intensive Care Society in 2020 (CE-SRLF20-21) in accordance with our local regulations. All patients or close relatives were informed that their data would be included in the SarSCoV-AKI cohort study and gave written informed consent for the storage and research use of residual blood from samples collected as part of routine care. All the procedures were followed in accordance with the ethical standards of the responsible committee on human experimentation (institutional or regional) and with the Helsinki Declaration of 1975.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data can be provided under request at the corresponding author.

Acknowledgments

We thank Jeffrey Watts for help in preparation of the English manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Noble, R.A.; Selby, N.M. The Changing Nature of COVID-19-Associated AKI: Where Are We Now? Nephrol. Dial. Transpl. 2022, 37, 201–202. [Google Scholar] [CrossRef] [PubMed]
  2. Nadim, M.K.; Forni, L.G.; Mehta, R.L.; Connor, M.J.; Liu, K.D.; Ostermann, M.; Rimmelé, T.; Zarbock, A.; Bell, S.; Bihorac, A.; et al. COVID-19-Associated Acute Kidney Injury: Consensus Report of the 25th Acute Disease Quality Initiative (ADQI) Workgroup. Nat. Rev. Nephrol. 2020, 16, 747–764. [Google Scholar] [CrossRef] [PubMed]
  3. Drake, T.M.; Riad, A.M.; Fairfield, C.J.; Egan, C.; Knight, S.R.; Pius, R.; Hardwick, H.E.; Norman, L.; Shaw, C.A.; McLean, K.A.; et al. Characterisation of In-Hospital Complications Associated with COVID-19 Using the ISARIC WHO Clinical Characterisation Protocol UK: A Prospective, Multicentre Cohort Study. Lancet 2021, 398, 223–237. [Google Scholar] [CrossRef] [PubMed]
  4. Palevsky, P.M. COVID-19 and AKI: Where Do We Stand? J. Am. Soc. Nephrol. 2021, 32, 1029–1032. [Google Scholar] [CrossRef]
  5. Chen, Y.-T.; Shao, S.-C.; Hsu, C.-K.; Wu, I.-W.; Hung, M.-J.; Chen, Y.-C. Incidence of Acute Kidney Injury in COVID-19 Infection: A Systematic Review and Meta-Analysis. Crit. Care 2020, 24, 346. [Google Scholar] [CrossRef]
  6. Chaibi, K.; Dao, M.; Pham, T.; Gumucio-Sanguino, V.D.; Di Paolo, F.A.; Pavot, A.; Cohen, Y.; Dreyfuss, D.; Pérez-Fernandez, X.; Gaudry, S. Severe Acute Kidney Injury in Patients with COVID-19 and Acute Respiratory Distress Syndrome. Am. J. Respir. Crit. Care Med. 2020, 202, 1299–1301. [Google Scholar] [CrossRef]
  7. Bowe, B.; Cai, M.; Xie, Y.; Gibson, A.K.; Maddukuri, G.; Al-Aly, Z. Acute Kidney Injury in a National Cohort of Hospitalized US Veterans with COVID-19. Clin. J. Am. Soc. Nephrol. 2020, 16, 14–25. [Google Scholar] [CrossRef]
  8. Fu, E.L.; Janse, R.J.; de Jong, Y.; van der Endt, V.H.W.; Milders, J.; van der Willik, E.M.; de Rooij, E.N.M.; Dekkers, O.M.; Rotmans, J.I.; van Diepen, M. Acute Kidney Injury and Kidney Replacement Therapy in COVID-19: A Systematic Review and Meta-Analysis. Clin. Kidney J. 2020, 13, 550–563. [Google Scholar] [CrossRef]
  9. Lowe, R.; Ferrari, M.; Nasim-Mohi, M.; Jackson, A.; Beecham, R.; Veighey, K.; Cusack, R.; Richardson, D.; Grocott, M.; Levett, D.; et al. Clinical Characteristics and Outcome of Critically Ill COVID-19 Patients with Acute Kidney Injury: A Single Centre Cohort Study. BMC Nephrol. 2021, 22, 92. [Google Scholar] [CrossRef]
  10. Lumlertgul, N.; Pirondini, L.; Cooney, E.; Kok, W.; Gregson, J.; Camporota, L.; Lane, K.; Leach, R.; Ostermann, M. Acute Kidney Injury Prevalence, Progression and Long-Term Outcomes in Critically Ill Patients with COVID-19: A Cohort Study. Ann. Intensive Care 2021, 11, 123. [Google Scholar] [CrossRef]
  11. da Costa, R.L.; Sória, T.C.; Salles, E.F.; Gerecht, A.V.; Corvisier, M.F.; Menezes, M.A. de M.; Ávila, C. da S.; Silva, E.C. de F.; Pereira, S.R.N.; Simvoulidis, L.F.N. Acute Kidney Injury in Patients with Covid-19 in a Brazilian ICU: Incidence, Predictors and in-Hospital Mortality. J. Bras. Nefrol. 2021, 43, 349–358. [Google Scholar] [CrossRef]
  12. Alessandri, F.; Pistolesi, V.; Manganelli, C.; Ruberto, F.; Ceccarelli, G.; Morabito, S.; Pugliese, F. Policlinico Umberto I COVID-19 Group Acute Kidney Injury and COVID-19: A Picture from an Intensive Care Unit. Blood Purif. 2021, 50, 767–771. [Google Scholar] [CrossRef]
  13. De Almeida, D.C.; do Franco, M.C.P.; dos Santos, D.R.P.; Santos, M.C.; Maltoni, I.S.; Mascotte, F.; de Souza, A.A.; Pietrobom, P.M.; Medeiros, E.A.; Ferreira, P.R.A.; et al. Acute Kidney Injury: Incidence, Risk Factors, and Outcomes in Severe COVID-19 Patients. PLoS ONE 2021, 16, e0251048. [Google Scholar] [CrossRef]
  14. Arrestier, R.; Gendreau, S.; Mokrani, D.; Bastard, J.-P.; Fellahi, S.; Bagate, F.; Masi, P.; d’Humières, T.; Razazi, K.; Carteaux, G.; et al. Acute Kidney Injury in Critically-Ill COVID-19 Patients. J. Clin. Med. 2022, 11, 2029. [Google Scholar] [CrossRef]
  15. Schaubroeck, H.; Vandenberghe, W.; Boer, W.; Boonen, E.; Dewulf, B.; Bourgeois, C.; Dubois, J.; Dumoulin, A.; Fivez, T.; Gunst, J.; et al. Acute Kidney Injury in Critical COVID-19: A Multicenter Cohort Analysis in Seven Large Hospitals in Belgium. Crit. Care 2022, 26, 225. [Google Scholar] [CrossRef]
  16. Kolhe, N.V.; Fluck, R.J.; Selby, N.M.; Taal, M.W. Acute Kidney Injury Associated with COVID-19: A Retrospective Cohort Study. PLoS Med. 2020, 17, e1003406. [Google Scholar] [CrossRef]
  17. Hirsch, J.S.; Ng, J.H.; Ross, D.W.; Sharma, P.; Shah, H.H.; Barnett, R.L.; Hazzan, A.D.; Fishbane, S.; Jhaveri, K.D.; Northwell COVID-19 Research Consortium; et al. Acute Kidney Injury in Patients Hospitalized with COVID-19. Kidney Int. 2020, 98, 209–218. [Google Scholar] [CrossRef] [PubMed]
  18. Cheng, Y.; Luo, R.; Wang, K.; Zhang, M.; Wang, Z.; Dong, L.; Li, J.; Yao, Y.; Ge, S.; Xu, G. Kidney Disease Is Associated with In-Hospital Death of Patients with COVID-19. Kidney Int. 2020, 97, 829–838. [Google Scholar] [CrossRef] [PubMed]
  19. Pan, X.-W.; Xu, D.; Zhang, H.; Zhou, W.; Wang, L.-H.; Cui, X.-G. Identification of a Potential Mechanism of Acute Kidney Injury during the COVID-19 Outbreak: A Study Based on Single-Cell Transcriptome Analysis. Intensive Care Med. 2020, 46, 1114–1116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Sharma, P.; Uppal, N.N.; Wanchoo, R.; Shah, H.H.; Yang, Y.; Parikh, R.; Khanin, Y.; Madireddy, V.; Larsen, C.P.; Jhaveri, K.D.; et al. COVID-19-Associated Kidney Injury: A Case Series of Kidney Biopsy Findings. J. Am. Soc. Nephrol. 2020, 31, 1948–1958. [Google Scholar] [CrossRef] [PubMed]
  21. Joseph, A.; Zafrani, L.; Mabrouki, A.; Azoulay, E.; Darmon, M. Acute Kidney Injury in Patients with SARS-CoV-2 Infection. Ann. Intensive Care 2020, 10, 117. [Google Scholar] [CrossRef]
  22. Gabarre, P.; Dumas, G.; Dupont, T.; Darmon, M.; Azoulay, E.; Zafrani, L. Acute Kidney Injury in Critically Ill Patients with COVID-19. Intensive Care Med. 2020, 46, 1339–1348. [Google Scholar] [CrossRef]
  23. Perinel, S.; Vincent, F.; Lautrette, A.; Dellamonica, J.; Mariat, C.; Zeni, F.; Cohen, Y.; Tardy, B.; Souweine, B.; Darmon, M. Transient and Persistent Acute Kidney Injury and the Risk of Hospital Mortality in Critically Ill Patients: Results of a Multicenter Cohort Study. Crit. Care Med. 2015, 43, e269–e275. [Google Scholar] [CrossRef]
  24. Darmon, M.; Truche, A.-S.; Abdel-Nabey, M.; Schnell, D.; Souweine, B. Early Recognition of Persistent Acute Kidney Injury. Semin. Nephrol. 2019, 39, 431–441. [Google Scholar] [CrossRef]
  25. Chawla, L.S.; Bellomo, R.; Bihorac, A.; Goldstein, S.L.; Siew, E.D.; Bagshaw, S.M.; Bittleman, D.; Cruz, D.; Endre, Z.; Fitzgerald, R.L.; et al. Acute Kidney Disease and Renal Recovery: Consensus Report of the Acute Disease Quality Initiative (ADQI) 16 Workgroup. Nat. Rev. Nephrol. 2017, 13, 241–257. [Google Scholar] [CrossRef] [Green Version]
  26. Abdel-Nabey, M.; Ghrenassia, E.; Mariotte, E.; Valade, S.; Morel, G.; Lemiale, V.; Zafrani, L.; Azoulay, E.; Darmon, M. Acute Kidney Injury Recovery Patterns in Critically Ill Patients: Results of a Retrospective Cohort Study. Crit. Care Med. 2021, 49, e683–e692. [Google Scholar] [CrossRef]
  27. Ng, J.H.; Hirsch, J.S.; Hazzan, A.; Wanchoo, R.; Shah, H.H.; Malieckal, D.A.; Ross, D.W.; Sharma, P.; Sakhiya, V.; Fishbane, S.; et al. Outcomes Among Patients Hospitalized With COVID-19 and Acute Kidney Injury. Am. J. Kidney Dis. 2021, 77, 204–215.e1. [Google Scholar] [CrossRef]
  28. Kellum, J.A.; Lameire, N. KDIGO AKI Guideline Work Group Diagnosis, Evaluation, and Management of Acute Kidney Injury: A KDIGO Summary (Part 1). Crit Care 2013, 17, 204. [Google Scholar] [CrossRef] [Green Version]
  29. Palevsky, P.M. Endpoints for Clinical Trials of Acute Kidney Injury. Nephron 2018, 140, 111–115. [Google Scholar] [CrossRef]
  30. Prischl, F.C.; Rossing, P.; Bakris, G.; Mayer, G.; Wanner, C. Major Adverse Renal Events (MARE): A Proposal to Unify Renal Endpoints. Nephrol. Dial. Transpl. 2021, 36, 491–497. [Google Scholar] [CrossRef]
  31. Calandra, T.; Cohen, J. International Sepsis Forum Definition of Infection in the ICU Consensus Conference The International Sepsis Forum Consensus Conference on Definitions of Infection in the Intensive Care Unit. Crit. Care Med. 2005, 33, 1538–1548. [Google Scholar] [CrossRef] [PubMed]
  32. Plachouras, D.; Lepape, A.; Suetens, C. ECDC Definitions and Methods for the Surveillance of Healthcare-Associated Infections in Intensive Care Units. Intensive Care Med. 2018, 44, 2216–2218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lau, B.; Cole, S.R.; Gange, S.J. Competing Risk Regression Models for Epidemiologic Data. Am. J. Epidemiol. 2009, 170, 244–256. [Google Scholar] [CrossRef] [PubMed]
  34. Geri, G.; Darmon, M.; Zafrani, L.; Fartoukh, M.; Voiriot, G.; Le Marec, J.; Nemlaghi, S.; Vieillard-Baron, A.; Azoulay, E. Acute Kidney Injury in SARS-CoV2-Related Pneumonia ICU Patients: A Retrospective Multicenter Study. Ann. Intensive Care 2021, 11, 86. [Google Scholar] [CrossRef]
  35. Hernandez-Romieu, A.C.; Adelman, M.W.; Hockstein, M.A.; Robichaux, C.J.; Edwards, J.A.; Fazio, J.C.; Blum, J.M.; Jabaley, C.S.; Caridi-Scheible, M.; Martin, G.S.; et al. Timing of Intubation and Mortality Among Critically Ill Coronavirus Disease 2019 Patients: A Single-Center Cohort Study. Crit. Care Med. 2020, 48, e1045–e1053. [Google Scholar] [CrossRef]
  36. COVID-ICU Group on behalf of the REVA Network and the COVID-ICU Investigators. Clinical Characteristics and Day-90 Outcomes of 4244 Critically Ill Adults with COVID-19: A Prospective Cohort Study. Intensive Care Med. 2021, 47, 60–73. [Google Scholar] [CrossRef]
  37. Khwaja, A. KDIGO Clinical Practice Guidelines for Acute Kidney Injury. Nephron Clin. Pract. 2012, 120, c179–c184. [Google Scholar] [CrossRef]
  38. Uchino, S.; Kellum, J.A.; Bellomo, R.; Doig, G.S.; Morimatsu, H.; Morgera, S.; Schetz, M.; Tan, I.; Bouman, C.; Macedo, E.; et al. Acute Renal Failure in Critically Ill Patients: A Multinational, Multicenter Study. JAMA 2005, 294, 813–818. [Google Scholar] [CrossRef] [Green Version]
  39. Mehta, R.L.; Pascual, M.T.; Gruta, C.G.; Zhuang, S.; Chertow, G.M. Refining Predictive Models in Critically Ill Patients with Acute Renal Failure. J. Am. Soc. Nephrol. 2002, 13, 1350–1357. [Google Scholar] [CrossRef] [Green Version]
  40. Hachim, I.Y.; Hachim, M.Y.; Hannawi, H.; Naeem, K.B.; Salah, A.; Hannawi, S. The Inflammatory Biomarkers Profile of Hospitalized Patients with COVID-19 and Its Association with Patient’s Outcome: A Single Centered Study. PLoS ONE 2021, 16, e0260537. [Google Scholar] [CrossRef]
  41. Iwamura, A.P.D.; Tavares da Silva, M.R.; Hümmelgen, A.L.; Soeiro Pereira, P.V.; Falcai, A.; Grumach, A.S.; Goudouris, E.; Neto, A.C.; Prando, C. Immunity and Inflammatory Biomarkers in COVID-19: A Systematic Review. Rev. Med. Virol. 2021, 31, e2199. [Google Scholar] [CrossRef]
  42. Cheng, Y.; Zhang, N.; Luo, R.; Zhang, M.; Wang, Z.; Dong, L.; Li, J.; Zeng, R.; Yao, Y.; Ge, S.; et al. Risk Factors and Outcomes of Acute Kidney Injury in Critically Ill Patients with Coronavirus Disease 2019. Kidney Dis. 2021, 7, 111–119. [Google Scholar] [CrossRef]
  43. Hardenberg, J.-H.B.; Stockmann, H.; Aigner, A.; Gotthardt, I.; Enghard, P.; Hinze, C.; Balzer, F.; Schmidt, D.; Zickler, D.; Kruse, J.; et al. Critical Illness and Systemic Inflammation Are Key Risk Factors of Severe Acute Kidney Injury in Patients With COVID-19. Kidney Int. Rep. 2021, 6, 905–915. [Google Scholar] [CrossRef]
  44. Wu, C.; Hou, D.; Du, C.; Cai, Y.; Zheng, J.; Xu, J.; Chen, X.; Chen, C.; Hu, X.; Zhang, Y.; et al. Corticosteroid Therapy for Coronavirus Disease 2019-Related Acute Respiratory Distress Syndrome: A Cohort Study with Propensity Score Analysis. Crit. Care 2020, 24, 643. [Google Scholar] [CrossRef]
  45. Eriksson, K.E.; Campoccia-Jalde, F.; Rysz, S.; Rimes-Stigare, C. Continuous Renal Replacement Therapy in Intensive Care Patients with COVID-19; Survival and Renal Recovery. J. Crit. Care 2021, 64, 125–130. [Google Scholar] [CrossRef]
  46. Gupta, S.; Coca, S.G.; Chan, L.; Melamed, M.L.; Brenner, S.K.; Hayek, S.S.; Sutherland, A.; Puri, S.; Srivastava, A.; Leonberg-Yoo, A.; et al. AKI Treated with Renal Replacement Therapy in Critically Ill Patients with COVID-19. J. Am. Soc. Nephrol. 2021, 32, 161–176. [Google Scholar] [CrossRef]
  47. Matsumoto, K.; Prowle, J.R. COVID-19-Associated AKI. Curr. Opin. Crit. Care 2022, 28, 630–637. [Google Scholar] [CrossRef]
  48. Lumlertgul, N.; Baker, E.; Pearson, E.; Dalrymple, K.V.; Pan, J.; Jheeta, A.; Weerapolchai, K.; Wang, Y.; Leach, R.; Barrett, N.A.; et al. Changing Epidemiology of Acute Kidney Injury in Critically Ill Patients with COVID-19: A Prospective Cohort. Ann. Intensive Care 2022, 12, 118. [Google Scholar] [CrossRef]
  49. Kellum, J.A.; Sileanu, F.E.; Bihorac, A.; Hoste, E.A.J.; Chawla, L.S. Recovery after Acute Kidney Injury. Am. J. Respir. Crit. Care Med 2017, 195, 784–791. [Google Scholar] [CrossRef] [Green Version]
  50. Lim, J.-H.; Park, S.-H.; Jeon, Y.; Cho, J.-H.; Jung, H.-Y.; Choi, J.-Y.; Kim, C.-D.; Lee, Y.-H.; Seo, H.; Lee, J.; et al. Fatal Outcomes of COVID-19 in Patients with Severe Acute Kidney Injury. J. Clin. Med. 2020, 9, 1718. [Google Scholar] [CrossRef]
  51. Johansson, M.A.; Quandelacy, T.M.; Kada, S.; Prasad, P.V.; Steele, M.; Brooks, J.T.; Slayton, R.B.; Biggerstaff, M.; Butler, J.C. SARS-CoV-2 Transmission From People Without COVID-19 Symptoms. JAMA Netw. Open 2021, 4, e2035057. [Google Scholar] [CrossRef] [PubMed]
  52. Pei, G.; Zhang, Z.; Peng, J.; Liu, L.; Zhang, C.; Yu, C.; Ma, Z.; Huang, Y.; Liu, W.; Yao, Y.; et al. Renal Involvement and Early Prognosis in Patients with COVID-19 Pneumonia. J. Am. Soc. Nephrol. 2020, 31, 1157–1165. [Google Scholar] [CrossRef] [PubMed]
  53. Heung, M.; Steffick, D.E.; Zivin, K.; Gillespie, B.W.; Banerjee, T.; Hsu, C.-Y.; Powe, N.R.; Pavkov, M.E.; Williams, D.E.; Saran, R.; et al. Acute Kidney Injury Recovery Pattern and Subsequent Risk of CKD: An Analysis of Veterans Health Administration Data. Am. J. Kidney Dis. 2016, 67, 742–752. [Google Scholar] [CrossRef] [PubMed]
Figure 1. COVID-19 patient flow diagram. ICU: intensive care unit; AKI: acute kidney injury; KDIGO: Kidney Disease Improving Global Outcomes.
Figure 1. COVID-19 patient flow diagram. ICU: intensive care unit; AKI: acute kidney injury; KDIGO: Kidney Disease Improving Global Outcomes.
Biomedicines 11 01001 g001
Figure 2. Cumulative incidence of early-onset acute kidney injury (EO-AKI) (A) and renal recovery (B) taking into account competing risks of discharge alive and mortality.
Figure 2. Cumulative incidence of early-onset acute kidney injury (EO-AKI) (A) and renal recovery (B) taking into account competing risks of discharge alive and mortality.
Biomedicines 11 01001 g002
Figure 3. Factors associated with occurrence of AKI, multivariate analyses, logistic regression model. Variables with a p-value < 0.1 in the univariate analyses were age, cardiovascular disease, impaired immune system, time from first symptoms to ICU admission, antimicrobial therapy, aminoglycosides, vasopressors, invasive mechanical ventilation, bacteriemia on admission and procalcitonin. Because of correlations, only the following covariates were tested in multivariate analysis: age, cardiovascular disease, impaired immune system, time from first symptoms to ICU admission, aminoglycosides, vasopressors and procalcitonin.
Figure 3. Factors associated with occurrence of AKI, multivariate analyses, logistic regression model. Variables with a p-value < 0.1 in the univariate analyses were age, cardiovascular disease, impaired immune system, time from first symptoms to ICU admission, antimicrobial therapy, aminoglycosides, vasopressors, invasive mechanical ventilation, bacteriemia on admission and procalcitonin. Because of correlations, only the following covariates were tested in multivariate analysis: age, cardiovascular disease, impaired immune system, time from first symptoms to ICU admission, aminoglycosides, vasopressors and procalcitonin.
Biomedicines 11 01001 g003
Figure 4. Factors associated with recovery from AKI, multivariate analyses, sub-distribution model. AKI: acute kidney injury; SubHR: sub hazard ratio. Variables with a p-value <0.1 in univariate analyses were age, lopinavir–ritonavir, diuretics, vasopressors and KDIGO stage. These covariates were considered into the final model.
Figure 4. Factors associated with recovery from AKI, multivariate analyses, sub-distribution model. AKI: acute kidney injury; SubHR: sub hazard ratio. Variables with a p-value <0.1 in univariate analyses were age, lopinavir–ritonavir, diuretics, vasopressors and KDIGO stage. These covariates were considered into the final model.
Biomedicines 11 01001 g004
Table 1. Characteristics of critically ill COVID-19 patients according to the occurrence of early-onset acute kidney injury.
Table 1. Characteristics of critically ill COVID-19 patients according to the occurrence of early-onset acute kidney injury.
VARIABLES, n (%) or Median (IQR)AllNo EO-AKIEO-AKI p-Value
Number of patients26618284
Period of admission
Wave 1 (before August 2020)15 (5.6)8 (4.4)7 (8.3)0.42
Wave 2 (between August 2020 and December 2020)101 (38)69 (37.9)32 (38.1)
Wave 3 (after January 2021)150 (56.4)105 (57.7)45 (53.6)
Age, yrs.68.8 (60.6–74.8)67.2 (59.8–73.3)73.7 (65.2–76.9)<0.01
Sex, men186 (70)128 (70.3)58 (69)0.83
Comorbidities
BMI > 30 kg/m2117 (44)76 (41.8)41 (48.8)0.28
Cardiovascular disease50 (18.8)29 (15.9)21 (25)0.08
Chronic respiratory disease17 (6.4)14 (7.7)3 (3.6)0.20
Impaired immune system 143 (16.2)24 (13.2)19 (22.6)0.05
Diabetes mellitus40 (15)23 (12.6)17 (20.2)0.11
Time from first symptoms to ICU admission (days)9 (6–11)10 (8–12)8 (4.5–10)<0.01
Time from hospital to ICU admission (days)2 (1–5)2 (1–5)2 (1–5)0.46
Treatments before ICU
ACE inhibitors and angiotensin receptor blockers4 (1.1)1 (0.5)3 (3.6)0.06
Lopinavir–ritonavir8 (3)4 (2.2)4 (4.8)0.27
Remdesivir45 (17.2)36 (20.1)9 (10.7)0.06
Steroids226 (85)158 (86.8)68 (81)0.21
Tocilizumab25 (9.4)20 (11)5 (6)0.19
Antimicrobial therapy93 (35.4)53 (29.6)40 (47.6)<0.01
Aminoglycosides9 (3.4)1 (0.6)8 (9.5)<0.01
Vancomycin3 (1.2)2 (1.1)1 (1.2)0.96
Diuretics207 (77.8)142 (78)65 (77.4)0.91
Organ failures at ICU admission
SAPS II36 (29–45)34.5 (28–41)42 (33–50)<0.01
SOFA4 (3–6)4 (3–5)5 (4–9)<0.01
PaO2/FiO298 (70–210)98 (71–248)90 (67–190)0.20
Need for vasopressors31 (11.6)6 (3.3)25 (29.8)<0.01
Invasive mechanical ventilation30 (11.2)8 (4.4)22 (26.2)<0.01
Renal replacement therapy3 (1.2)0 (0)3 (3.6)0.01
Pulmonary bacterial co-infection18 (6.8)12 (6.7)6 (7.1)0.90
Bacteriemia on admission11 (4.2)4(2.2)7(8.3)0.02
Admission laboratory profile
Neutrophils (NA = 20), G/L6.6 (4.8–9.8)6.4 (4.8–8.9)7.6 (4.5–11.9)0.04
Lymphocytes (NA = 19), G/L0.6 (0.4–1)0.7 (0.5–1)0.6 (0.4–0.9)0.50
Creatininemia, µmol/L81.6 (65–105)73.5 (61–89)119 (85.5–161)<0.01
Procalcitonin (NA = 17), g/L0.2 (0.2–0.6)0.2 (0.1–0.4)0.5 (0.2–2.2)<0.01
C Reactive protein (NA = 52), mg/L119 (70–170)113 (70–163)125 (82–171)0.73
D-dimers (NA = 6), µg/L1214 (777–2027)1168 (727–1939)1216 (1000–2900)0.05
Fibrinogen (NA = 4), g/L7.2 (6.2–8)7.2 (6.5–8)7 (5.8–7.9)0.18
Ferritin (NA = 19), ng/mL1144 (644–1945)1130 (684–1850)1177 (584–2142)0.58
Organ failures during ICU stay
Need for vasopressors85 (32)33 (18.1)52 (61.9)<0.01
Invasive mechanical ventilation88 (33)35 (19.2)53 (63.1)<0.01
Renal replacement therapy38 (14.2)13 (7.1)25 (29.8)<0.01
General outcomes
Pulmonary embolism 14 (5.2)9 (5)5 (6)0.74
Ventilator-associated pneumonia35 (13.4)13 (7.3)22 (26.2)<0.01
Decision not to intubate during ICU stay40 (15)18 (9.9)22 (26.2)<0.01
Length of ICU stay, d8 (5–13)6 (4–10)10.5 (7–16)<0.01
Death in ICU79 (29.6)32 (17.6)47 (56)<0.01
Length of hospital stay, d14 (10–23)14 (10–24)15 (9.5–22.5)0.50
In-hospital death90 (33.8)38 (20.8)52 (61.9)<0.01
RRT at hospital discharge 1 (0.6)1 (0.8)0 (0)0.63
General outcomes at 90 days (MAKE-90)
eGFR < 60 mL/min at 90 days17/158 (10.8)9/131 (6.9)8/27 (29.6)<0.01
RRT at 90 days0/1630/1320/31
Death at 90 days87/244 (35.6)38/168 (22.6)49/76 (64.5)<0.01
MAKE-90104/244(42.6)47/168 (28)57/76 (75)<0.01
ACE: angiotensin converting enzyme—AKI, acute kidney injury; BMI, body mass index; ECMO, extracorporeal membrane oxygenation; GFR, glomerular filtration rate; ICU, intensive care unit; NSAID, non-steroidal anti-inflammatory drug; RRT, renal replacement therapy; SAPS II, simplified acute physiology score; SOFA, sequential organ failure assessment. 1 Aplasia (lymphocytes < 1 G/L; or corticosteroids (if treatment duration > 1 month or if treatment amount > 2 mg/kg regardless of duration); or HIV (positive serology); AIDS (positive HIV serology and clinical complications: pneumocystis pneumonia, Kaposi’s sarcoma, tuberculosis, toxoplasmosis.
Table 2. Characteristics of the critically ill COVID-19 patients according to the severity of the acute kidney injury (KDIGO stage).
Table 2. Characteristics of the critically ill COVID-19 patients according to the severity of the acute kidney injury (KDIGO stage).
VARIABLES, n (%) or Median (IQR)No AKIAKI Stage 1AKI Stage 2AKI Stage 3p-Value
Number of patients182421725
Period of admission
Wave 1 (before August 2020)8 (4.4)2 (4.8)1 (5.9)4 (16)0.37
Wave 2 (between August 2020 and December 2020)69 (37.9)18 (42.9)7 (41.2)7 (28)
Wave 3 (after January 2021)105 (57.7)22 (52.4)9 (52.9)14 (56)
Age, yrs.67.2 (59.8–73.3)74.5 (66.4–78.2)74.7 (64.7–77.4)72.7 (63.4–75.1)<0.01
Sex, men128 (70.3)30 (71.4)8 (47.1)20 (80)0.14
BMI, kg/m228.7 (25.1–32.1)27.4 (24.9–31.1)28.3 (26.9–34.7)30.8 (25.5–36)0.52
Comorbidities, no (%)
BMI > 30 kg/m276 (41.8)18 (42.9)8 (47.1)15 (60)0.38
Cardiovascular disease29 (15.9)9 (21.4)4 (23.5)8 (32)0.23
Chronic respiratory disease14 (7.7)1 (2.4)2 (11.8)00.24
Chronic kidney disease2 (1.1)5 (11.9)03 (12)<0.01
Impaired immune system 124 (13.2)10 (23.8)2 (11.8)7 (28)0.12
Diabetes mellitus23 (12.6)9 (21.4)1 (5.9)7 (28)0.09
Time from first symptoms to ICU admission10 (8–12)8.5 (4–11)9 (5–11)6 (5–10)<0.01
Treatment before ICU
ACE inhibitors and angiotensin receptor blockers1 (0.6)2 (4.8)1 (5.9)00.08
Lopinavir–ritonavir4 (2.2)1 (2.4)1 (5.9)2 (8)0.39
Remdesivir36 (20.1)6 (14.3)1 (5.9)2 (8)0.23
Steroids158 (86.8)38 (90.5)14 (82.4)16 (64)0.02
Tocilizumab20 (11)2 (4.8)1 (5.9)2 (8)0.59
Antimicrobial therapy53 (29.6)20 (47.6)7 (41.2)13 (52)0.04
Aminoglycosides1 (0.6)3 (7.1)1 (5.9)4 (16)<0.01
Vancomycin2 (1.1)001 (4)0.48
Diuretics142 (78)36 (85.7)11 (64.7)18 (72)0.30
Organ failures at ICU admission
SAPS II34.5 (28–4)40.5 (3–47)42 (31–50)47 (34–56)<0.01
SOFA4 (3–5)5 (4–7)7 (5–8)9 (4–12)<0.01
PaO2/FiO298 (71–247)82 (67–176)123 (65–243)98 (67–141)0.50
Need for vasopressors6 (3.3)6 (14.3)6 (35.3)13 (52)<0.01
Invasive mechanical ventilation8 (4.4)6 (14.3)6 (35.3)10 (40)<0.01
Renal replacement therapy0003 (12)<0.01
Pulmonary bacterial co-infection12 (6.7)1 (2.4)05 (20)0.03
Bacteriemia on admission4(2.23)4(9.52)03(12)0.03
Admission laboratory profile
Neutrophils (NA = 20), G/L6.4 (4.8–8.9)7.5 (5.5–11.9)5.5 (3.7–14.4)7.8 (4.1–11.8)0.19
Lymphocytes (NA = 19), G/L0.7 (0.49–1.04)0.57 (0.4–0.77)0.86 (0.69–1.64)0.57 (0.36–0.9)0.01
Creatininemia, µmol/L74 (61–89)121 (97–135)91 (64–156)119 (85–202)<0.01
Procalcitonin (NA = 17), g/L0.18 (0.11–0.44)0.38 (0.23–3.69)0.31 (0.15–0.99)0.71 (0.26–2.24)<0.01
C Reactive protein (NA = 52), mg/L113 (70–163)121 (83–158)108 (53–147)150 (45–214)0.72
D-dimers (NA = 6), µg/L1168 (727–1939)1198 (953–2531)1926 (1143–3381)1308 (927–3730)0.19
Fibrinogen (NA = 4), g/L7.2 (6.5–8)7.2 (6.4–7.9)6.3 (5.3–7.5)7 (6–7.9)0.27
Ferritin (NA = 19), ng/mL1130 (684–1850)1129 (528–1846)1253 (535–2429)1258 (681–2753)0.92
Organ failures during ICU stay
Need for vasopressors33 (18.1)18 (42.9)13 (76.5)21 (84)<0.01
Invasive mechanical ventilation35 (19.2)18 (42.9)14 (82.4)21 (84)<0.01
Pulmonary embolism 9 (5.0)3 (7.1)1 (5.9)1 (4)0.94
Ventilator-associated pneumonia13 (7.3)9 (21.4)6 (35.3)7 (28)<0.01
Type of Renal Recovery, no (%)
Transient 32 (76.2)6 (35.3)2 (8)<0.01
Persistent 7 (16.7)6 (35.3)2 (8)
Acute kidney disease 3 (7.1)5 (29.4)21 (84)
General Outcomes
Renal replacement therapy13 (7.1)6 (14.3)4 (23.5)15 (60)<0.01
Decision not to intubate during ICU stay18 (9.9)12 (28.6)5 (29.4)5 (20)0.01
Length of ICU stay, d6 (4–10)9.5 (6–15)11 (7–16)13 (8–19)<0.01
Death in ICU32 (17.6)19 (45.2)9 (52.9)19 (76)<0.01
Length of hospital stay, d 14 (10–24)15.5 (10–22)16 (9–35)15 (10–21)0.86
In-hospital death38 (20.9)22 (52.4)9 (52.9)21 (84)<0.01
RRT at hospital discharge 1 (0.75)0000.97
General Outcomes at 90 days (MAKE-90)
eGFR < 60 mL/min at 90 days9/1316/171/61/4<0.01
RRT at 90 days0/1320/190/70/5
Death at 90 days38/168 (22.6)22/39 (56.4)9/15 (60)18/22 (81.8)<0.01
MAKE-9047/168 (28.0)28/39 (71.8)10/15 (66.7)19/22 (86.4)<0.01
ACE, angiotensin converting enzyme; AKI, acute kidney injury; BMI, body mass index; ECMO, extra corporeal membrane oxygenation; GFR, glomerular filtration rate; ICU, intensive care unit; NSAID, non-steroidal anti-inflammatory drug; RRT, renal replacement therapy; SAPS II, simplified acute physiology score; SOFA, sequential organ failure assessment. 1 Aplasia (lymphocytes < 1 G/L); or corticosteroids (if treatment duration > 1 month or if treatment amount > 2 mg/kg regardless of duration); or HIV (positive serology); AIDS (positive HIV serology and clinical complications: pneumocystis pneumonia, Kaposi’s sarcoma, tuberculosis, toxoplasmosis.
Table 3. Characteristics of the critically ill COVID-19 patients according to renal recovery.
Table 3. Characteristics of the critically ill COVID-19 patients according to renal recovery.
VARIABLES, n (%) or Median (IQR)No AKITransient AKIPersistent AKIAcute Kidney Diseasep-Value
Number of patients182401529
Period of admission
Wave 1 (before August 2020)8 (4.4)1 (2.5)1 (6.7)5 (17.2)0.10
Wave 2 (between August 2020 and December 2020)69 (37.9)14 (35)8 (53.3)10 (34.5)
Wave 3 (after January 2021)105 (57.7)25 (62.5)6 (40)14 (48.3)
Age, yrs.67.2 (59.8–73.3)73.9 (65.2–77.8)73.8 (68.8–75.9)72.81 (63.4–76.4)<0.01
Sex, men128 (70.3)27 (67.5)9 (60)22 (75.9)0.73
BMI, kg/m228.7 (25.1–32.1)27.44 (24.0–31.6)28.3 (26.3–32)30.8 (26.1–36.0)0.57
Comorbidities
BMI > 30 kg/m276 (41.8)17 (42.5)7 (46.7)17 (58.6)0.40
Cardiovascular disease29 (15.9)10 (25)6 (40)5 (17.2)0.09
Chronic respiratory disease14 (7.7)2 (5)01 (3.5)0.55
Chronic kidney disease2 (1.1)6 (15)02 (6.9)<0.01
Impaired immune system 124 (13.2)8 (20)4 (26.7)7 (24.1)0.24
Diabetes mellitus23 (12.6)7 (17.5)2 (13.3)8 (27.6)0.20
Time from first symptoms to ICU admission, d10 (8–12)8 (4–10.5)9 (4–11)7 (5–10)<0.01
Treatment before ICU
ACE inhibitors and angiotensin receptor blockers1 (0.6)1 (2.5)1 (6.7)1 (3.5)0.19
Lopinavir–ritonavir4 (2.2)01 (6.7)3 (10.3)0.06
Remdesivir36 (20.1)6 (15)1 (6.7)2 (6.9)0.21
Steroids158 (86.8)36 (90)11 (73.3)21 (72.4)0.09
Tocilizumab20 (11)4 (10)01 (3.5)0.35
Antimicrobial therapy53 (29.6)19 (47.5)7 (46.7)14 (48.3)0.04
Aminoglycosides1 (0.6)3 (7.5)05 (17.2)<0.01
Vancomycin2 (1.1)001 (3.4)0.57
Diuretics142 (78)30 (75)12 (80)23 (79.3)0.97
Organ failures at ICU admission
SAPS II34.5 (28–41)40.5 (31–46.5)47 (38–58)44 (34–54)<0.01
SOFA4 (3–5)5 (4–7)6 (5–10)8 (4–10)<0.01
PaO2/FiO298 (71.3–247.6)82.5 (70–220)104 (66–200)86.7 (63.8–138.8)0.59
Need for vasopressors6 (3.3)8 (20)5 (33.3)12 (41.4)<0.01
Invasive mechanical ventilation8 (4.4)9 (22.5)2 (13.3)11 (37.9)<0.01
ECMO0001 (3.45)0.04
Pulmonary bacterial co-infection12 (6.7)02 (13.3)4 (13.8)0.11
Bacteriemia on admission4 (2.23)4(10)03(10.34)0.04
Admission laboratory profile
Neutrophils (NA = 20), G/L6.37 (4.76–8.94)7.24 (4.63–11.2)9.75 (5.1–14.44)7.59 (4.09–13.32)0.15
Lymphocytes (NA = 19), G/L0.7 (0.49–1.04)0.64 (0.44–0.82)0.77 (0.37–1.33)0.62 (0.4–0.88)0.71
Creatininemia, µmol/L74 (61–89)122 (92.5–147)122 (88–190)105 (81–176)<0.01
Procalcitonin (NA = 17), g/L0.18 (0.11–0.44)0.38 (0.18–3.43)0.71 (0.31–3.86)0.56 (0.22–2)<0.01
C Reactive protein (NA = 52), mg/L113 (70–163)117 (82–147)108 (83–214)135 (45–175)0.92
D-dimers (NA = 6), µg/L1168 (727–1939)1217 (841.5–2542)1209 (1000–3961)1241 (1100–2041)0.20
Fibrinogen (NA = 4), g/L7.2 (6.5–8)7.2 (5.8–7.7)7.9 (6–9.5)7 (5.8–7.7)0.31
Ferritin (NA = 19), ng/mL1130 (684–1850)1129 (528–2142)1191 (538–1831)1391 (701–2720)0.72
Organ failures during ICU stay
Need for vasopressors33 (18.1)20 (50)9 (60)23 (79.3)<0.01
Invasive mechanical ventilation35 (19.2)21 (52.5)8 (53.3)24 (82.8)<0.01
Pulmonary embolism 9 (5)3 (7.5)1 (6.7)1 (3.5)0.88
Ventilator-associated pneumonia13 (7.3)11 (27.5)3 (20)8 (27.6)<0.01
Severity of Acute Kidney Injury
KDIGO 1 32 (80)7 (46.7)3 (10.3)<0.01
KDIGO 2 6 (15)6 (40)5 (17.2)
KDIGO 3 2 (5)2 (13.3)21 (72.4)
General outcomes
Renal replacement therapy13 (7.1)5 (12.5)3 (20)17 (58.6)<0.01
Decision not to intubate during ICU Stay18 (9.9)12 (30)3 (20)7 (24.1)<0.01
Length of ICU stay, d6 (4–10)9 (6–15.5)10 (8–13)13 (8–19)<0.01
Death in ICU32 (17.6)19 (47.5)7 (46.7)21 (72.4)<0.01
Length of hospital stay, d 14 (10–24)15.5 (10.5–24)16 (9–31)15 (10–21)0.87
In-hospital death38 (20.9)21 (52.5)8 (53.3)23 (79.3)<0.01
RRT at hospital discharge 1 (0.75)0000.97
General outcomes at 90 days (MAKE-90)
eGFR < 60 mL/min at 90 days9/1316/161/61/5<0.01
RRT at 90 days0/1320/180/60/7
Death at 90 days38/168 (22.6)20/36 (55.6)8/14 (57.1)21/26 (80.8)<0.01
MAKE-9047/168 (28.0)26/36 (72.2)9/14 (64.3)22/26 (84.6)<0.01
ACE, angiotensin converting enzyme; AKI, acute kidney injury; BMI, body mass index; ECMO, extra corporeal membrane oxygenation; GFR, glomerular filtration rate; ICU, intensive care unit; NSAID, non-steroidal anti-inflammatory drug; RRT, renal replacement therapy; SAPS II, simplified acute physiology score; SOFA, sequential organ failure assessment. 1 Aplasia (lymphocytes < 1000/mm3); or corticosteroids (if treatment duration > 1 month or if treatment amount > 2 mg/kg regardless of duration); or HIV (positive serology); AIDS (positive HIV serology and clinical complications: pneumocystis pneumonia, Kaposi’s sarcoma, tuberculosis, toxoplasmosis.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ruault, A.; Philipponnet, C.; Sapin, V.; Evrard, B.; Bouzgarrou, R.; Calvet, L.; Thouy, F.; Grapin, K.; Bonnet, B.; Adda, M.; et al. Epidemiology and Outcome of Early-Onset Acute Kidney Injury and Recovery in Critically Ill COVID-19 Patients: A Retrospective Analysis. Biomedicines 2023, 11, 1001. https://doi.org/10.3390/biomedicines11041001

AMA Style

Ruault A, Philipponnet C, Sapin V, Evrard B, Bouzgarrou R, Calvet L, Thouy F, Grapin K, Bonnet B, Adda M, et al. Epidemiology and Outcome of Early-Onset Acute Kidney Injury and Recovery in Critically Ill COVID-19 Patients: A Retrospective Analysis. Biomedicines. 2023; 11(4):1001. https://doi.org/10.3390/biomedicines11041001

Chicago/Turabian Style

Ruault, Alice, Carole Philipponnet, Vincent Sapin, Bertrand Evrard, Radhia Bouzgarrou, Laure Calvet, François Thouy, Kévin Grapin, Benjamin Bonnet, Mireille Adda, and et al. 2023. "Epidemiology and Outcome of Early-Onset Acute Kidney Injury and Recovery in Critically Ill COVID-19 Patients: A Retrospective Analysis" Biomedicines 11, no. 4: 1001. https://doi.org/10.3390/biomedicines11041001

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