Sie sind auf Seite 1von 41

Platelets

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/iplt20

Platelets and renal failure in the SARS-CoV-2


syndrome

Muhanad Taha , Dahlia Sano , Samer Hanoudi , Zahia Esber , Morvarid Elahi ,
Ali Gabali , Teena Chopra , Sorin Draghici & Lobelia Samavati

To cite this article: Muhanad Taha , Dahlia Sano , Samer Hanoudi , Zahia Esber , Morvarid Elahi ,
Ali Gabali , Teena Chopra , Sorin Draghici & Lobelia Samavati (2020): Platelets and renal failure in
the SARS-CoV-2 syndrome, Platelets, DOI: 10.1080/09537104.2020.1817361

To link to this article: https://doi.org/10.1080/09537104.2020.1817361

Published online: 06 Sep 2020.

Submit your article to this journal

Article views: 740

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iplt20
http://www.tandfonline.com/iplt
ISSN: (print), (electronic)

Platelets, Early Online: 1–8


© 2020 Taylor & Francis Group, LLC. DOI: https://doi.org/10.1080/09537104.2020.1817361

Platelets and renal failure in the SARS-CoV-2 syndrome


Muhanad Taha1, Dahlia Sano2, Samer Hanoudi3, Zahia Esber1, Morvarid Elahi4, Ali Gabali5, Teena Chopra5,
Sorin Draghici3, & Lobelia Samavati 1,6
1
Department of Internal Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State University, School of Medicine and Detroit
Medical Center, Detroit, MI, USA, 2Department of Internal Medicine, Division Hematology and Oncology; Wayne State University, School of Medicine
and Detroit Medical Center, Detroit, MI, USA, 3Department of Computer Science, Wayne State University, Detroit, MI, USA, 4Department of Pathology,
Wayne State University, School of Medicine and Detroit Medical Center, Detroit, MI, USA, 5Division of Infectious Diseases, Wayne State University,
Detroit, MI, USA, and 6Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA

Abstract Keywords
The coronavirus disease 19 (COVID-19) is a highly transmittable viral infection caused by the AKI, COVID-19, D-Dimer, hypercoagulable,
severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 utilizes metallo­ mean platelet volume, renal failure, SARS-
carboxyl peptidase angiotensin receptor (ACE) 2 to gain entry into human cells. Activation of CoV-2
several proteases facilitates the interaction of viral spike proteins (S1) and ACE2 receptor. This
leads to cleavage of host ACE2 receptors. ACE2 activity counterbalances the angiotensin II History
effect, its loss may lead to elevated angiotensin II levels with modulation of platelet function,
Received 27 June 2020
size and activity.
Revised 3 August 2020
COVID-19 disease encompasses a spectrum of systemic involvement far beyond respiratory
Accepted 22 August 2020
failure alone. Several features of this disease, including the etiology of acute kidney injury (AKI)
Published online xx xxx xxxx
and the hypercoagulable state, remain poorly understood. Here, we show that there is a high
incidence of AKI (81%) in the critically ill adults with COVID-19 in the setting of elevated
D-dimer, elevated ferritin, C reactive protein (CRP) and lactate dehydrogenase (LDH) levels.
Strikingly, there were unique features of platelets in these patients, including larger, more
granular platelets and a higher mean platelet volume (MPV). There was a significant correlation
between measured D-dimer levels and MVP; but a negative correlation between MPV and
glomerular filtration rates (GFR) in critically ill cohort. Our data suggest that activated platelets
may play a role in renal failure and possibly hypercoagulability status in COVID19 patients.

Introduction SARS‐CoV‐2 is single-stranded positive-sense RNA virus,


containing an approximately 26–32 kilobase (kb) genome [6].
The coronavirus disease 19 (COVID-19) is a highly transmittable
Similar to other CoV, during viral entry into the host cell the
viral infection caused by the severe acute respiratory syndrome
spike proteins (S) on the envelope of SARS‐CoV‐2 are cleaved
coronavirus 2 (SARS-CoV-2) [1]. On presentation, most infected
into S1 and S2 subunits [7]. Through the S1 receptor-binding
individuals exhibit dry cough, dyspnea, and bilateral ground‐glass
domain (RBD), S1 directly binds to the peptidase domain (PD) of
opacities on radiographic images [2]. While the critical impor­
metallocarboxyl peptidase angiotensin receptor (ACE) 2 to gain
tance of respiratory derangement in COVID-19 is well appre­
entry into human cells [7–9]. ACE receptors (ACE and ACE2)
ciated, several clinical features of COVID-19 infection remain
are expressed in almost all tissues and ACE2 is expressed pre­
poorly understood including the increased incidence of throm­
dominantly on the alveolar epithelial type II cells and capillary
boembolic disease despite being on prophylactic anticoagulation
endothelial cells [10,11]. ACE and ACE2 regulate vascular tone,
and acute renal injury requiring renal replacement therapy. For
fibrinolysis and the coagulation cascade [12,13]. ACE activity
instance, based on our observation and those of others, several
increases vascular tone and activates the coagulation cascade by
patients rapidly decompensate days after stabilization of oxygena­
promoting the generation of angiotensin (Ang) II. In contrast,
tion during or after cessation of mechanical ventilation. Findings
ACE2 receptor activity decreases vascular tone and inhibits acti­
of micro and macrothrombi are common in these patients [3].
vation of the coagulation cascade by generating Ang [1–7], which
Others have documented cerebral ischemic infarcts in these sub­
activates a MAS/G protein receptor. SARS‐CoV‐2 infection leads
jects [4]. Microvascular thrombosis appears to be one defining
to loss of function of ACE2 and increased angiotensin II levels
feature of novel SARS-CoV-2 infection [5]. Formation of vascular
[14]. Activation of the renin-angiotensin–aldosterone system
thrombi in association with progressive severe endothelial injury
(RAAS) plays critical role in renal injury [15]. Angiotensin II is
in COVID-19-infected subjects may determine case fatality and
a potent vasoconstrictor and activates the coagulation cascade by
renal dysfunction in this disease.
increasing platelet activation and size, inducing microvascular
thrombosis [12,16].
Correspondence: Lobelia Samavati Department of Internal Medicine, The coagulation cascade, fibrinolytic system and the comple­
Division of Pulmonary, Critical Care and Sleep Medicine, Wayne State ment system closely collaborate to control a wide range of biolo­
University School of Medicine, Detroit, MI 48201, USA gical and pathological functions including immune responses to
ay6003@wayne.edu
2 Platelets, Early Online: 1–8

pathogens, cell migration and tissue hemostasis. A variety of relationship between the clinical variables (including AKI) and
microorganisms can trigger endothelial injury, alternative com­ coagulation profiles. Acute kidney injury is defined as per guide­
plement activation and hence activate the coagulation pathway lines of American society of Nephrology [17]. For all analyses,
perpetuating inflammation and organ dysfunction. We hypothe­ p-values <0.05 were considered significant. For the GFR data,
sized that platelet activation through multiple mechanisms plays a trend analysis was performed on the data from each day for up
an important role in COVID-19 associated coagulopathy and to 8 days. Furthermore, trend analysis was performed on the daily
acute kidney injury (AKI). Our data indicate significant increase data for GFR and MPV. Each interval represents data from every
incidence of AKI in critically ill patients due to COVID-19 3 day except for the last interval, which includes all data beyond
infection. Here we show evidence that mean platelet volume 16 days. We compared the presence and absence of AKI and
(MPV) reflects platelet activation and activation of coagulation tested the significance of each interval. A similar analysis was
cascades and plays a major role in the development of acute renal applied on the daily MPV data. Statistical analyses were per­
failure. formed using SPSS statistical software (Version 27, Chicago,
IL) and the programing language R.
Methods
Study Design and Data Collection Results
We included adult patients with laboratory-confirmed COVID-19 During early March through April 15 we reviewed the charts of
infection admitted to the intensive care units (ICU) at Harper 81 subjects who were positive for COVID-19 based on the PCR
University Hospital in Metro Detroit between March 15 to assay. Among 81 subjects, 41 (51%) were female. Mean age was
April 20, 2020. A confirmed case of COVID-19 was defined by 60 ± 13.8 y (range 24–95 y). Almost all subjects in our study had
a positive result on a reverse-transcriptase–polymerase-chain- hypertension (95%), over half (57%) had diabetes mellitus (DM)
reaction (RT-PCR) assay of a specimen collected on and many had a combination of both (55%). Patients demo­
a nasopharyngeal swab. Only laboratory-confirmed cases were graphics are shown in Table I. The mean body mass index
included. Eighty-one adults (18 years of age or older) were (BMI) was 33 ± 8.9 kg·m−2. Overall mortality in our study was
identified. Informed consent was waived, and researchers ana­ 65%. Among study subjects, 66 patients (81%) developed acute
lyzed only deidentified (anonymized) data. Data from each insti­ renal injury and 49% required renal replacement therapy. As
tution’s electronic medical record were obtained through shown in Table II, we followed a wide range of laboratory para­
a research form. We obtained demographic data, information on meters, including CRP, ferritin, CPK and LDH. We also evaluated
clinical symptoms or signs at presentation, laboratory, and radi­ D-dimer levels at two distinct time points during the ICU stay.
ologic results. Furthermore, we reviewed the peripheral blood D-dimer 1 (D1) is the first reported result after ICU admission
smears of patients during ICU admission. All laboratory tests and D-dimer 2 (D2) was drawn approximately 5 days later. In
were performed at the discretion of the treating physicians. This total, 45 patients had elevated D-dimer levels on the day of ICU-
study was reviewed and approved by the Detroit Medical Center admission. Therapeutic anticoagulation, either intravenous
and the Wayne State University Institutional Review Boards. unfractionated heparin (IV-UFH) or low molecular weight
heparin (Enoxaparin 1 mg/kg every 12 − 24 h depending on
creatinine clearance) was administered to 18 patients in this
Specimen Collection and Testing
study. All other ICU patients were on pharmacologic venous
Clinical specimens for COVID-19 diagnostic testing were thromboembolism prophylaxis with subcutaneous UFH.
obtained in accordance with Centers for Disease Control and There was a significant increase in D2 ranging from 0.25 to
Prevention (CDC) guidelines. Nasal swabs were performed in 35 mg/dl with a mean±SD of 6.1453 ± 0.98 compared to D1 that
all patients using a real-time RT-PCR assay developed by the was ranging from 0.9 to 35 mg/dl with a mean±SD of
CDC at the Michigan State Public Health Laboratory to detect the 13.6 ± 1.38. Platelet counts varied among patients, ranging
virus. from 52,000 to 482,000/µL, however, most patients had normal
to high platelet levels. Only one patient had baseline thrombocy­
Laboratory Data topenia with a platelet count of 52,000 µL, likely from his

All laboratory data were part of routine clinical data obtained


during hospitalization and ICU admission. The following data Table I. Clinical characteristics patients.
were analyzed: total white blood cell counts (WBC), absolute
lymphocytes counts (ALC), granulocyte count, platelets, Characteristic All Patients (N = 81)
Age (y, Mean ± SD) 60 ± 13.8
D-dimer, fibrinogen, prothrombin time (PT), partial prothrombin BMI (Mean ± SD) 32 ± 11.9
time (PTT), international normalized ratio (INR). C reactive pro­ Gender, N (%)
tein (CRP), alanine aminotransferase (ALT), aspartate transami­ Female 41 (50.9)
nase (AST), lactate dehydrogenase (LDH); creatine kinase (CPK) Male 40 (49.1)
and mean platelet volume (MPV). Glomerular filtration rates Race, N (%)
(GFR) were calculated based on creatinine clearance, gender African American 63(78%)
Caucasian 3 (3.7%)
and race.
Hispanic 1(1.7%)
Unknown 14 (17.3%)
Statistical Analysis Smoking history, N (%) 14(6%)
Coexisting disorder, N (%)
Categorical data are presented as frequency and percentage. Hypertension 75 (92%)
Continuous data are presented either as box plots (WBC, LDH, Diabetes 46 (57%)
CRP, D-D1, D-D2 and ferritin) or line plots with error bars Chronic kidney disease 19 (23%)
between two groups. Independent unpaired t-tests were used for Cardiovascular diseases 19 (23%)
Respiratory diseases 13 (16%)
analyzes of continuous variables and the bivariate Spearman’s Cancer 2 (2%)
rank correlation coefficient was calculated to measure the
DOI: https://doi.org/10.1080/09537104.2020.1817361 3
Table II. Initial laboratory findings. creatinine values in AKI group were 2.3 ± 1.9 (mg/dL) versus
1.38 ± 1.1 (mg/dL) in the group without AKI. Figure 1 shows the
Variable Mean ± SD No. Reference GFR trends during 8 days of ICU stay between two groups:
ranges absence of AKI (Figure 1A) and presence of AKI (Figure 1B).
White blood cell count, K/CUMM 8.3 ± 4.5 81 3.5–10.6
Platelet count, K/CUMM 229 ± 90 81 150–450 A Student t-test was performed to detect differences in outcome
C-reactive protein, mg/L 189.8 ± 121.4 81 0.0–5.0 variables of selected laboratory values between the group with
Ferritin, ng/mL 1036 ± 1189 79 24–336 and without AKI. Figure 1 shows Box plots overlaid with dot
Alanine aminotransferase, U/L 38 ± 35 72 7–52 plots illustrating mean±SD of the relevant laboratory data com­
Aspartate aminotransferase, U/L 58 ± 53 73 13–39 paring subjects, who developed AKI versus, who did not. As seen
Alkaline phosphatase, U/L 69 ± 29 61 45–115 in Figure 2, we found no significant differences in mean±SD of
Lactate dehydrogenase, U/L 569 ± 313 56 140–271
WBC (Figure 2A) between two groups. Similarly, there were no
Creatine phosphokinase, U/L 565 ± 1404 61 30–223
D-Dimer 1, mg/L* 6.15 ± 8.27 70 0.0–0.50 significant differences in the neutrophil and lymphocytes counts
D-Dimer 2 mg/L ** 13.5 ± 11.2 65 0.0–0.50 between two groups (data not shown). In contrast, we observed
Partial thromboplastin 31.7 ± 7.9 58 23.1–33.1 significant differences (p < .05) in CRP values (Figure 2B),
time, second(s) ferritin (Figure 2C), and LDH (Figure 2D), between these two
Prothrombin time, second(s) 12.2 ± 3.4 59 9.4–11.7 groups. Among patients who required renal replacement therapy,
Fibrinogen, mg/dL 482 ± 236 42 186–466 we observed a high number of subjects (65%), who exhibited
Mean platelet volume, FL 11.4 ± 1.6 81 7.3–11.4
*D-Dimer 1: At day 1 admission
circuit clotting and clotting in central lines, requiring heparin or
**D-Dimer 2: At day 5 admission alteplase treatment. Most striking, we found that patients, who
developed AKI had significantly higher D-dimer levels upon ICU
Abbreviation: WBC: White blood cell count; CRP: C reactive protein; admission (D1) (Figure 2E). Interestingly, the differences in
ALT: Alanine aminotransferase; AST: Aspartate transaminase; LDH: D-dimer levels were more pronounced, when we used the
lactate dehydrogenase; CPK: Creatine kinase; DD1: D-Dimer 1; DD2 D-dimer 2 (D2), which was measured on average at 5th
D-Dimer 2. PTT: Partial Prothrombin time; PT: Prothrombin time.
MPV: Mean platelet volume.
ICU day. Figure 1F shows the mean±SD of D2 between the two
groups. Given the important role of platelets and prothrombin
time in the activation of the coagulation pathway, we analyzed the
underlying cancer and chemotherapy. Similarly, we observed data to identify parameters that correlate with D1and D2. Next,
a wide variation in other laboratory values, including CRP, we performed non-parametric Spearman Correlation Test to iden­
LDH, and ferritin as shown in Table II. Interestingly, we observed tify laboratory parameters correlating with D-dimer levels. We
elevated fibrinogen levels among patients with COVID-19 as used all laboratory data, including PT, PTT, fibrinogen, CRP, Hb,
shown in Table II. ferritin, platelet count, and mean platelet volume (MPV). We
Acute renal injury (AKI) has been associated with worse out­ found a significant correlation between D1 and CRP (r = 0.32;
comes in patients with COVID-19, therefore we analyzed labora­ p < .05), as well as D2 and CRP (r = 0.558; p < .01). Strikingly,
tory data based on the presence or absence of AKI. There were we found a significant correlation of D1 and MVP (0.454;
significant differences (p value< .05:CI<0.095) in renal function p < .01) but we found no correlation between the number of
between subjects with and without AKI. In the AKI group the platelets and D1. When we used D2, we found a significant
mean±SD of blood urea nitrogen (BUN) was 39 ± 24 (mg/dL) correlation of D2 with ferritin (r = 0.303; p < .05), CRP
versus 17 ± 11 (mg/dL) in the group without AKI. The mean±SD (r = 0.475; p < .01), and MPV (r = 0.420; p < .01). Again, we

Figure 1. GFR trend among COVID-19 subjects from day one to day eight of ICU- stay. (A) Mean± SD of GFR (y-axis) in COVID-19 patients without
AKI from day one of admission plotted against time, showing the GFR trend. (B) The trend of GFR among COVID-19 patients with AKI from day1-8
of ICU-stay. There was a significant difference between AKI and non- AKI group in terms of GFR. Trend of GFR during ICU admission among
patients with COVID-19 infection for each day. Error bars plots to show the trend of GFR (y-axis) in COVID-19 patients without acute renal injury (A)
and with acute renal injury (B) during ICU stay. Each error bars plot shows the data from each day. The trend line connects the means of each day, and
the caps represent the SD. The GFR plot shows a continuous deterioration for patients with AKI starting from day 3, in contrast the GFR of patients
without AKI shows continuous improvement.
4 Platelets, Early Online: 1–8

Figure 2. Box blots of main laboratory values among COVID-19 subjects with and without renal failure. Mean± SEM levels of altered laboratory
values among COVID −19 patients who developed acute renal failure as compared to subjects who did not. (A) total WBC count (C/dL). (B)
C reactive Protein (CRP) mg/L. (C) Ferritin (ng/mL). (D) Lactate dehydrogenase (LDH) (U/L). E) D-Dimer 1 (mg/L) at ICU admission. (F) D-Dimer 2
(mg/L). The bottom and top of the boxes represent the 25th and 75th percentile, respectively. The top and bottom bars represent the entire stretch of the
data points for the subjects, except the extreme points, which are indicated with circles (o). The hyphen indicates the median value. The y-axis
represents the concentrations of the laboratory values.

found no significant correlation between D2 and PT, PTT, fibri­ expected, there was a significant difference between GFRs of
nogen, or platelet counts. In order to further determine the kinetic subjects with AKI versus no AKI, and deterioration in most
of GFR variation in a time-dependent manner during ICU admis­ patients occurred within 4–7 days of ICU stay. Similarly, most
sion, we collected the daily GFR values of all study subjects and patients with AKI died despite renal replacement therapy. Only
compare the kinetics of GFR based on the presence and absence six patients, who showed improvement of GFR after 16 days,
of AKI. Figure 3A shows violin plots for GFR in a time- survived. We performed similar analysis using MPV. As shown in
dependent manner based on presence and absence of AKI. As Figure 3B, COVID-19 subjects with AKI exhibited significantly

Figure 3. Trend of GFR and MPV during ICU stay among patients with COVID-19 infection. (A) The GFR trend presented as line plot with error bars
to show GFR (y-axis) in COVID-19 patients with and without acute renal injury in time-dependent manner (X-axis) during every 3 days of ICU stay.
Patients with AKI (red) versus without AKI (black). B) MPV trend of patients with AKI (red) without AKI (black). Days of admission (x-axis). Each
error bars plot shows the data of every 3 days. The dot represents the mean, and the cap represents the SD. The FDR p-values of each comparison are
shown at the bottom of the plots. The GFR plot shows a deterioration after day 3 for patients with AKI. After 16 days, this trend showed upwards as
some of the patients were either discharged or expired. The MPV plot shows a consistently higher level in AKI patients when compared to patients
without AKI. After 16 days, this trend of MPV showed downwards. At each time point for GFR and MPV, the comparisons of AKI patients and
without AKI were significant as shown of p-value on the bottom of each interval.
DOI: https://doi.org/10.1080/09537104.2020.1817361 5

larger platelets based on MPV. It is important to mention that all required renal replacement therapy. Furthermore, the mortality
subjects with COVID-19 have higher MPV values (see normal among the critically ill cohort of COVID-19 patients, who devel­
value), however, in the AKI group the MPV was significantly oped AKI was significantly higher. Previously, a large study
higher. MVP values in our study patients did fluctuate day by day reported the AKI incidence of 17% in critically ill patients
but on average, a higher MVP correlated with the presence admitted to ICU due to H1N1influenza infection [18]. The inci­
of AKI. dence of AKI in bacterial sepsis and ARDS has been reported
Because automated platelet counts may be unprecise, espe­ between 23% and 41% during ICU stay [19,20]. Our data indicate
cially with varying platelets size, we reviewed patients’ peripheral that the prevalence of AKI in COVID-19 infected individuals was
blood smears, which consistently showed findings of large plate­ substantially higher as compared to AKI incidence due to H1N1
lets. Figure 4A-F shows representative blood smears of six dif­ or bacterial sepsis. In COVID-19 infection multiple mechanisms
ferent subjects. As shown in Figure 4, most patients had enlarged may lead to microcirculatory alterations such as endothelial
platelets and several patients had giant platelets (Figure 3C). injury, autonomic nervous system response, activation of the
Furthermore, we analyzed the data to identify correlation between coagulation cascade and RAAS pathway.
laboratory data and outcome of AKI. We found that elevation of Soon after the onset of the COVID-19 pandemic, reports of
D-dimer at ICU admission or during ICU stay highly correlated increased activation of the coagulation pathway and thrombotic
with the development of AKI with a significantly lower false events emerged [21,22]. Patients may present with acute pul­
discovery rate (FDR) of 2.11E-09 for D-dimer 2 as compared monary embolism, deep venous thrombosis, acute ischemic
with FDR = 0.000 for the correlation of AKI with D-dimer events, or exhibit clotting of arterial and venous lines. Recent
values. This suggests rising D-Dimer levels during ICU stay autopsy results confirmed the presence of thrombosis in many
may be viewed as a poor prognostic marker. organs including lungs [23]. Our data indicate that a COVID-
19 infection leads to unique patterns of hypercoagulability and
platelet activation play a major role in initiation of coagulation
Discussion cascade. These patterns seem to be distinct to the usual DIC
Our study showed that 81% of critically ill COVID-19 patients pattern reported in other forms of dysfunctional coagulation
developed AKI during their ICU course and half of these patients pathways in severe gram-negative sepsis and Acute Respiratory

Figure 4. Peripheral blood smears of subjects with COVID-19 infection. Peripheral blood smear showing (A) platelets with variation in granulation,
shape, and size including large platelets (thin arrow), Wright-Giemsa stain, Objective 100x, immersion oil; (B) large platelets clumps (thin arrow),
Wright-Giemsa stain, Objective 100x, immersion oil (C) giant platelet that is larger than a normal red blood cell and showing a pseudo-nucleus (thin
arrow), Wright-Giemsa stain, Objective 100x, immersion oil; (D) platelets with variation in granulation, shape, and size, Wright-Giemsa stain,
Objective 100x, immersion oil; (E) several large platelets (thin arrow) with variation in shape and size partially adherent to a myelocytes (thick arrow)
(platelet satellitism), Wright-Giemsa stain, Objective 100x, immersion oil; (F) reactive plasmacytoid lymphocytes (thick arrow), Wright-Giemsa stain,
Objective 100x, immersion oil.
6 Platelets, Early Online: 1–8

Distress Syndrome (ARDS) [24,25]. One criterion for DIC is a limited efficacy of conventional anticoagulant therapy in
decreased fibrinogen levels and presence of fibrin monomers COVID-19 associated coagulopathy, warranting to evaluate
[26]. In the contrary to DIC, we observed increased fibrinogen the efficacy of antiplatelet drug therapy in this disease.
level, elevated D-dimers, but normal platelet counts in these SARS‐CoV‐2 utilizes the novel ACE 2 receptor to gain
subjects. Our data corroborate with previous reports that entry into human cells [9]. This process leads to shedding of
patients with COVID-19 have elevated fibrinogen levels. In host ACE2 receptor and the loss of ACE2 function [22].
a limited number of subjects, the level of protein C and protein Because ACE2 counterbalances the deleterious effect of the
S was decreased, which is consistent with consumption coagu­ RAAS, through effect of Angiotensin [1–7,36,37], the loss of
lopathy. Strikingly, we observed a unique feature of platelets in ACE2 function amplifies the deleterious effect of angiotensin II
these patients, including larger, granular platelets with a higher (Ang II). It has been shown that Ang II stimulation leads to
MPV and increased frequency of large platelets clumps in the increased platelet size and activation, and that aspirin does not
presence of normal platelet counts. This has been defined as prevent such activation [16,38]. Other experimental models
a sign of platelet activation and indicator of increased throm­ suggested that heparin has minimal effect on the Ang II
bopoiesis [27]. Interestingly, we found increased immature induced platelet activation [12,38]. In contrast, ACE2 activity
platelet fraction IPF (IPF>7) in COVID-19-infected subjects, or activation of the MAS receptor promotes antithrombotic
who had evidence of elevated D-dimers and AKI. Immature activity and analogues of Angiotensin [1–7] prevent thrombosis
platelets usually have larger size and are considered to be more formation [13,36,39]. Alteration of ACE2 function and
reactive and increased IPF has been associated with smoking- increased Ang II have been associated with chronic kidney
related cardiovascular disease and inflammation [28,29]. Day disease and diabetes nephropathy [40]. Furthermore, SARS‐
to day variation in MVP can be related to various factors, CoV‐2 infection causes endothelial injury, which amplifies
including misinterpretation of the true platelet size due to platelet activation. Limited available renal autopsy results of
platelets clumps, and some more activated platelets will be COVID-19 deceased subjects showed presence of micro­
consumed in clot formation and not counted by automatic thrombi and fibrin thrombi in glomerular capillaries [41].
reader. It has been shown that larger platelets have denser α- Several studies have shown that hypertension, diabetes and
granules, which store biologically active molecules such as obesity are associated with dysregulation of Ang II and
GPIIb/IIIa, fibrinogen, and von Willebrand factor (vWf) ACE2 [42]. In both diseases increased platelet size and activa­
which can initiate the coagulation cascade [30]. Dense granules tion have been reported [16,43,44]. It is important to note that
store a variety of molecules which are secreted during platelet 75% of our patients had hypertension and 58% had diabetes
activation; including catecholamines, serotonin, calcium, ade­ mellitus. The SARS‐CoV‐2 infection and the loss of function
nosine 5′-diphosphate (ADP), and adenosine 5′-triphosphate of ACE2 may potentiate the existing prothrombotic state of
(ATP) that involved in initiating coagulation cascade during subjects with diabetes and hypertension. The unique features
inflammation [31]. Higher MPV is also seen with DIC and it of COVID-19 infection are therefore, the loss of ACE2 func­
has been shown that MPV plays an important role in activation tion and increased Ang II activity promote severe endothelial
of coagulation and/or hyperfibrinolysis. If hyperfibrinolysis is dysfunction, platelet activation, an increased burden of micro­
the predominant feature, it is associated with remarkable bleed­ vascular thrombosis, and subsequent microcirculatory compro­
ing rather than increased vascular thrombosis. Reductions in mise in many organs including lungs, heart, CNS, and kidney
the levels of natural anticoagulants, such as antithrombin III associated with fatal outcome.
and protein C are common in patients with DIC.
Hyperfibrinolysis with subsequent increased bleeding is asso­
ciated with an increase in the levels of cytokines which further Acknowledgements
induces plasminogen activator inhibitor I (PAI-I). In viral This work was supported by a grant (R01HL150474 (LS) as well as the
hemorrhagic fevers, including in Ebola and Hantavirus pulmon­ Department of Medicine, Wayne State University School of Medicine
ary syndrome, aberrant coagulation and bleeding have been (LS).
reported [20,21]. The infection with Ebola virus is associated
with elevated PAI-I levels with increased bleeding [32], Funding
Elevated PIA-I is associated with high mortality in these
This work was supported by the National Heart, Lung, and Blood
patients [33]. Other predominant features of Ebola virus infec­ Institute [R01HL150474].
tion are thrombocytopenia, coagulation factor deficiencies and
elevated D-dimers. In contrast to viral hemorrhagic fevers,
COVID-19 has not been associated with hemorrhage. Soon Author Contributions
after the reports of activation of coagulation and increased
MT and ME collected clinical data and contributed in writing the
incidence of pulmonary embolisms and DVT emerged, many manuscript. DS and AG reviewed the peripheral blood smear and
centers adopted guidelines to initiate therapy with unfractio­ contributed in writing the manuscript. SH and SD performed the
nated or low molecular weight heparin in patients with statistical analysis of data. TC contributed to data interpretation and
COVID-19 infection. However, our observation and others writing the manuscript. LS conceived and designed the study, partici­
suggest that some of these patients continue to be in a pro- pated in all areas of the research such as patients’ selection, data
coagulable state as indicated by clinical observations of analysis and writing of the manuscript.
ongoing thrombotic disease and laboratory data, for example
increased in D-dimers, despite therapeutic anticoagulation [34].
Competing Financial Interests
Experimental data in animals suggested that neither heparin nor
hirudin can overcome the thrombogenic effects of Angiotensin None of the authors of this manuscript had any financial relationship with
II (Ang II). Interestingly antithrombin III could reverse the Ang a commercial company.
II associated prothrombotic state in experimental animal model
[12]. It was reported by Llitjos et al. that 56% of patients
ORCID
infected with COVID-19, who were receiving full dose antic­
oagulation developed VTE [35]. These observations suggest Lobelia Samavati http://orcid.org/0000-0002-3327-2585
DOI: https://doi.org/10.1080/09537104.2020.1817361 7

References 19. Bagshaw SM, George C, Bellomo R, Committee ADM. Early acute
kidney injury and sepsis: a multicentre evaluation. Critical Care
1. Baud D, Qi X, Nielsen-Saines K, Musso D, Pomar L, Favre G. Real 2008;12:R47. doi: 10.1186/cc6863
estimates of mortality following COVID-19 infection. Lancet Infect 20. Rangel-Frausto MS, Pittet D, Costigan M, Hwang T, Davis CS,
Dis 2020 Jul;20(7):773. doi: 10.1016/S1473-3099(20)30195-X. Wenzel RP. The natural history of the systemic inflammatory
2. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, Zhang L, Fan G, response syndrome (SIRS): a prospective study. JAMA 1995;273
Xu J, Gu X. Clinical features of patients infected with 2019 novel (2):117–123. doi: 10.1001/jama.1995.03520260039030
coronavirus in Wuhan, China. The Lancet 2020;395 21. Di Micco P, Russo V, Carannante N, Imparato M, Rodolfi S,
(10223):497–506. doi: 10.1016/S0140-6736(20)30183-5 Cardillo G, Lodigiani C. Clotting Factors in COVID-19: epidemio­
3. Xiong M, Liang X, Wei YD. Changes in blood coagulation in logical association and prognostic values in different clinical pre­
patients with severe coronavirus disease 2019 (COVID-19): a sentations in an Italian cohort. J Clin Med. 2020 May;9(5):1371.
meta-analysis. Br J Haematol 2020;189(6):1050–1052. doi: doi: 10.3390/jcm9051371.
10.1111/bjh.16725 22. Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M,
4. Zhang Y, Xiao M, Zhang S, Xia P, Cao W, Jiang W, Chen H, Delabranche X, Merdji H, Clere-Jehl R, Schenck M, Fagot
Ding X, Zhao H, Zhang H. Coagulopathy and antiphospholipid Gandet F, et al. High risk of thrombosis in patients with severe
antibodies in patients with Covid-19. N Engl J Med 2020;382(17): SARS-CoV-2 infection: a multicenter prospective cohort study.
e38. doi: 10.1056/NEJMc2007575 Intensive Care Med 2020. doi: 10.1007/s00134-020-06062-x
5. Lodigiani C, Iapichino G, Carenzo L, Cecconi M, Ferrazzi P, 23. Wichmann D, Sperhake JP, Lutgehetmann M, Steurer S, Edler C,
Sebastian T, Kucher N, Studt J-D, Sacco C, Alexia B. Venous Heinemann A, Heinrich F, Mushumba H, Kniep I, Schroder AS,
and arterial thromboembolic complications in COVID-19 et al. Autopsy findings and venous thromboembolism in patients
patients admitted to an academic hospital in Milan, Italy. with COVID-19: A prospective cohort study. Ann Intern Med 2020.
Thromb Res 2020;191:9–14. doi: 10.1016/j. doi: 10.7326/M20-2003
thromres.2020.04.024 24. Simmons J, Pittet J-F. The coagulopathy of acute sepsis. Curr Opin
6. Fehr AR, Perlman S. Coronaviruses: an overview of their replication Anaesthesiol 2015;28(2):227. doi: 10.1097/
and pathogenesis. In: Maier H., Bickerton E., Britton P. (eds) ACO.0000000000000163
Coronaviruses. Methods in Molecular Biology, vol 1282. New 25. LaRosa SP, Opal SM, Utterback B, Yan SCB, Helterbrand J,
York, NY: Humana Press. doi: 10.1007/978-1-4939-2438-7_1 Simpson AJ, Chaowagul W, White NJ, Fisher CJ Jr. Decreased
7. Li F, Li W, Farzan M, Harrison SC. Structure of SARS coronavirus protein C, protein S, and antithrombin levels are predictive of poor
spike receptor-binding domain complexed with receptor. Science outcome in Gram-negative sepsis caused by Burkholderia
2005;309(5742):1864–1868. doi: 10.1126/science.1116480 pseudomallei. Int J Infect. Dis 2006;10:25–31. doi: 10.1016/j.
8. Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, ijid.2005.06.001
Erichsen S, Schiergens TS, Herrler G, Wu N-H, Nitsche A. SARS- 26. Venugopal A. Disseminated intravascular coagulation. Indian
CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked J Anaesth 2014;58(5):603. doi: 10.4103/0019-5049.144666
by a clinically proven protease inhibitor. Cell 2020;181(2):271–280. 27. Park Y, Schoene N, Harris W. Mean platelet volume as an indicator
e8. doi: 10.1016/j.cell.2020.02.052 of platelet activation: methodological issues. Platelets 2002;13(5–­
9. Li W, Moore MJ, Vasilieva N, Sui J, Wong SK, Berne MA, 6):301–306. doi: 10.1080/095371002220148332
Somasundaran M, Sullivan JL, Luzuriaga K, Greenough TC. 28. Schmoeller D, Picarelli MM, Paz Munhoz T, Poli de Figueiredo CE,
Angiotensin-converting enzyme 2 is a functional receptor for the Staub HL. Mean platelet volume and immature platelet fraction in
SARS coronavirus. Nature 2003;426(6965):450–454. doi: 10.1038/ autoimmune disorders. Front Med 2017;4:146. doi: 10.3389/
nature02145 fmed.2017.00146
10. Hamming I, Timens W, Bulthuis M, Lely A, Navis G, van Goor H. 29. Nardin M, Verdoia M, Negro F, Rolla R, Tonon F, De Luca G.
Tissue distribution of ACE2 protein, the functional receptor for SARS Impact of active smoking on the immature platelet fraction and its
coronavirus. A first step in understanding SARS pathogenesis. J Pathol relationship with the extent of coronary artery disease. Eur J Clin
2004;203(2):631–637. doi: 10.1002/path.1570 Invest 2020;50(2):e13181. doi: 10.1111/eci.13181
11. Tikellis C, Thomas M. Angiotensin-converting enzyme 2 (ACE2) is 30. Blair P, Flaumenhaft R. Platelet α-granules: basic biology and
a key modulator of the renin angiotensin system in health and clinical correlates. Blood Rev 2009;23(4):177–189. doi: 10.1016/j.
disease. Int J Pept 2012;2012:256294. doi: 10.1155/2012/256294. blre.2009.04.001
12. Senchenkova EY, Russell J, Esmon CT, Granger DN. Roles of 31. Chen Y, Yuan Y, Li W. Sorting machineries: how platelet-dense
coagulation and fibrinolysis in angiotensin II‐enhanced microvascu­ granules differ from α-granules. Biosci Rep. 2018 Oct 31; 38(5):
lar thrombosis. Microcirc 2014;21:401–407. doi: 10.1111/ BSR20180458. Published online 2018 Sep 7. doi: 10.1042/
micc.12120 BSR20180458.
13. Fraga-Silva RA, Sorg BS, Wankhede M, deDeugd C, Jun JY, 32. West TE, von Saint André-von AA. Clinical presentation and man­
Baker MB, Li Y, Castellano RK, Katovich MJ, Raizada MK. agement of severe Ebola virus disease. Ann Am Thorac Soc
ACE2 activation promotes antithrombotic activity. Mol Med 2014;11(9):1341–1350. doi: 10.1513/AnnalsATS.201410-481PS
2010;16(5–6):210–215. doi: 10.2119/molmed.2009.00160 33. McElroy AK, Erickson BR, Flietstra TD, Rollin PE, Nichol ST,
14. Liu N, Hong Y, Chen R-G, Zhu H-M. High rate of increased level of Towner JS, Spiropoulou CF. Biomarker correlates of survival in
plasma Angiotensin II and its gender difference in COVID-19: an pediatric patients with Ebola virus disease. Emerg Infect Dis
analysis of 55 hospitalized patients with COVID-19 in a single hospi­ 2014;20(10):1683. doi: 10.3201/eid2010.140430
tal, Wuhan, China. medRxiv 2020. doi: 10.1101/ 34. Poissy J, Goutay J, Caplan M, Parmentier E, Duburcq T, Lassalle F,
2020.04.27.20080432. Jeanpierre E, Rauch A, Labreuche J, Susen S. Pulmonary embolism
15. Siragy HM, Carey RM. Role of the intrarenal renin-angiotensin- in COVID-19 patients: awareness of an increased prevalence.
aldosterone system in chronic kidney disease. Am J Nephrol Circulation 2020 Jul 14;142(2):184–186. doi: 10.1161/
2010;31(6):541–550. doi: 10.1159/000313363 CIRCULATIONAHA.120.047430.
16. Jagroop I, Mikhailidis D. Angiotensin II can induce and potentiate 35. Llitjos JF, Leclerc M, Chochois C, Monsallier JM, Ramakers M,
shape change in human platelets: effect of losartan. J Hum Auvray M, Merouani K. High incidence of venous thromboembolic
Hypertens 2000;14(9):581–585. doi: 10.1038/sj.jhh.1001102 events in anticoagulated severe COVID‐19 patients. J Thromb
17. Waikar SS, Curhan GC, Wald R, McCarthy EP, Chertow GM. Haemost. 2020 Jul;18(7):1743–1746. doi: 10.1111/jth.14869.
Declining mortality in patients with acute renal failure, 1988 to 36. Rabelo LA, Alenina N, Bader M. ACE2–angiotensin-(1–7)–Mas
2002. J Am Soc Nephrol 2006;17(4):1143–1150. doi: 10.1681/ axis and oxidative stress in cardiovascular disease. Hypertens Res
ASN.2005091017 2011;34(2):154–160. doi: 10.1038/hr.2010.235
18. Martin-Loeches I, Papiol E, Rodríguez A, Diaz E, Zaragoza R, 37. Samavati L, Uhal BD. ACE2, much more than just a receptor for
Granada RM, Socias L, Bonastre J, Valverdú M, Pozo JC. Acute SARS-COV-2. Front. Cell. Infect. Microbiol., 05 June 2020.
kidney injury in critical ill patients affected by influenza A (H1N1) doi:10.3389/fcimb.2020.00317.
virus infection. Critical Care 2011;15(1):R66. doi: 10.1186/cc10046 38. Larsson P, Schwieler J, Wallen N. Platelet activation during angio­
tensin II infusion in healthy volunteers. Blood Coagulation
8 Platelets, Early Online: 1–8

Fibrinolysis 2000;11(1):61–69. doi: 10.1097/00001721-200011010- 42. Ingelfinger JR. ACE2: a new target for prevention of diabetic
00007 nephropathy? Am Soc Nephrol 2006;17(11):2957–2959. doi:
39. Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, Liu FF, 10.1681/ASN.2006090986
Zhang K, Zhang C. Angiotensin-converting enzyme 2 and angio­ 43. Shah B, Sha D, Xie D, Mohler ER, Berger JS. The relationship
tensin 1–7: novel therapeutic targets. Nat Rev Cardiol 2014;11:413 between diabetes, metabolic syndrome, and platelet activity as mea­
40. Williams VR, Scholey JW. Angiotensin-converting enzyme 2 and sured by mean platelet volume: the National Health And Nutrition
renal disease. Curr Opin Nephrol Hypertens 2018;27(1):35–41. doi: Examination Survey, 1999–2004. Diabetes Care
10.1097/MNH.0000000000000378 2012;35:1074–1078. doi: 10.2337/dc11-1724
41. Menter T, Haslbauer J, Nienhold R, Savic S, Hopfer H, 44. Ueba T, Nomura S,Inami N, Nishikawa T, Kajiwara M, Iw
ata R,Yamashit­
Deigendesch N, Frank S, Turek D, Willi N, Pargger H. Post‐mortem Ueba T, Nomura S,Inami N, Nishikawa T, Kajiwara M, Iw
ata R,Yamashit­
examination of COVID19 patients reveals diffuse alveolar damage a K. Plasma level of platelet-derived microparticles is associated
with severe capillary congestion and variegated findings of lungs with coronary heart disease risk score in healthy men, 2010;17
and other organs suggesting vascular dysfunction. The (4):342–349. doi: 10.5551/jat.2964.
Histopathology of COVID‐19, August 2020;77(2):198–209.
Joseph et al. Ann. Intensive Care (2020) 10:117
https://doi.org/10.1186/s13613-020-00734-z

RESEARCH Open Access

Acute kidney injury in patients


with SARS‑CoV‑2 infection
Adrien Joseph1, Lara Zafrani1,3*, Asma Mabrouki1, Elie Azoulay1,2 and Michael Darmon1,2

Abstract 
Background:  Acute Kidney Injury (AKI) is a frequent complication of severe SARS-CoV-2 infection. Multiple mecha-
nisms are involved in COVID-19-associated AKI, from direct viral infection and secondary inflammation to comple-
ment activation and microthrombosis. However, data are limited in critically-ill patients. In this study, we sought to
describe the prevalence, risk factors and prognostic impact of AKI in this setting.
Methods:  Retrospective monocenter study including adult patients with laboratory confirmed SARS-CoV-2 infec-
tion admitted to the ICU of our university Hospital. AKI was defined according to both urinary output and creatinine
KDIGO criteria.
Results:  Overall, 100 COVID-19 patients were admitted. AKI occurred in 81 patients (81%), including 44, 10 and 27
patients with AKI stage 1, 2 and 3 respectively. The severity of AKI was associated with mortality at day 28 (p = 0.013).
Before adjustment, the third fraction of complement (C3), interleukin-6 (IL-6) and ferritin levels were higher in AKI
patients. After adjustment for confounders, both severity (modified SOFA score per point) and AKI were associated
with outcome. When forced in the final model, C3 (OR per log 0.25; 95% CI 0.01–4.66), IL-6 (OR per log 0.83; 95% CI
0.51–1.34), or ferritin (OR per log 1.63; 95% CI 0.84–3.32) were not associated with AKI and did not change the model.
Conclusion:  In conclusion, we did not find any association between complement activation or inflammatory mark-
ers and AKI. Proportion of patients with AKI during severe SARS-CoV-2 infection is higher than previously reported and
associated with outcome.
Keywords:  Acute kidney injury, COVID-19, Complement system proteins, Interleukin-6, Outcome, Intensive care units

Background the critically ill (23%; 14–35%) [2–4]. Different applica-


Since December 2019, severe acute respiratory coronavi- tions of the Kidney Disease Improving Global Outcomes
rus 2 (SARS-CoV-2) has spread worldwide, causing more (KDIGO) criteria for AKI, in particular different methods
than 6.6 million cases and 390 000 deaths [1]. This pan- to estimate missing baseline creatinine and handling uri-
demic has put unprecedented pressure on healthcare sys- nary output, can cause important variations of estimated
tems and especially on intensive care units (ICUs). incidence [5, 6] and may contribute to the discrepancies
Acute Kidney Injury (AKI) is a frequent complication among these studies.
of severe SARS-CoV-2 infection but data are scarce in Multiple mechanisms are involved in COVID-19-as-
ICUs. AKI has been previously reported with an average sociated AKI, ranging from direct viral infection of
incidence of 11% (8–17%) overall, with highest ranges in the kidney and secondary inflammation to comple-
ment activation and microthrombosis [7]. In particular,
severe COVID-19 is associated with uncontrolled sys-
*Correspondence: lara.zafrani@aphp.fr temic inflammatory response with high levels of IL-6
1
Service de médecine Intensive et de réanimation médicale, Hôpital [8] that could potentially lead to intrarenal inflamma-
Saint-Louis, Assistance-Publique Hôpitaux de Paris, Paris University, 1
avenue Claude Vellefaux, 75010 Paris, France
tion and increased vascular permeability and share sev-
Full list of author information is available at the end of the article eral features with hyperferritinemic syndromes such as

© The Author(s) 2020. This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and
the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material
in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material
is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the
permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creat​iveco​
mmons​.org/licen​ses/by/4.0/.
Joseph et al. Ann. Intensive Care (2020) 10:117 Page 2 of 8

macrophage activation syndrome [9]. Furthermore, unre- from baseline or urinary output < 0.5 ml/kg/h for 6–12 h
strained activation of complement leads to endothelial within 7 days; stage 2—2.9 times increase in serum cre-
cell dysfunction and intravascular coagulation that could atinine or urinary output < 0.5 ml/kg/h for ≥ 12 h within
participate in COVID-19-associated AKI [10]. Both IL-6 7 days; stage 3—3 times or more increase in serum cre-
[11] and complement [12] have been proposed as thera- atinine or to ≥ 4.0  mg/dl or initiation of RRT or urinary
peutic targets and understanding their role in COVID- output < 0.3 ml/kg/h for ≥ 24 h or anuria for ≥ 12 h within
19-associated AKI is therefore a priority. 7  days. Patients were stratified according to the highest
However, most of the studies performed to date gave AKI stage attained during the first 7 days of ICU stay.
little data regarding definition of AKI or influence of Baseline creatinine was defined as the best value in the
inflammation and complement markers on AKI. 3 preceding months or if unavailable as the lowest value
In this study, we sought to describe the prevalence, risk during ICU stay or was back calculated based on a glo-
factors and prognostic impact of AKI during COVID-19 merular filtration rate of 60 mL/min/1.73m2 with MDRD
in the ICU. equation in patients without known chronic kidney dis-
ease. Chronic kidney disease (CKD) was defined accord-
Methods ing to the KDIGO definition.
Study design and cohort Modified SOFA was defined as SOFA score [14] exclud-
We conducted a retrospective monocenter study includ- ing the renal component.
ing adult patients with laboratory confirmed SARS-
CoV-2 infection admitted to the ICU of our university Statistical analysis
Hospital. All adult patients (age ≥ 18  years) who tested Continuous variables were described as median (inter-
positive by polymerase chain reaction testing of a naso- quartile range [IQR]) and compared between groups
pharyngeal sample for COVID-19 and were hospitalized using the non-parametric Wilcoxon rank-sum test. Cat-
from March 1, 2020 to June 1, 2020 were eligible. egorical variables were described as frequency (percent-
This study was approved by an institutional review ages) and compared between groups using Fisher’s exact
board (French Intensive Care Society—CE SRLF n°20– test. Mortality was assessed using survival analysis.
32). Need for informed consent was waived as regard to Independent risk factors of day 28 mortality were
the study observational design and in accordance with assessed using Cox model. Conditional stepwise vari-
the French law. This study was conducted in accordance able selection was performed with 0.2 as the critical
with the principles of the Declaration of Helsinki. p-value for entry into the model, and 0.1 as the p-value
for removal. Interactions and correlations between the
Data collection, definitions and measurements explanatory variables were carefully checked. Validity of
All data were obtained from medical records and proportional hazards assumption, influence of outliers,
patients’ charts. Baseline patients’ characteristics were and linearity in relationship between the log hazard and
collected, including demographics and comorbidities the covariates were carefully checked.
before ICU admission. The variables recorded regarding Independent risk factors of AKI were assessed using
ICU admission and treatments were relative to clinical logistic regression. Conditional backward stepwise vari-
presentation, reason for ICU admission, diagnosis, thera- able selection was performed with 0.2 as the critical
pies implemented and outcomes. p-value for entry into the model, and 0.1 as the p-value
Blood sampling and routine biological testing were per- for removal. Interactions and correlations between the
formed on the day of admission according to the stand- explanatory variables were carefully checked. Influence
ard laboratory protocols. of outliers, and linearity in relationship between the
All samples were immediately centrifuged at 3000 rpm log hazard and the covariates were carefully checked. It
at 4 °C for 10 min, separated from the cells and stored at was preplanned to force, one by one, in the final model
−80 °C until biochemical assays of complement proteins inflammation biomarker, ferritin, complement pathway
C3, C4 (nephelometry) and sC5B9 (ELISA), IL-6 (ELISA) dosage and PEEP level at admission should these variable
were performed. The primary outcome was mortality at not be selected.
day 28. Kaplan–Meier graphs were used to express the prob-
ability of death from inclusion to day 28. Comparisons
Definition of AKI were performed using the log-rank test.
AKI was defined according to both urinary output and Overall, rate of missing data was 6.6% with rate of miss-
serum creatinine KDIGO criteria [13] as follows: stage ing data among major outcome or covariates was < 5%. As
1—increase in serum creatinine by 0.3  mg/dl within regard to completeness of the dataset, no imputation of
48  h or a 1.5–1.9 times increase in serum creatinine missing data was performed.
Joseph et al. Ann. Intensive Care (2020) 10:117 Page 3 of 8

Statistical analyses were performed with R statistical associated with a lower risk for AKI stage 2 or 3, com-
software, version 3.6.2 (available online at https​://www.r- pared to no AKI or AKI stage 1 (OR 0.17 95% CI [0.05–
proje​ct.org/) and the ‘Survival’ package was used. A p 0.54], p = 0.004), while the association between AKI stage
value < 0.05 was considered significant. 2 or 3 and IL-6, Ferritin, sC5b9 and PEEP levels did not
reach statistical significance (Additional file  1: Table  S1
and Figure S1).
Results
Patients’ characteristics
Outcome analysis
Overall, 100 COVID-19 patients were admitted in our
Twenty-nine (29%) patients had died by day 28, 28
ICU between March and May 2020 and included in this
(35%) patients with AKI and 1 (5%) patient without AKI
study. Patients’ characteristics are described in Table  1.
(p = 0.02). More than half of the patients with AKI stage
Median age was 59  years [53–67] and 70 patients (70%)
2 (n = 5, 50%) and 3 (n = 15, 56%) died before day 28. The
were of male gender. Only 15 patients (15%) had no
severity of AKI was associated with mortality at day 28
underlying disease. Hypertension (n = 56, 56%), diabetes
(p = 0.013) (Fig. 2 and Additional file 1: Figure S2).
(n = 30, 30%) and chronic kidney disease (n = 29, 29%)
Patients who died were older (66 versus 57  years,
were the main comorbidities. Thirty-six patients were
p = 0.001), suffered more frequently from chronic kid-
obese (n = 21, 21%) or overweight (n = 25, 25%) and 30
ney disease (52 versus 20%, p = 0.003), had higher SOFA
(30%) were treated with angiotensin converting enzyme
score (8 versus 2, p < 0.001), ferritin levels (1805 versus
inhibitors or angiotensin-receptor blockers. Median
1148 mg/L, p = 0.004) and IL-6 levels (204 versus 103 ng/
SOFA score at admission was 4 [2–7]. Median modified
mL, p = 0.02), and were less often immunocompetent (46
SOFA score [without the renal component] was 3 [2–7].
versus 18%, p = 0.01). Neither comorbidities nor BMI
Fifty-six patients (55%) required mechanical ventilation
were associated with death at day 28. Patients who died
and 51 (51%) vasopressor therapy.
more often required mechanical ventilation (90 versus
41%, p < 0.001), vasopressors (90 versus 36%, p < 0.001)
Risk factors of AKI in COVID‑19 patients and renal replacement therapy (31 versus 6%, p = 0.002).
Rate of patients with missing baseline serum creatinine After adjustment for confounders, both AKI and sever-
was 67% (n = 67) and did not differ between AKI and ity (modified SOFA score per point) were associated with
patients without AKI (74 vs. 65% respectively; p = 0.67). survival (Table 2).
AKI occurred in 81 patients (81%), including 44 There was a similar trend towards poorer survival
patients, 10 patients, 27 patients with AKI stage 1, 2 and in patients with (n = 33) and without (n = 67) baseline
3 respectively. Among patients with AKI, 33 (41%) met serum creatinine (Additional file 1: Figure S3).
only urinary output KDIGO criteria, 28 (35%) met only We also tested the interaction between missing baseline
creatinine criteria and 20 (25%) met both. Urinary output serum creatinine and reported results and did not find
criteria alone was more frequently involved in diagno- any significant interaction (Additional file 1: Table S2).
sis of milder stage of AKI (AKI stage 1 (n = 20, 45%) and Treatments with lopinavir/ritonavir (n = 10), tocili-
stage 2 (n = 6, 60%) compared to 26% in stage 3 (n = 7, zumab (n = 1), eculizumab (n = 2) or chloroquine (n = 3)
p = 0.007). Thirteen (13%) required renal replacement were associated neither with mortality nor with the
therapy during the first 7 days in ICU. development of AKI.
Before adjustment, C3 (p = 0.01), IL-6 (p = 0.03) and
ferritin levels (p = 0.03) were associated with AKI sever- Discussion
ity, whereas soluble C5b9 fraction was not different In this study, we describe the incidence of AKI in 100
(p = 0.42) (Fig. 1). COVID-19 patients admitted to the ICU, and the link
In a multivariate model incorporating baseline chronic between AKI, inflammation markers and complement
kidney disease, only modified SOFA was significantly levels. The main results of this case series are the higher
associated with the development of AKI (OR per point than previously reported incidence of AKI and its lack
1.29; 95% CI 1.04–1.70). of association with IL-6, ferritin or complement fac-
When forced in the final model, C3 (OR per log 0.25; tors C3 and sC5B9. The importance of AKI in COVID-
95% CI 0.01–4.66), IL-6 (OR per log 0.83; 95% CI 0.51– 19 patients has been increasingly recognized. If initial
1.34), ferritin (OR per log 1.63; 95% CI 0.84–3.32), or reports from China reported rates of AKI as low as 2.9–
PEEP level (OR per mmHg 1.04; 95% CI 0.91–1.22) were 8% in severe patients [2, 15, 16], incidences from later
not associated with AKI and did not change the model. studies ranged between 15% [17] and 44% [18] in criti-
However, after adjustment for modified SOFA and cally ill patients. Most reports lack a clear operational
CKD, C3 value higher than median was significantly AKI definition [19–21], but even in studies that used
Joseph et al. Ann. Intensive Care (2020) 10:117 Page 4 of 8

Table 1  Patients’ characteristics according to AKI


Overall No AKI AKI p value
n = 100 n = 19 n = 81

Age (year) 59 [53–67] 54 [45–61] 60 [54–68] 0.05


Male gender 70 (70%) 11 (58%) 59 (73%) 0.32
Absence of underlying comorbidity 15 (15%) 6 (32%) 9 (11%) 0.06
Chronic Obstructive Pulmonary Disease 2 (2%) 0 (0) 2 (3%) 1.00
Asthma 8 (8%) 2 (11%) 6 (8%) 1.00
History of hypertension 56 (56%) 8 (42%) 48 (60%) 0.25
Diabetes 30 (30%) 3 (16%) 27 (34%) 0.21
Immunocompromized 26 (26%) 4 (21%) 22 (28%) 0.78
Heart failure 15 (15%) 0 (0) 15 (19%) 0.09
Chronic kidney disease 29 (29%) 2 (11%) 27 (33%) 0.09
Body Mass Index (Kg/m2) 28 [24–31] 26 [23–31] 28 [24–31] 0.48
Obesity and overweight 0.37
Overweight 25 (25%) 3 (16%) 22 (27%)
Obese 21 (21%) 3 (16%) 18 (22%)
Other 54 (54%) 13 (68%) 41 (51%)
Chronic use of ACE/ARB 30 (30%) 3 (16%) 27 (33%) 0.22
Baseline serum creatinine (µmol/L) 65 [50–93] 63 [48–70] 67 [50–100] 0.22
SOFA score 4 [2–7] 2 [2, 3] 5 [2–7] 0.003
Nonsteroidal anti-inflammatory drugs (%) 1 (1%) 0 (0) 1 (1%) 1.00
Invasive mechanical ventilation 55 (55%) 6 (32%) 49 (61%) 0.04
PEEP at day 1 0 [0–10] 0 [0–4] 8 [0–10] 0.04
Renal replacement therapy 13 (13%) 0 (0) 13 (16%) 0.14
Vasopressors 51 (51%) 7 (37%) 44 (55%) 0.24
IL6 at day 0 (ng/mL) 118 [67–287] 136 [63–292] 113 [69–287] 0.98
C3 at day 0 (ng/mL) 1305 [1173–1550] 1495 [1285, 1630] 1260 [1160–1543] 0.06
C4 at day 0 (ng/mL) 348 [275–418] 351 [282–493] 348 [272–416] 0.69
sC5b9 at day 0 (ng/mL) 373 [270–471] 425 [317–516] 363 [266–450] 0.22
Ferritin at day 0 (mg/L) 1272 [636–2234] 1182 [495–1584] 1311 [695–2322] 0.12
Fibrinogen at day 0 (g/L) 6.8 [5.8–7.8] 7.2 [5.4–7.7] 6.7 [5.8–7.8] 0.94
Acute kidney injury - 81 (81%) 0 (0) 81 (100%)  < 0.001
KDIGO stage  < 0.001
No AKI 19 (19%) 19 (100%) 0 (0)
Stage 1 44 (44%) 0 (0) 44 (54%)
Stage 2 10 (10%) 0 (0) 10 (12%)
Stage 3 27 (27%) 0 (0) 27 (33%)
KDIGO criteria fulfilled  < 0.001
Creatinine criteria alone 28 (28%) 0 (0) 28 (35%)
Oliguria alone 33 (33%) 0 (0) 33 (41%)
Both criteria 20 (20%) 0 (0) 20 (25%)
None 19 (19%) 19 (100%) 0 (0)
Day-28 mortality 29 (29%) 1 (5%) 28 (35%) 0.02
Specific treatment during ICU stay 0.35
Chloroquine or Hydroxychloroquine 3 (3%) 0 (0) 3 (4%)
Eculizumab 2 (2%) 0 (0) 2 (3%)
Lopinavir/ritonavir 10 (10%) 0 (0) 10 (12%)
Tocilizumab 1 (1%) 0 (0) 1 (1%)
None 84 (84%) 19 (100%) 65 (80%)
Results are presented as median (interquartile) or n (%)
ACE angiotensin converting enzyme inhibitors, ARB angiotensin receptor blockers
Joseph et al. Ann. Intensive Care (2020) 10:117 Page 5 of 8

Fig. 1  Boxplots depicting relationship between AKI severity and C3 [ng/mL] (p = 0.01) (a), IL-6 [ng/mL] (p = 0.02) (b), sC5b9 [ng/mL] (p = 0.42) (c)
and ferritin levels [mg/L] (p = 0.03) (d) levels

Fig. 2  Kaplan–Meier curves for day-28 survival in patients with (n = 81) and without AKI (n = 19) (a) and with AKI stage 1 (n = 44), 2 (n = 10) and 3
(n = 27) (b)

KDIGO serum creatinine criteria, diuresis was inconsist- With 81% of patients diagnosed with AKI, the rate of AKI
ently taken into account, and none of them reported AKI in our study is much higher than previously reported.
stages. Our study is the largest published to date report- Interestingly, one of the largest cohort from the United
ing incidence of AKI specifically in critically ill patients. States reported a prevalence of AKI in COVID-19
Joseph et al. Ann. Intensive Care (2020) 10:117 Page 6 of 8

Table 2  Factors associated with survival censored at day 28 (Cox Model) and risk of AKI (logistic regression)
Estimate 95% CI p value

Model 1 (survival censored at day 28) Hazard ratio


 Modified SOFA score at admission (per point) 1.23 1.04–1.45 0.01
 Acute Kidney Injury 3.08 1.12–8.45 0.03
Model 2 (acute kidney injury) Odds ratio
 Chronic kidney disease 3.91 0.97–26.20 0.09
 Modified SOFA score at admission (per point) 1.29 1.04–1.70 0.04
Model selection was performed according to forward variable selection conditioned on p value (critical entry p value < 0.2, critical exit p value < 0.1)

patients requiring mechanical ventilation of 80% [22], in COVID-19-induced-AKI, even though complement
close to our finding of 90%. dysregulation may be involved in the most severe forms
One strength of our study is the rigorous use of KDIGO of AKI.
criteria, including urinary output. Furthermore, given Although our study did not include extensive coagu-
that many patients will present with AKI without a reli- lation explorations [31], the lack of difference in fibrin-
able baseline serum creatinine on record, estimation of ogen levels in patients with AKI when compared to
the latter is of tremendous importance. Several methods patients without AKI does not support the evidence
to estimate missing baseline creatinine can lead to varia- of a role of hypercoagulability in COVID-19-induced-
tions in incidence of AKI up to 15% [6]. Using complete AKI, suggested by the presence of thrombi in glomeru-
KDIGO definition we found a 80% incidence of AKI dur- lar loops described by others [32, 33]. Last, high levels
ing the first 7  days of ICU stay and a 90% incidence in of PEEP in COVID-19 patients [34, 35] have also been
patients requiring mechanical ventilation. suggested as a potential factor for increased AKI in
We also tested the hypotheses that COVID-19-as- severe COVID-19 patients [36]. In our study, PEEP lev-
sociated-AKI is linked to the elevation of cytokines els were not independently associated with the devel-
induced by SARS-CoV-2 infection [23], complement opment of AKI, but the lack of statistical power and
dysregulation [12] induced by SARS-CoV-2 infection longitudinal data on PEEP levels does not allow for any
or mechanical ventilation settings. High levels of IL-6 definite conclusion.
have been associated with the development of severe Our study also has limitations. First, despite being the
disease [24, 25] and acute respiratory distress syn- largest focusing on COVID-19-induced-AKI in the ICU,
drome [8] during COVID-19 infection, but the role the limited number of patients can translate into a lack
of inflammation markers in COVID-19-induced-AKI of statistical power. Nevertheless, rate of AKI and sample
remains speculative [7]. The deleterious role of IL-6 has size allow confirming a high AKI incidence with a rea-
been demonstrated in different models of AKI, includ- sonable level of confidence (81%; 95% CI 72–89). In addi-
ing ischemic AKI, nephrotoxin-induced AKI and sep- tion, the monocenter design may have limited external
sis-induced AKI [26, 27]. In our study, IL-6 and ferritin validity of our findings. The proportion of missing base-
levels correlated with severity but were not indepen- line serum creatinine value (67%) may also be an issue.
dently associated with AKI. The complement system Missing baseline creatinine value is a common problem
represents the first response of the host immune sys- in research focusing on AKI, and although most studies
tem. It participates in the development of AKI [28] and do not report the proportion of missing baseline values,
has also been suspected to play a role in AKI in the rates as high as 85% are common [22]. We provide sensi-
context of SARS-CoV-2 infection [10, 12]. Recent stud- tivity analyses (Additional file 1: Figure S3 and Table S2)
ies showed a strong immunohistochemical staining of to show that back calculation of missing baseline creati-
complement cascade components in the lungs [29] and nine values unlikely results in a significant bias. Last, lack
kidneys [30] of severe COVID-19 patients. However, of association between inflammation biomarker, IL-6 or
even if C3 levels were associated with AKI severity in complement component do not preclude participation
univariate analysis, the association did not persist when of these mechanisms to AKI. They only underline that
forced into a multivariate model, and soluble C5b9 in this setting and when adjusted to severity, these fac-
showed no association with AKI. C3 level was only tors have little influence on AKI rate. Additional stud-
independently associated with AKI stage 2 and 3 com- ies, assessing levels of proteinuria and hematuria [22,
pared to no AKI or AKI stage 1. Overall, our results 37], pathological findings and translational research are
do not support a role for complement dysregulation needed to further explore different mechanisms that may
participate to AKI during severe SARS-CoV-2 infection.
Joseph et al. Ann. Intensive Care (2020) 10:117 Page 7 of 8

In conclusion, we did not find any association between 2. Guan W, Ni Z, Hu Y, Liang W, Ou C, He J, et al. Clinical characteristics of
coronavirus disease 2019 in China. N Engl J Med. 2020. https​://www-
complement activation or inflammatory markers and nejm-org.proxy​.inser​mbibl​io.inist​.fr/doi/10.1056/NEJMo​a2002​032.
AKI. Our study suggests a tremendously high incidence 3. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of
of AKI in our cohort of critically ill COVID-19 patients, patients infected with 2019 novel coronavirus in Wuhan China. Lancet.
2020;395(10223):497–506.
along with an independent association between AKI and 4. Gabarre P, Dumas G, Dupont T, Darmon M, Azoulay E, Zafrani L. Acute
outcome. Because of its marked influence on outcome, kidney injury in critically ill patients with COVID-19. Intensive Care Med.
AKI should be identified promptly during SARS-CoV-2 2020;46:1339.
5. Koeze J, Keus F, Dieperink W, van der Horst ICC, Zijlstra JG, van Meurs
infection. M. Incidence, timing and outcome of AKI in critically ill patients varies
with the definition used and the addition of urine output criteria. BMC
Supplementary information Nephrol. 2017;18(1):70.
6. Wiersema R, Jukarainen S, Eck RJ, Kaufmann T, Koeze J, Keus F, et al. Differ-
Supplementary information accompanies this paper at https​://doi.
ent applications of the KDIGO criteria for AKI lead to different incidences
org/10.1186/s1361​3-020-00734​-z.
in critically ill patients: a post hoc analysis from the prospective observa-
tional SICS-II study. Crit Care Lond Engl. 2020;24(1):164.
Additional file 1: Figure S1. Boxplots depicting relationship between AKI 7. Batlle D, Soler MJ, Sparks MA, Hiremath S, South AM, Welling PA, et al.
stage 2 and 3 and C3 [ng/mL] (A), IL-6 [ng/mL] (B), sC5b9 [ng/mL] (C) and Acute kidney injury in COVID-19: emerging evidence of a distinct patho-
ferritin levels [mg/L] (D) levels, and predicted probabilities of severe AKI physiology. J Am Soc Nephrol. 2020;31:1380. https​://jasn-asnjo​urnal​s-org.
according to ferritin (E) and C3 (F) deciles (per log). Figure S2. Kaplan- proxy​.inser​mbibl​io.inist​.fr/conte​nt/early​/2020/05/04/ASN.20200​40419​
Meier curve for day-28 survival in patients without AKI or with AKI stage 8. Wu C, Chen X, Cai Y, Xia J, Zhou X, Xu S, et al. Risk factors associated
1 (n=63) compared to AKI stage 2 or 3 (n= 37). Figure S3. Kaplan-Meier with acute respiratory distress syndrome and death in patients with
curves for day-28 survival according to AKI (A and B) and AKI stages (C and coronavirus disease 2019 pneumonia in Wuhan China. JAMA Intern Med.
D) in patients with (n=33, B and D) and without (n= 67, A and C) baseline 2020;18:1.
creatinine value. Table S1. Factors associated with risk of AKI stage 2 and 9. Colafrancesco S, Alessandri C, Conti F, Priori R. COVID-19 gone bad: a new
3, compared to no AKI or AKI stage 1, after adjustment for modified SOFA character in the spectrum of the hyperferritinemic syndrome? Autoim-
and chronic kidney disease (logistic regression). Table S2. Interaction mun Rev. 2020;5:102573.
between missing baseline serum creatinine and reported results. 10. Noris M, Benigni A, Remuzzi G. The case of Complement activation in
COVID-19 multiorgan impact. Kidney Int. 2020;98:314.
11. Xu X, Han M, Li T, Sun W, Wang D, Fu B, et al. Effective treatment
Abbreviations of severe COVID-19 patients with tocilizumab. Proc Natl Acad Sci.
ACE2: Angiotensin-converting enzyme 2; AKI: Acute kidney injury; COVID-19: 2020;117(20):10970–5.
Coronavirus disease 2019; ICU: Intensive care unit; KDIGO: Kidney disease 12. Risitano AM, Mastellos DC, Huber-Lang M, Yancopoulou D, Garlanda C,
improving global outcome; PEEP: Positive end expiratory pressure; SARS- Ciceri F, et al. Complement as a target in COVID-19? Nat Rev Immunol.
COV-2: Severe acute respiratory coronavirus 2. 2020;20(6):343–4.
13. Kellum JA, Lameire N, Aspelin P, Barsoum RS, Burdmann EA, Goldstein SL,
Authors’ contributions et al. Kidney disease: improving global outcomes (KDIGO) acute kidney
MD, LZ and EA contributed to the study conception and design. AJ, MD, AM injury work group. KDIGO clinical practice guideline for acute kidney
and LZ performed the data collection and the initial data analysis. AJ, LZ and injury. Kidney Int Suppl. 2012;2(1):1–138.
MD prepared the first draft of the manuscript. All authors contributed to the 14. Vincent J-L, Moreno R, Takala J, Willatts S, De Mendonça A, Bruining
data analysis and to the critical revision. All authors read and approved the H, et al. The SOFA (Sepsis-related Organ Failure Assessment) score to
final manuscript. describe organ dysfunction/failure: on behalf of the working group
on sepsis-related problems of the European Society of Intensive Care
Ethics approval and competing interests Medicine (see contributors to the project in the appendix). Intensive Care
MD declares having received grant from MSD, speaker fees from MSD, Astelas Med. 1996;22(7):707–10.
and Gilead-Kite and having attended an advisory board for Gilead-Kite. LZ 15. Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al. Clinical characteristics of
declares having received research grant from Jazz Pharmaceuticals. The other 138 hospitalized patients with 2019 novel coronavirus-infected pneumo-
authors have no conflict of interest to declare. nia in Wuhan, China. JAMA. 2020;323:1061.
16. Wang L, Li X, Chen H, Yan S, Li D, Li Y, et al. Coronavirus disease 19 infec-
Consent for publication tion does not result in acute kidney injury: an analysis of 116 hospitalized
Not applicable. patients from Wuhan, China. Am J Nephrol. 2020;51(5):343–8.
17. Zhang G, Hu C, Luo L, Fang F, Chen Y, Li J, et al. Clinical features and short-
Author details term outcomes of 221 patients with COVID-19 in Wuhan, China. J Clin
1
 Service de médecine Intensive et de réanimation médicale, Hôpital Saint- Virol. 2020;1(127):104364.
Louis, Assistance-Publique Hôpitaux de Paris, Paris University, 1 avenue Claude 18. Cao J, Hu X, Cheng W, Yu L, Tu W-J, Liu Q. Clinical features and short-term
Vellefaux, 75010 Paris, France. 2 ECSTRA Team, UMR 1153, Center of Epidemiol- outcomes of 18 patients with corona virus disease 2019 in intensive care
ogy and Biostatistics, INSERM, Université de Paris, Paris, France. 3 INSERM U976, unit. Intensive Care Med. 2020;46(5):851–3.
Université de Paris, Paris, France. 19. Xu S, Fu L, Fei J, Xiang H-X, Xiang Y, Tan Z-X, et al. Acute kidney injury at
early stage as a negative prognostic indicator of patients with COVID-
Received: 11 June 2020 Accepted: 27 August 2020 19: a hospital-based retrospective analysis. medRxiv. 2020. https​://doi.
org/10.1101/2020.03.24.20042​408.
20. Luo X, Xia H, Yang W, Wang B, Guo T, Xiong J, et al. Characteristics of
patients with COVID-19 during epidemic ongoing outbreak in Wuhan,
China. medRxiv. 2020. https​://doi.org/10.1101/2020.03.19.20033​175.
References 21. Hu L, Chen S, Fu Y, Gao Z, Long H, Wang J-M, et al. Risk factors associated
1. COVID-19 Map. Johns Hopkins Coronavirus Resource Center. https​:// with clinical outcomes in 323 COVID-19 hospitalized patients in Wuhan,
coron​aviru​s.jhu.edu/map.html. China. Clin Infect Dis. 2020. https​://doi.org/10.1093/cid/ciaa5​39.
22. Hirsch JS, Ng JH, Ross DW, Sharma P, Shah HH, Barnett RL, et al. Acute kid-
ney injury in patients hospitalized with COVID-19. Kidney Int. 2020;98:209.
Joseph et al. Ann. Intensive Care (2020) 10:117 Page 8 of 8

https​://www.kidne​y-inter​natio​nal.org/artic​le/S0085​–2538(20)30532​–9/ 31. Mortus JR, Manek SE, Brubaker LS, Loor M, Cruz MA, Trautner BW, et al.
abstr​act Thromboelastographic results and hypercoagulability syndrome in
23. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ, et al. patients with coronavirus disease 2019 who are critically ill. JAMA Netw
COVID-19: consider cytokine storm syndromes and immunosuppression. Open. 2020;3(6):e2011192–e20111922011192.
Lancet Lond Engl. 2020;395(10229):1033–4. 32. Su H, Yang M, Wan C, Yi L-X, Tang F, Zhu H-Y, et al. Renal histopathological
24. Liu F, Li L, Xu M, Wu J, Luo D, Zhu Y, et al. Prognostic value of interleukin-6, analysis of 26 postmortem findings of patients with COVID-19 in China.
C-reactive protein, and procalcitonin in patients with COVID-19. J Clin Kidney Int. 2020;98:219. https​://linki​nghub​.elsev​ier.com/retri​eve/pii/
Virol. 2020;127:104370. S0085​25382​03036​90
25. Zhang X, Tan Y, Ling Y, Lu G, Liu F, Yi Z, et al. Viral and host factors related 33. Helms J, Tacquard C, Severac F, Leonard-Lorant I, Ohana M, Delabranche
to the clinical outcome of COVID-19. Nature [Internet]. 2020 May 20 X, et al. High risk of thrombosis in patients with severe SARS-CoV-2
[cited 2020 Jun 6]; Available from: https​://www.natur​e.com/artic​les/s4158​ infection: a multicenter prospective cohort study. Intensive Care Med.
6–020–2355–0 2020;46:1089.
26. Vuille-dit-Bille RN, Camargo SM, Emmenegger L, Sasse T, Kummer E, 34. Beloncle FM, Pavlovsky B, Desprez C, Fage N, Olivier P-Y, Asfar P, et al.
Jando J, et al. Human intestine luminal ACE2 and amino acid transporter Recruitability and effect of PEEP in SARS-Cov-2-associated acute respira-
expression increased by ACE-inhibitors. Amino Acids. 2015;47(4):693–705. tory distress syndrome. Ann Intensive Care. 2020;10(1):55.
27. Ferrario CM, Jessup J, Chappell MC, Averill DB, Brosnihan KB, Tallant EA, 35. Gattinoni L, Chiumello D, Caironi P, Busana M, Romitti F, Brazzi L, et al.
et al. Effect of angiotensin-converting enzyme inhibition and angiotensin COVID-19 pneumonia: different respiratory treatments for different
II receptor blockers on cardiac angiotensin-converting enzyme 2. Circula- phenotypes? Intensive Care Med. 2020;46:1099.
tion. 2005;111(20):2605–10. 36. Husain-Syed F, Slutsky AS, Ronco C. Lung-kidney cross-talk in the critically
28. McCullough JW, Renner B, Thurman JM. The role of the complement ill patient. Am J Respir Crit Care Med. 2016;194(4):402–14.
system in acute kidney injury. Semin Nephrol. 2013;33(6):543–56. 37. Cheng Y, Luo R, Wang K, Zhang M, Wang Z, Dong L, et al. Kidney disease
29. Gao T, Hu M, Zhang X, Li H, Zhu L, Liu H, et al. Highly pathogenic corona- is associated with in-hospital death of patients with COVID-19. Kidney Int.
virus N protein aggravates lung injury by MASP-2-mediated complement 2020;97(5):829–38.
over-activation. Infect Dis. 2020. https​://doi.org/10.1101/2020.03.29.20041​
962.
30. Diao B, Feng Z, Wang C, Wang H, Liu L, Wang C, et al. Human Publisher’s Note
kidney is a target for novel severe acute respiratory syndrome Springer Nature remains neutral with regard to jurisdictional claims in pub-
coronavirus 2 (SARS-CoV-2) infection. medRxiv. 2020. https​://doi. lished maps and institutional affiliations.
org/10.1101/2020.03.04.20031​120.
CLINICAL RESEARCH www.jasn.org

Clinicopathological Features and Outcomes of Acute


Kidney Injury in Critically Ill COVID-19 with Prolonged
Disease Course: A Retrospective Cohort
Peng Xia,1 Yubing Wen,1 Yaqi Duan,2,3 Hua Su,4 Wei Cao,5 Meng Xiao,6 Jie Ma,1
Yangzhong Zhou,1 Gang Chen,1 Wei Jiang,7 Huanwen Wu,8 Yan Hu,1 Sanpeng Xu,2
Hanghang Cai,2 Zhengyin Liu,5 Xiang Zhou,9 Bin Du ,7 Jinglan Wang,10 Taisheng Li,5
Xiaowei Yan,11 Limeng Chen,1 Zhiyong Liang,8 Shuyang Zhang,11 Chun Zhang ,4
Yan Qin ,1 Guoping Wang,2,3 and Xuemei Li1
Due to the number of contributing authors, the affiliations are listed at the end of this article.

ABSTRACT
Background The incidence, severity, and outcomes of AKI in COVID-19 varied in different reports. In patients
critically ill with COVID-19, the clinicopathologic characteristics of AKI have not been described in detail.
Methods This is a retrospective cohort study of 81 patients critically ill with COVID-19 in an intensive care
unit. The incidence, etiologies, and outcomes of AKI were analyzed. Pathologic studies were performed in
kidney tissues from ten deceased patients with AKI.
Results A total of 41 (50.6%) patients experienced AKI in this study. The median time from illness to AKI
was 21.0 (IQR, 9.5–26.0) days. The proportion of Kidney Disease Improving Global Outcomes (KDIGO)
stage 1, stage 2, and stage 3 AKI were 26.8%, 31.7%, and 41.5%, respectively. The leading causes of AKI
included septic shock (25 of 41, 61.0%), volume insufficiency (eight of 41, 19.5%), and adverse drug effects
(five of 41, 12.2%). The risk factors for AKI included age (per 10 years) (HR, 1.83; 95% CI, 1.24 to 2.69;
P50.002) and serum IL-6 level (HR, 1.83; 95% CI, 1.23 to 2.73; P50.003). KDIGO stage 3 AKI predicted
death. Other potential risk factors for death included male sex, elevated D-dimer, serum IL-6 level, and
higher Sequential Organ Failure Assessment score. The predominant pathologic finding was acute tubular
injury. Nucleic acid tests and immunohistochemistry failed to detect the virus in kidney tissues.
Conclusions AKI was a common and multifactorial complication in patients critically ill with COVID-19 at
the late stage of the disease course. The predominant pathologic finding was acute tubular injury. Older
age and higher serum IL-6 level were risk factors of AKI, and KDIGO stage 3 AKI independently predicted death.

JASN 31: 2205–2221, 2020. doi: https://doi.org/10.1681/ASN.2020040426

Received April 9, 2020. Accepted July 22, 2020.


Coronavirus disease 2019 (COVID-19) is a new P.X., Y.W., Y.D., H.S., and W.C. contributed equally to this work.
contagious disease caused by severe acute respi-
Published online ahead of print. Publication date available at www.jasn.org.
ratory syndrome coronavirus 2 (SARS-CoV-2),
causing .10 million cases and 500,000 deaths Correspondence: Dr. Yan Qin, Department of Nephrology, Pe-
worldwide.1,2 Most of patients had mild cases of king Union Medical College Hospital, Chinese Academy of
Medical Sciences, No.1 Shuaifuyuan, Wangfujing Street, Beijing
the disease which primarily presented as acute re- 100730, China, or Dr. Guoping Wang, Institute of Pathology,
spiratory illness.3–7 About 15.7%–26.0% of the Tongji Hospital, Tongji Medical College, Huazhong University of
Science and Technology, No.1095 Jie Fang Avenue, Hankou,
COVID-19 cases were classified as severe or criti-
Wuhan 430030, China and Department of Pathology, School of
cally ill, with mortality as high as 60%.3,7,8 Based on Basic Medical Science, Tongji Medical College, Huazhong Uni-
the published studies, the median time from illness versity of Science and Technology, Wuhan 430030, China.
E-mail: qinyanbeijing@126.com or wanggp@hust.edu.cn
to death in early severe cases was 16–18.5 days.3,7,9
Early reports suggested the renal involvement in Copyright © 2020 by the American Society of Nephrology

JASN 31: 2205–2221, 2020 ISSN : 1046-6673/3109-2205 2205


CLINICAL RESEARCH www.jasn.org

patients with COVID-19 might not be very common.7,10,11


Significance Statement
Recent studies showed the incidence of AKI in severe cases
varied from 2.9% to 50%, due to different definitions and Coronavirus disease 2019 (COVID-19) is a new contagious disease.
clinical situations.7,8,10,12–14 The detection of SARS-CoV-2 Previous studies reported AKI with varying results regarding the
incidence, severity, and outcomes. This study provides detailed
in urine samples and kidney tissues of patients with
clinical data of 81 patients critically ill with COVID-19 and a prolonged
COVID-19 also showed varying results (B. Diao, C. Wang, disease course, and provides renal pathologic findings from ten de-
R. Wang, Z. Feng, Y. Tan, H. Wang, et al.: Human kidney is ceased patients with AKI in a single intensive care unit in Wuhan,
a target for novel severe acute respiratory syndrome corona- China. The incidence of AKI was 50.6%, with 41.5% of cases of AKIs
virus 2 [SARS-CoV-2] infection, 2020; doi:10.1101/2020.03. were Kidney Disease Improving Global Outcomes (KDIGO) stage 3.
The primary pathological findings were those of acute tubular injury.
04.20031120).15–19 Elevated inflammatory markers, such as
Nucleic acid tests and immunohistochemistry failed to detect the virus
high-sensitivity C-reactive protein and IL-6, in patients with in kidney tissues. Older age and serum IL-6 levels were risk factors of
COVID-19 were described.7,8 It is reasonable to raise the con- AKI. KDIGO stage 3 AKI independently predicted death.
cern that kidney damage might be related to direct viral in-
fection of the kidney and the following hyperinflammation
process.20 So far, the limited evidence of kidney histologic requiring mechanical ventilation, and a combination of other
changes in COVID-19 cases revealed mainly tubular in- organ failures and admission to ICU.
jury.10,18,21 Electron microscopic (EM) evidence of virus in-
Data Collection and Definitions
fection in kidney was also under debate.18,22,23 In summary, it
is a pressing demand to improve our understanding of the The electronic medical records, laboratory results, and medical
kidney involvement in COVID-19 in different clinical con- order lists were carefully reviewed to extract the data. AKI was
texts, especially in patients who are critically ill with a relative- diagnosed according to the Kidney Disease Improving Global
ly long disease course. In this study, we provide a detailed Outcomes (KDIGO) clinical practice guidelines.25 For those
description of the clinical data from 81 patients who were who had no baseline serum creatinine (sCr) data, the diagnosis
critically ill, and renal pathologic findings from ten deceased was made by the clinical evaluations of three independent ne-
patients with AKI in intensive care unit (ICU) settings. phrologists. eGFR was calculated based on the CKD–
Epidemiology Collaboration equation using sCr. Volume
insufficiency–induced AKI was diagnosed based on the history
of poor intake and/or volume loss and rapid recovery of AKI
METHODS
after volume supplementation. Fever was defined as an axillary
temperature of at least 37.3°C. Sepsis and septic shock were
Study Design and Participants
defined according to the 2016 Third International Consensus
This is a single-center, retrospective study conducted in an Definitions for Sepsis and Septic Shock.26 Acute respiratory
ICU designated for patients critically ill with COVID-19 at distress syndrome (ARDS) was diagnosed according to the Ber-
the Sino-French New City Campus of Tongji Hospital in Wu- lin definition.27 Cardiac injury was diagnosed if serum levels of
han, China, which is run by the medical team from Peking cardiac biomarkers (e.g., high-sensitivity cardiac troponin I)
Union Medical College Hospital (PUMCH). All of the pa- were above the 99th percentile upper reference limit. Coagul-
tients in ICU who met the criteria of having a critical case of opathy was defined as a 3-second extension of prothrombin
the disease from February 5 to March 20, 2020 were time (PT) or a 5-second extension of activated partial throm-
included in this study. The deceased adult patients whose boplastin time. Hypoalbuminemia was defined as serum albu-
family member had provided oral consent for a minimally min ,25 g/L. The primary outcome was defined as death in
invasive autopsy received needle autopsies of the kidneys. hospital before March 20, 2020.
The written consent was waived due to the visiting restric-
tions during the epidemic. This study was approved by the Minimally Invasive Needle Autopsy of Kidneys
PUMCH Institutional Review Board (ZS-2328, SK-1197). The minimally invasive needle autopsies were performed by
The diagnosis of COVID-19 was made according to the the nephrologists using the 16-guage biopsy needle (BARD
Guideline of Chinese National Health Commission (Fifth MN1620) under ultrasound guidance. The autopsies were
Trial Edition).24 The clinical diagnosis criteria were as fol- performed in the right kidneys of the deceased patients. The
lows: (1) fever or respiratory symptoms, (2) leukopenia or first slices of kidney tissues were sent for PCR tests of SARS-
lymphopenia, and (3) computerized tomography scan CoV-2. Other samples were fixed in 4% neutral formaldehyde
showing radiographic abnormalities in lung. Those with and 2.5% glutaraldehyde for further pathologic processing.
two or more clinical diagnosis criteria and a positive result
to high-throughput sequencing or RT-PCR assay of SARS- Light Microscopic Examinations and Transmission EM
CoV-2 were diagnosed with COVID-19. Examinations
Critically ill COVID-19 cases were defined as including at Specimens were fixed with 4% neutral formaldehyde and em-
least one of the following: septic shock, respiratory failure bedded in paraffin wax. Sections (2 mm thick) were cut;

2206 JASN JASN 31: 2205–2221, 2020


www.jasn.org CLINICAL RESEARCH

Table 1. Demographic and clinical characteristics of patients


Presence of AKI Survival
All Patients
Characteristics Non-AKI P Nonsurvivors Survivors
(n581) AKI (n541) P Value
(n540) Value (n560) (n521)
Clinical characteristics
Age (yr), mean6SD 66.6611.4 69.669.3 63.6612.7 0.02a 68.2610.4 62.0613.3 0.03a
Sex, n (%)
Female 27 (33.3%) 11 (26.8%) 16 (40.0%) 0.21 15 (25.0%) 12 (57.1%) 0.007a
Male 54 (66.7%) 30 (73.2%) 24 (60.0%) – 45 (75.0%) 9 (42.9%) –
APACHE II score on ICU day 1, 15 (11–19) 16 (12–22) 14 (10–16) 0.02a 16 (12–20) 12 (10–16) 0.01a
median (IQR)
SOFA score on ICU day 1, median 6 (4–8) 7 (5–10) 6 (4–8) 0.03a 7 (5–10) 4 (3–5) ,0.001a
(IQR)
Median arterial pressure (mm Hg), 93 (82–103) 93 (82–104) 92 (82–103) 0.79 90 (80–103) 97 (89–104) 0.16
median (IQR)
Comorbidity, n (%)b
Hypertension 43 (53.1%) 23 (56.1%) 20 (50.0%) 0.58 30 (50.0%) 13 (61.9%) 0.35
Diabetes mellitus 19 (23.5%) 11 (26.8%) 8 (20.0%) 0.47 12 (20.0%) 7 (33.3%) 0.22
Coronary heart disease 17 (21.0%) 8 (19.5%) 9 (22.5%) 0.74 12 (20.0%) 5 (23.8%) 0.71
Cerebrovascular disease 11 (13.6%) 2 (4.9%) 9 (22.5%) 0.02a 6 (10.0%) 5 (23.8%) 0.11
CKD 3 (3.7%) 1 (2.4%) 2 (5%) 0.62 1 (1.6%) 2 (9.5%) 0.16
Current smoker, n (%)c 12 (16.0%) 7 (18.4%) 5 (13.5%) 0.56 9 (16.7%) 3 (14.3%) .0.99
Complications, n (%)
ARDS 77 (95.1%) 37 (90.2%) 40 (100.0%) – 57 (95.0%) 20 (95.2%) .0.99
Septic shock 51 (63.0%) 28 (68.3%) 23 (57.5%) 0.32 44 (73.3%) 7 (33.3%) 0.001a
Altered mental status 39 (48.1%) 19 (46.3%) 20 (50.0%) 0.74 30 (50.0%) 9 (42.9%) 0.57
Cardiac injury 61 (79.0%) 33 (80.5%) 31 (77.5%) 0.74 51 (85.0%) 13 (61.9%) 0.03a
Arrythmias 29 (35.8%) 17 (41.5%) 12 (30.0%) 0.28 25 (41.7%) 4 (19.0%) 0.07
Coagulopathy 51 (63.0%) 30 (73.2%) 21 (52.5%) 0.05 43 (71.7%) 8 (38.1%) 0.006a
Liver injury 32 (39.5%) 19 (46.3%) 13 (32.5%) 0.20 28 (46.7%) 4 (19.0%) 0.03a
Hypercapnia 44 (54.3%) 26 (63.4%) 18 (45.0%) 0.10 37 (61.7%) 7 (33.3%) 0.03a
Secondary infection 47 (58.0%) 26 (63.4%) 21 (52.5%) 0.32 34 (56.7%) 13 (61.9%) 0.68
Pneumothorax 7 (8.6%) 5 (12.2%) 2 (5.0%) 0.43 7 (11.7%) 0 (0.0%) –
AKI 41 (50.6%) – – – 34 (56.7%) 7 (33.3%) 0.07
Initial symptoms, n (%)
Fever 72 (88.9%) 34 (82.9%) 38 (95.0%) 0.08 54 (90.0%) 18 (85.7%) 0.69
Cough 63 (77.8%) 33 (80.5%) 30 (75.0%) 0.55 46 (76.7%) 17 (81.0%) 0.77
Sputum 33 (40.7%) 15 (36.6%) 18 (45.0%) 0.44 22 (36.7%) 11 (52.4%) 0.21
Fatigue 46 (56.8%) 21 (51.2%) 25 (62.5%) 0.31 34 (56.7%) 12 (57.1%) 0.97
Anorexia 26 (32.1%) 11 (26.8%) 15 (37.5%) 0.30 22 (36.7%) 4 (19.0%) 0.18
Vomit 8 (9.9%) 4 (9.8%) 4 (10.0%) 0.97 5 (8.3%) 3 (14.3%) 0.42
Diarrhea 20 (24.7%) 9 (22.0%) 11 (27.5%) 0.56 17 (28.3%) 3 (14.3%) 0.25
Headache 9 (11.1%) 6 (14.6%) 3 (7.5%) 0.48 8 (13.3%) 1 (4.8%) 0.43
Myalgia 15 (18.5%) 6 (14.6%) 9 (22.5%) 0.36 8 (13.3%) 7 (33.3%) 0.04a
Dyspnea 55 (67.9%) 29 (70.7%) 26 (65.0%) 0.58 42 (70.0%) 13 (61.9%) 0.49
a
P,0.05.
b
Hypertension and diabetes histories were diagnosed based on self-reports from patients or family members. Coronary heart disease was diagnosed based the self-
reports from patients or family members about the coronary angiography results or histories of coronary stenting. Cerebrovascular disease was diagnosed based on
the self-reports from patients or family members about the histories of ischemic or hemorrhagic cerebrovascular incidents and positive image findings. CKD was
defined as abnormalities of kidney structure or function for at least 3 months.
c
All patients, n575; AKI, n538; non-AKI, n537; nonsurvivors, n554; survivors, n521.

stained with hematoxylin and eosin, Periodic acid–Schiff, Pe- tetroxide postfixation and gradient dehydration, Epon-
riodic acid–silver methenamine, and Masson trichrome; and embedded, Toluidine Blue–stained semithin sections were
scanned as high-resolution digital images (Nanozoomer Dig- examined, and selected areas were chosen for thin sections.
ital Pathology, NDP-2.0RS; Hamamatsu Photonics, Hama- Thin sections were then cut and stained with uranyl acetate
matsu, Japan). and lead citrate. EM grids were then viewed with a trans-
For EM examination, tissues were fixed in 2.5% glutaral- mission electron microscope (HT-7800; Hitachi, Tokyo,
dehyde before the following procedures. After osmium Japan).

JASN 31: 2205–2221, 2020 AKI Outcome in Critically Ill COVID-19 2207
CLINICAL RESEARCH www.jasn.org

Table 2. The laboratory findings of patients


Presence of AKI Survival
All Patients
Laboratory Findings Nonsurvivors P
(n581) AKI (n541) Non-AKI (n540) P Value Survivors (n521)
(n560) Value
White blood cell count 12.20 13.11 11.60 0.41 13.05 9.93 (6.40–15.13) 0.05a
(3109/L), median (IQR) (9.00–17.29) (9.88–17.81) (8.66–15.30) (10.20–18.30)
.10, n (%) 56 (69.1%) 30 (73.2%) 26 (65.0%) 0.43 46 (76.7%) 10 (47.6%) 0.01a
4–10, n (%) 24 (29.6%) 11 (26.8%) 13 (32.5%) – 13 (21.7%) 11 (52.4%) –
,4, n (%) 1 (1.2%) 0 (0.0%) 1 (2.5%) – 1 (1.7%) 0 (0.0%) –
Neutrophil count (3109/L), 11.13 12.49 10.65 0.31 11.89 9.22 (5.64–13.10) 0.03a
median (IQR) (8.14–16.02) (9.14–16.43) (7.80–14.42) (9.33–16.87)
Lymphocytes (3109/L), 0.54 (0.34–0.74) 0.50 (0.34–0.60) 0.65 (0.34–0.88) 0.02a 0.53 (0.32–0.70) 0.69 (0.42–0.94) 0.10
median (IQR)
,0.4, n (%) 24 (29.6%) 13 (31.7%) 11 (27.5%) 0.08 19 (31.7%) 5 (23.8%) 0.59
0.4–0.8, n (%) 39 (48.1%) 23 (56.1%) 16 (40.0%) – 30 (50.0%) 9 (42.9%) –
Hemoglobin (g/L), 122.1622.8 124.5625.6 119.8619.5 0.36 124.8621.5 114.7625.2 0.08
mean6SD
Anemia, n (%) 29 (35.8%) 14 (34.1%) 15 (37.5%) 0.75 19 (31.7%) 10 (47.6%) 0.19
Platelets (3109/L), median 159.0 145.0 176.5 0.03a 153.0 202.0 0.03a
(IQR) (100.0–225.5) (77.5–209.5) (122.5–268.0) (82.5–210.8) (138.0–271.0)
,100, n (%) 19 (23.5%) 12 (29.3%) 7 (17.5%) 0.21 17 (28.3%) 2 (9.5%) 0.13
Serum albumin (g/L), 28.5 (26.1–31.0) 28.4 (25.2–31.8) 28.7 (26.2–31.0) 0.87 27.9 (24.1–30.4) 29.0 (28.1–33.0) 0.06
median (IQR)
,25, n (%) 18 (22.2%) 10 (24.4%) 8 (20.0%) 0.64 17 (28.3%) 1 (4.8%) 0.03a
ALT (U/L), median (IQR) 29.0 (19.5–45.0) 32.0 (19.0–52.5) 25.0 (20.2–39.8) 0.44 29.5 (20.2–45.8) 24.0 (16.5–38.5) 0.33
AST (U/L), median (IQR) 33.0 (20.5–63.5) 37.0 (28.0–59.0) 29.0 (19.0–68.0) 0.27 33.5 (21.0–68.2) 33.0 (20.0–54.5) 0.38
LDH (U/L), median (IQR) 523.0 562.0 504.5 0.45 558.0 373.0 0.01a
(373.5–700.5) (382.5–688.0) (345.2–742.5) (417.2–742.5) (274.5–618.5)
Total bilirubin (mmol/L), 12.9 (8.6–20.6) 12.2 (8.2–21.6) 13.4 (8.8–20.6) 0.81 14.5 (9.8–21.7) 10.3 (7.6–17.0) 0.05a
median (IQR)
sCr (mmol/L), median (IQR) 80.0 (58.0–113.0) 104.0 65.5 (48.0–80.0) ,0.001a 85.5 (65.0–114.5) 61.0 (43.5–82.0) 0.02a
(74.0–188.5)
Elevated sCr, n (%) 24 (29.6%) 21 (51.2%) 3 (7.5%) ,0.001a 20 (33.3%) 4 (19.0%) 0.27
Cystatin C (mg/L), median 1.26 (0.99–2.01) 1.74 (1.22–2.44) 1.06 (0.86–1.28) ,0.001a 1.32 (0.95–2.01) 1.22 (1.05–1.82) 0.96
(IQR)b
.1.55, n (%) 23 (35.3%) 20 (60.6%) 3 (9.1%) ,0.001a 17 (36.2%) 6 (31.6%) 0.72
eGFR (ml/min per 1.73 m2), 83.2 (50.1–99.6) 53.0 (27.0–85.8) 96.0 (79.8–107.8) ,0.001a 78.3 (45.2–97.4) 94.8 (64.5–115.9) 0.06
median (IQR)
,60, n (%) 26 (32.1%) 22 (53.7%) 4 (10.0%) ,0.001a 21 (35.0%) 5 (23.8%) 0.34
BUN (mmol/L), median 9.6 (6.4–15.4) 12.5 (8.6–27.8) 7.1 (5.5–10.6) ,0.001a 10.2 (7.0–17.8) 7.3 (5.2–10.9) 0.02a
(IQR)
Serum potassium (mmol/L), 4.37 (3.88–5.01) 4.50 (4.09–5.04) 4.19 (3.78–4.82) 0.16 4.48 (3.96–5.04) 4.16 (3.76–4.60) 0.18
median (IQR)
.5.5, n (%) 12 (14.8%) 6 (14.6%) 6 (15.0%) 0.96 9 (15.0%) 3 (14.3%) .0.99
3.5–5.5, n (%) 62 (76.5%) 31 (75.6%) 31 (77.5%) – 46 (76.7%) 16 (76.2%) –
,3.5, n (%) 7 (8.6%) 4 (9.8%) 3 (7.5%) – 5 (8.3%) 2 (9.5%) –
Serum sodium (mmol/L), 142.6 144.4 139.9 0.10 144.2 137.8 0.001a
median (IQR) (138.0–146.2) (138.9–148.2) (137.5–145.1) (139.4–146.9) (135.2–140.0)
.145, n (%) 28 (34.6%) 18 (43.9%) 10 (25.0%) 0.07 25 (41.7%) 3 (14.3%) 0.03a
135–145, n (%) 43 (53.1%) 18 (43.9%) 25 (75.0%) – 29 (48.3%) 14 (66.7%) –
,135, n (%) 10 (12.3%) 5 (12.2%) 5 (12.5%) – 6 (10.0%) 4 (19.0%) –
Serum calcium (mmol/L), 2.10 (1.96–2.29) 2.12 (1.98–2.26) 2.09 (1.95–2.29) 0.77 2.08 (1.96–2.22) 2.24 (1.98–2.34) 0.05a
median (IQR)
Serum phosphorus 0.98 (0.84–1.24) 1.12 (0.84–1.48) 0.94 (0.82–1.09) 0.09 0.99 (0.86–1.32) 0.92 (0.74–1.14) 0.14
(mmol/L), median (IQR)c
,0.81, n (%) 16 (21.9%) 8 (22.2%) 8 (21.6%) 0.95 9 (17.0%) 7 (35.0%) 0.08

2208 JASN JASN 31: 2205–2221, 2020


www.jasn.org CLINICAL RESEARCH

Table 2. Continued

Presence of AKI Survival


All Patients
Laboratory Findings Nonsurvivors P
(n581) AKI (n541) Non-AKI (n540) P Value Survivors (n521)
(n560) Value
Serum uric acid (mmol/L), 201.0 289.5 164.0 ,0.001a 208.0 187.0 0.28
median (IQR)d (137.5–327.0) (184.8–448.8) (119.2–219.2) (147.0–346.0) (125.0–259.0)
Proteinuria at admission to
ICU, n (%)e
.21 11 (15.5%) 6 (18.2%) 5 (13.2%) 0.78 10 (19.2%) 1 (5.3%) 0.27
611 50 (70.4%) 23 (69.7%) 27 (71.1%) – 37 (71.2%) 13 (68.4%) –
Negative 10 (14.1%) 4 (12.1%) 6 (15.8%) – 5 (9.6%) 5 (26.3%) –
Hematuria at admission to
ICU, n (%)e
.21 23 (32.4%) 11 (33.3%) 12 (31.6%) 0.75 20 (38.5%) 3 (15.8%) 0.09
611 24 (33.8%) 12 (36.4%) 12 (31.6%) – 18 (34.6%) 6 (31.6%) –
Negative 24 (33.8%) 10 (30.3%) 14 (36.8%) – 14 (26.9%) 10 (52.6%) –
Proteinuria at hospital
presentation, n (%)f
.21 5 (13.9%) 3 (15.8%) 2 (11.8%) .0.99 5 (17.9%) 0 (0.0%) –
611 26 (72.2%) 14 (73.7%) 12 (70.6%) – 20 (71.4%) 6 (75%) –
Negative 5 (13.9%) 2 (10.5%) 3 (17.6%) – 3 (10.7%) 2 (25.0%) –
Hematuria at hospital
presentation, n (%)f
.21 10 (25.0%) 6 (31.6%) 4 (23.5%) 0.72 9 (32.1%) 1 (12.5%) 0.40
611 17 (47.2%) 9 (47.4%) 8 (47.4%) – 14 (50.0%) 3 (37.5%) –
Negative 9 (27.8%) 4 (21.1%) 5 (21.1%) – 5 (17.9%) 4 (50.5%) –
ESR (mm/h), median (IQR)g 40.5 (27.8–65.5) 47.5 (32.0–72.0) 35.0 (22.0–61.8) 0.10 38.5 (24.5–58.5) 67.0 (31.0–91.0) 0.02a
hsCRP (mg/L), median 105.5 109.0 66.0 (36.1–140.7) 0.10 108.9 66.0 (29.0–122.5) 0.06
(IQR)h (50.8–142.2) (66.8–147.9) (58.8–149.4)
IL-6 (pg/ml), median (IQR)i 59.0 (31.0–167.2) 100.4 36.8 (20.3–103.2) 0.01a 93.2 (35.5–214.9) 31.8 (22.1–69.5) 0.002a
(36.4–265.6)
Ferritin (mg/ml), median 1386.4 1301.8 1484.8 0.62 1469.0 973.3 0.34
(IQR)j (758.7–2207.8) (758.7–2207.8) (765.2–2229.8) (831.3–2261.3) (649.7–2207.8)
PT (s), median (IQR) 16.3 (15.3–18.1) 17.2 (15.3–18.5) 16.0 (15.1–17.2) 0.04a 17.0 (15.3–18.2) 15.6 (14.6–16.2) 0.006a
aPTT (s), median (IQR) 41.0 (37.2–45.5) 41.7 (37.4–46.8) 40.9 (35.9–43.6) 0.37 40.7 (37.0–45.9) 43.0 (38.2–45.4) 0.48
Fibrinogen (g/L), median 3.95 (3.08–5.56) 4.08 (2.92–5.57) 3.92 (3.10–5.55) 0.88 3.86 (2.77–5.52) 4.60 (3.42–5.62) 0.21
(IQR)
INR, median (IQR) 1.29 (1.19–1.48) 1.40 (1.20–1.52) 1.26 (1.18–1.36) 0.02a 1.36 (1.20–1.49) 1.24 (1.08–1.28) 0.009a
D-dimer (mg/ml FEU), n (%)
.21.0 36 (44.4%) 19 (46.3%) 17 (42.5%) 0.73 32 (53.3%) 4 (19.0%) 0.01a
5.0–21.0 19 (23.5%) 10 (24.4%) 9 (22.5%) – 14 (23.3%) 5 (23.8%) –
0.5–5.0 26 (32.1%) 12 (29.3%) 14 (35.0%) – 14 (23.3%) 12 (57.1%) –
,0.5 0 0 0 – 0 0 –
cTnI (pg/ml), median (IQR)k 62.6 (15.1–520.4) 69.8 (18.8–320.8) 52.4 (8.3–555.8) 0.49 67.1 (19.1–653.2) 20.5 (3.9–167.4) 0.02a
NT-proBNP (pg/ml), 992.0 1902.0 843.0 0.03a 1021.5 992.0 0.74
median (IQR)l (402.0–3589.0) (617.0–5590.0) (320.2–2929.2) (486.0–3634.5) (193.0–3808.0)
ALT, alanine aminotransferase; AST, aspartate aminotransferase; LDH, lactate dehydrogenase; ESR, erythrocyte sedimentation rate; hsCRP, high-sensitivity
C-reactive protein; aPTT, activated partial thromboplastin time; INR, international normalized ratio; FEU, fibrinogen equivalent units; cTnI; cardiac troponin I;
NT-proBNP, N-terminal prohormone of brain natriuretic peptide.
a
P#0.05.
b
All patients, n566; AKI, n533; non-AKI, n533; nonsurvivors, n547; survivors, n519.
c
All patients, n573; AKI, n536; non-AKI, n537; nonsurvivors, n553; survivors, n520.
d
All patients, n580; AKI, n540; non-AKI, n540; nonsurvivors, n559; survivors, n521.
e
All patients, n571; AKI, n533; non-AKI, n538; nonsurvivors, n552; survivors, n519.
f
All patients, n536; AKI, n519; non-AKI, n517; nonsurvivors, n528; survivors, n58.
g
All patients, n558; AKI, n530; non-AKI, n528; nonsurvivors, n542; survivors, n516.
h
All patients, n577; AKI, n539; non-AKI, n538; nonsurvivors, n556; survivors, n521.
i
All patients, n564; AKI, n532; non-AKI, n532; nonsurvivors, n545; survivors, n519.
j
All patients, n563; AKI, n531; non-AKI, n532; nonsurvivors, n548; survivors, n515.
k
All patients, n580; AKI, n540; non-AKI, n540; nonsurvivors, n559; survivors, n521.
l
All patients, n579; AKI, n539; non-AKI, n540; nonsurvivors, n558; survivors, n521.

JASN 31: 2205–2221, 2020 AKI Outcome in Critically Ill COVID-19 2209
CLINICAL RESEARCH www.jasn.org

Table 3. The treatment and outcome of patients during the hospitalization


Presence of AKI Survival
Treatment and Outcome Total (n581) Non-AKI P Nonsurvivors Survivors
AKI (n541) P Value
(n540) Value (n560) (n521)
Therapies, n (%)
Glucocorticoids 71 (87.7%) 34 (82.9%) 37 (92.5%) 0.31 54 (90.0%) 17 (81.0%) 0.28
Antiviral medication 66 (81.5%) 33 (80.5%) 33 (82.5%) 0.82 49 (81.7%) 17 (81.0%) 0.94
Antibiotics 75 (92.6%) 38 (92.7%) 37 (92.5%) .0.99 57 (95.0%) 18 (85.7%) 0.18
Intravenous Ig 66 (81.5%) 33 (80.5%) 33 (82.5%) 0.82 48 (80.0%) 18 (85.7%) 0.75
Blood transfusion 22 (27.2%) 14 (34.1%) 8 (20.0%) 0.15 18 (30.0%) 4 (19.0%) 0.40
Anticoagulation 41 (50.6%) 21 (51.2%) 20 (50.0%) 0.91 29 (48.3%) 12 (57.1%) 0.49
Anti-inflammatory therapies 13 (16.0%) 8 (19.5%) 5 (12.5%) 0.39 9 (15.0%) 4 (19.0%) 0.73
Life-sustaining treatment, n (%)
Invasive mechanic ventilation 66 (81.5%) 36 (87.8%) 30 (75.0%) 0.14 53 (88.3%) 13 (61.9%) 0.007
ECMO 5 (6.2%) 2 (4.9%) 3 (7.5%) 0.68 3 (5.0%) 2 (9.5%) 0.60
Vasopressors 63 (77.8%) 36 (87.8%) 27 (67.5%) 0.03a 56 (93.3%) 7 (33.3%) ,0.001a
CRRT 8 (9.9%) 8 (19.5%) 0 (0.0%) – 8 (13.3%) 0 (0.0%) –
Outcome, n (%)
Death 60 (74.1%) 34 (82.9%) 26 (65.0%) 0.07 – – –
Survive before this study 21 (25.9%) 7 (17.1%) 14 (35.0%) – – – –
Time from illness to admission (d), 9.0 (5.5–13.5) 8.0 (5.0–11.0) 10.5 (7.0–16.0) 0.09 9.0 (5–11.8) 11.0 (6.0–16.5) 0.11
median (IQR)
Time from illness to intubation (d), 15.5 14.0 17.0 0.13 16.0 (12.5–21.0) 13.0 0.65
median (IQR)b (12.0–21.0) (11.0–20.8) (13.0–23.5) (10.0–22.5)
Time of mechanic ventilation (d), 9.5 (3.8–16.5) 10.0 9.0 (4.0–15.0) 0.89 8 (3.0–13.5) 27.0 ,0.001a
median (IQR)b (3.0–18.0) (13.5–41.0)
Time from illness to AKI (d), median 21.0 21.0 – – 20.5 (9.0–24.2) 21.0 0.40
(IQR)c (9.5–26.0) (9.5–26.0) (15.0–35.0)
Time from illness to death (d), median 24.0 24.0 24.5 0.96 24.0 (18.0–31.8) – –
(IQR)d (18.0–31.8) (18.0–32.2) (18.8–31.0)
Time of hospitalization (d), median 17.0 18.0 15.5 (9.5–23.8) 0.76 14.0 (9.2–22.8) 29.0 ,0.001a
(IQR) (10.0–26.0) (10.0–28.0) (14.5–39.0)
Disease course before this study (d), 28.0 26.0 28.5 0.42 24.0 (18.0–31.8) 39.0 ,0.001a
median (IQR) (19.0–36.0) (18.0–36.0) (19.2–38.2) (29.0–52.5)
ECMO, extracorporeal membrane oxygenation.
a
P,0.05.
b
All patients, n566; AKI, n530; non-AKI, n536; nonsurvivors, n553; survivors, n513.
c
All patients, n541; nonsurvivors, n534; survivors, n57.
d
All patients, n560; AKI, n534; non-AKI, n526.

Immunohistochemistry Staining cell renal carcinoma was used as control for ACE2 staining.
Formalin-fixed, paraffin-embedded sections were heated in the Lung tissue from an included patient was used as control for
pressure cooker in 1003 Tris-EDTA buffer (pH 9.0) for 2 minutes staining of SARS-CoV-2.
to allow antigen retrieval. Standard immunohistochemistry (IHC)
was performed using the EnVision Detection System (K5007; Nucleic Acid Tests of SARS-CoV-2 in Urine Samples and
Dako) according to the manufacturer’s protocol. The primary an- Kidney Tissues
tibodies used were as follows: anti-CD3 (clone number F7.2.38, Patients’ urine samples were extracted on a Nextractor auto-
1:400; DAKO), anti-CD4 (clone number UMAB64, ready to use; matic extraction system (Genolution Pharmaceuticals, Seoul,
ZSGB-BIO), anti-CD8 (clone number SP16, ready to use; South Korea) with the manufacturer’s RNA extraction kits.
ZSGB-BIO), anti-CD20 (clone number L28, 1:400; DAKO), Kidney tissues were extracted manually with Qiagen RNeasy
anti-CD34 (clone number QBend-10, 1:400; DAKO), anti- Plus Universal Mini Kit (Thermo Fisher Scientific, Dreieich,
CD68 (clone number KP1, 1:400; DAKO), anti-CD138 (ready Germany) following the manufacturer’s instructions. RT-PCR
to use; ZSGB-BIO), anti-granzyme B (polyclonal, ready to use; of SARS-CoV-2 was performed using the commercially avail-
ZSGB-BIO), anti-angiotensin converting enzyme 2 (anti- able kit targeting the SARS-CoV-2-specific N-gene (BGI Bio-
ACE2; polyclonal, 1:1600; Proteintech), anti-SARS-CoV-2 technology [Wuhan] Co., Ltd., Wuhan, China) that has been
spike (clone number HA14FE2402, 1:1000; Sino Biologic). approved by the Chinese National Medical Products Admin-
Paracarcinoma normal kidney tissue from a patient with clear istration on the ABI 7500 system (Applied Biosystems,

2210 JASN JASN 31: 2205–2221, 2020


www.jasn.org CLINICAL RESEARCH

Table 4. Risk factors for AKI identified by multivariate Cox proportional hazards models

Thermo Fisher Scientific). Any positive or suspected positive with lymphocytopenia occurred in .70% of the patients.
results were repeated for a second time for confirmation. Significant elevation of D-dimer and inflammatory mark-
ers, including high-sensitivity C-reactive protein, IL-6, and
Statistical Analyses ferritin, were also observed (Table 2). The median Acute
Categoric variables were described as frequency and percent- Physiology and Chronic Health Evaluation II (APACHE
age, and continuous variables were described as using mean, II) and Sequential Organ Failure Assessment (SOFA) scores
median, and interquartile range (IQR) values. When the data were 15 (IQR, 11–19) and 6 (IQR, 4–8), respectively, on ICU
were normally distributed, independent t tests were used to day 1 (Table 1). ARDS, septic shock, and coagulopathy were
compare the means of continuous variables. Otherwise, the present in 95.1%, 63.0%, and 68.0% of the patients. A total
Mann–Whitney test was used. Although the Fisher exact test of 58.0% of patients experienced secondary infections
was used with limited data, the chi-squared test was used to after hospitalization (Supplemental Figure 1). Aggressive
compare the proportion of categoric variables. Cox propor- treatment including glucocorticoids, antiviral medication,
tional hazards models were used to identify the potential risk antibiotics, and intravenous Ig were deployed. Invasive me-
factors. Time trends of creatinine and eGFR for different chanical ventilation and vasopressors were required in
groups of patients with AKI were calculated by a cubic poly- 81.5% (66/81) and 77.8% (63/81) of the patients. Five patients
nomial regression model based on least square means fit using received extracorporeal membrane oxygenation support
GraphPad Prism 6.0. All statistical analyses were performed (Table 3).
using SPSS version 22.0 software. A P value of ,0.05 is statis- A total of 60 patients died in hospital before conclusion of
tically significant. the study. The median time from illness onset to death was
24.0 (IQR, 18.0–31.8) days. Nonsurvivors were mainly male
(45/60, 75.0%). They were also of an older age, had a higher
RESULTS white blood cell count, prolonged PT and international nor-
malized ratio, higher baseline sCr and serum IL-6 levels, and
Clinical Characteristics of Patients Critically Ill with higher APACHE II and SOFA scores (Table 1). They also
COVID-19 displayed higher prevalence of organ dysfunction including
A total of 1752 patients with COVID-19 were admitted to septic shock, liver injury, cardiac injury, and coagulopathy
the Sino-French New City Campus of Tongji Hospital from (Table 1). At the time of this study, 57.2% (12/21) of the
February 5 to March 20, 2020. Of these, 95 were transferred survivors had been discharged, 23.8% (5/21) of them had
to our ICU. Five patients transferred for non-COVID-19- been transferred to general wards, and 19.0% (4/21) of them
related conditions, and nine patients missing important were still in ICU.
data were excluded. Finally, 81 patients were included. Elevated sCr and eGFR ,60 ml/min per 1.73 m2 were pre-
The mean age of these patients was 66.6611.4 years. Male/ sent in 29.6% and 35.3% of patients, respectively, at admission
female ratio was 2:1. Comorbidities were present in over half to ICU. Of the tested patients, 15.5% had proteinuria .21,
of the patients, with hypertension being the most common whereas 32.4% had hematuria .21. AKI occurred in 41/81
(Table 1). Neutrophil-predominant leukocytosis complicated (50.6%) of patients. Patients with AKI were of an older age,

JASN 31: 2205–2221, 2020 AKI Outcome in Critically Ill COVID-19 2211
CLINICAL RESEARCH www.jasn.org

Table 5. Different multivariate Cox proportional hazards models for risk factors of in-hospital death

2212 JASN JASN 31: 2205–2221, 2020


www.jasn.org CLINICAL RESEARCH

Death within 21 days


400
Death between 21-35 days
350 Death after 35 days
Survivors with AKI
300
sCr (umol/L)
250

200

150

100

50

0
0 7 14 21 28 35 42 49 56 63
Days

Death within 21 days


120 Death between 21-35 days
Death after 35 days
100 Survivors with AKI
eGFR(ml/min.1.73m2)

80

60

40

20

0
0 7 14 21 28 35 42 49 56 63
Days

Figure 1. The patients with AKI who had a shorter disease course before death showed faster decline of renal function. Cubic
polynomial regression of (A) sCr and (B) eGFR of patients with AKI who died within 21 days (n511), between 21 and 35 days (n517),
after 35 days (n56), and those who survived (n57).

had lower lymphocyte and platelet counts, slightly prolonged SOFA score (HR, 1.08; 95% CI, 1.01 to 1.15; P50.03) predic-
PT, and higher serum IL-6 levels (Table 2). ted death (Table 5).

Risk Factors of AKI and Death Trends of sCr, AKI Staging, and Etiologies during the
The risk factors for AKI included older age (per 10 years) Disease Course
(hazard ratio [HR], 1.83; 95% CI, 1.24 to 2.69; P50.002) The median time from illness to AKI was 21.0 (IQR, 9.5–26.0)
and serum IL-6 levels (ng/ml) (HR, 1.83; 95% CI, 1.23 to days. Cubic polynomial regressions suggested the decline of
2.73; P50.003) (Supplemental Table 1, Table 4). Univariate renal function was faster in patients with AKI who died within
Cox proportional hazards models showed that older age, male shorter course of the disease (Figure 1). Seven of the 81 (8.6%)
sex, elevated baseline sCr, reduced eGFR, leukocytosis, hypo- patients had AKI at hospital presentation, whereas 21.0% (17/
albuminemia, hypernatremia, elevated D-dimer, and higher 81) of the patients had AKIs at ICU admission. The proportion
APACHE II and SOFA scores were associated with death of KDIGO stage 1, stage 2, and stage 3 AKI cases were 26.8%,
(Supplemental Table 2). Different multivariate Cox propor- 31.7%, and 41.5%, respectively. Eight patients with AKI re-
tional hazards models demonstrated that, besides male sex quired continuous RRT (CRRT). Most of the KDIGO stage 3
and KDIGO stage 3 AKI, serum IL-6 levels (ng/ml) (HR, AKI cases occurred in the first 4 weeks (Figure 2A). Various
2.12; 95% CI, 1.27 to 3.53; P50.004), higher levels of AKI etiologies were identified: 61.0% (25/41) of the AKIs oc-
D-dimer (HR, 1.57; 95% CI, 1.13 to 2.18; P50.008), and curred either on the same day of septic shock, or within

JASN 31: 2205–2221, 2020 AKI Outcome in Critically Ill COVID-19 2213
CLINICAL RESEARCH www.jasn.org

A 12
KDIGO Stage I
KDIGO Stage II
10 KDIGO Stage III

4
8
Patients

3
6 2
5
4
4 2
4
1 1
2
2 3 3
2 2
1 1
0
1 2 3 4 5 6 7
Weeks

B 12
Other
Drug Adverse Effect
1 Volume Insufficiency
10
Shock
2

8
1 1
2
Patients

1 1
6
3 1 1

4
6
5 5 1
2 4
3
2
1
0
1 2 3 4 5 6 7
Weeks

Figure 2. The compositions of AKI stages and etiologies during the disease course. (A) The proportion of KDIGO stage 1, stage 2, and
stage 3 AKI were 26.8%, 31.7%, and 41.5%, respectively. Most stage 3 AKI occurred in the first 4 weeks. KDIGO stage 1: increase in sCr
to 1.5–1.9 times baseline within 7 days or $0.3 mg/dl ($26.5 mmol/L) increase within 48 hours, or urine output ,0.5 ml/kg per hour for
6–12 hours. KDIGO stage 2: increase in sCr to 2.0–2.9 times baseline within 7 days, or urine output ,0.5 ml/kg per hour for $12 hours.
KDIGO stage 3: increase in sCr to three or more times baseline, or increase in sCr to $4.0 mg/dl ($353.6 mmol/L) within 7 days, or initiation of
RRT, or anuria for $12 hours. (B) Various AKI etiologies were identified: 61.0% (25/41) of the AKIs were considered to be related to septic
shock; eight patients (19.5%) had histories of poor intake or volume loss before AKI; 12.2% (5/41) of AKIs occurred after administration of
antiviral drugs or antibiotics, and no other triggers were identified. In general, AKI caused by septic shock and adverse drug effects occurred
throughout the whole disease course, whereas volume insufficiency usually occurred in the early phase of the disease.

24 hours after septic shock. Four patients with septic shock enteral nutrition support. Five (12.2%) of the AKIs occurred
had AKI complicated with possible rhabdomyolysis (signifi- after the administration of antiviral drugs or antibiotics, and
cant elevated serum myoglobin and lactate dehydrogenase). no other triggers were identified. One patient experienced di-
Eight patients (19.5%) had histories of poor intake or volume abetic ketoacidosis before AKI. One patient who had received
loss (high fever, vomit, diarrhea, or diuresis) before AKI; most kidney transplantation 6 years previously suffered AKI possi-
of these patients (6/8) recovered from AKI quickly after vol- bly caused by acute rejection and cardiac failure. One patient
ume supplementations consisting of isotonic fluids and with AKI had positive anti-phospholipid antibodies and a

2214 JASN JASN 31: 2205–2221, 2020


www.jasn.org CLINICAL RESEARCH

Table 6. Light microscopic pathologic findings of kidney tissues from ten deceased patients critically ill with COVID-19 and
AKI
Sex/ Baseline sCr Peak sCr Disease Glomerular
Patient PMH Tubulointerstitial Changes Vascular Changes
Age (mmol/L) (mmol/L) Course (d) Changes
1 M/62 HTN N/A 427 33 Endothelial CV, focal TA and IF, crystallization Intimal hyperplasia
swollen casts, inflammatory cells
infiltration
2 M/82 DM 115 136 14 NR CV, focal ATI NR
3 M/62 – 73 220 29 NR CV, focal ATI Intimal hyperplasia
4 M/78 HTN 93 197 30 Focal ischemic CV, focal TA and IF Intimal hyperplasia,
sclerosis hyalinosis
5 M/73 HTN, HBV 70 147 24 NR CV, focal TA and IF, focal ATI Intimal hyperplasia,
venous
thrombosis
6 M/59 HTN 48 139 39 Hypertrophy CV, focal ATI Intimal hyperplasia,
hyalinosis
7 M/86 HTN, CAD 82 195 45 NR CV, focal ATI Intimal hyperplasia,
hyalinosis
8 M/66 – 68 121 48 Focal ischemic CV, focal ATI, focal TA and IF Intimal hyperplasia,
sclerosis hyalinosis
9 F/57 – 32 128 36 NR CV, focal ATI NR
10 F/66 DM 80 125 35 NR CV, focal ATI NR
M, male; PMH, past medical history; HTN, hypertension; DM, diabetes Mellitus; CAD, coronary artery disease; HBV, Hepatitis B virus infection; N/A, not available;
CV, cytoplasmic vacuolization; TA, tubular atrophy, IF, interstitial fibrosis; NR, nonremarkable; ATI, acute tubular injuries; F, female.

probable cerebrovascular incident; his renal pathology showed (Figure 4F). Strong ACE2 expression was seen in both parietal
venous thrombosis. In general, AKI caused by septic shock and epithelial cells of glomeruli and tubular epithelial cells
adverse drug effects occurred throughout the disease course, (Figure 5A), which was not significantly different from normal
whereas volume insufficiency usually occurred in the early kidney tissues (Figure 5B). SARS-CoV-2 was not detected by
phase of the disease (Figure 2B). IHC in kidney tissues (Figure 5C).
A few particles of a diameter of about 60–100 nm were
Pathologic Findings in Ten Deceased Patients with observed in the cytoplasm of renal proximal tubular epithelial
COVID-19 and AKI cells under EM (Figure 6A). Some of these particles were en-
Kidney pathologic findings of ten deceased patients with AKI closed in vesicles (Figure 6B). Wrinkling of the glomerular
are summarized in Table 6. The autopsied patients with AKI basement membrane, varying degrees of foot process efface-
had comparable demographic and clinical characteristics with ment, and subendothelial space expansion were present occa-
other patients with AKI (Supplemental Table 3). All of the sionally (Figure 6C). Cytoplasmic vacuolization and remnants
patients showed differing degrees of tubular injury and cyto- of mitochondrial cristae at the periphery were observed in
plasmic vacuolization in tubular epithelial cells (Figure 3A). proximal tubular epithelial cells (Figure 6D). Peritubular cap-
Patient 1 developed anuric AKI requiring CRRTafter receiving illary lumens were obstructed by abundant erythrocytes.
antiviral medications. Crystallizations were observed in the
proximal tubular cells and casts (Figure 3B), which suggested Nucleic Acid Tests of Urine Samples and Kidney Tissues
drug-induced AKI. Localized inflammatory cell infiltration Kidney tissues from nine patients with AKI (except patient 8 in
was present in the interstitial area (Figure 3C). Glomerular Table 6) were sent for nucleic acid RT-PCR tests for SARS-
lesions were not remarkable, except swollen endothelial cells CoV-2, and the results were all negative. The PCR results of
and wrinkling of the glomerular basement membrane with urine samples from another nine patients were also negative.
double contours were occasionally observed (Figure 4, A and The median time from disease onset to these tests was 29.0
B). Arteriolar hyalinosis and intimal fibrosis were observed in (26.0–35.0) days.
several patients (Figure 4, C and D). Patient 5 had high titers
of positive anti-phospholipid antibodies and probable cere-
brovascular incidents before death. His renal pathology re- DISCUSSION
vealed venous thrombosis (Figure 3D).
The IHC staining of CD3, CD4, CD8, CD20, and CD68 COVID-19 is currently a worldwide pandemic affecting .10
displayed scattered distributed lymphocytes and macrophages million people, causing .500,000 deaths. Most previous stud-
in the interstitial area (Figures 3, E–H and 4E). CD3-positive ies were conducted in patients with a short disease course
lymphocytes were also occasionally seen within the glomerulus (median time from illness to death was 16–18.5 days).3,7,9 As

JASN 31: 2205–2221, 2020 AKI Outcome in Critically Ill COVID-19 2215
CLINICAL RESEARCH www.jasn.org

A B

C D

E F

G H

Figure 3. Pathologic findings in kidney tissues from patients critically ill with COVID-19 and AKI. (A) Diffuse tubular injury and cyto-
plasmic vacuolization in tubular epithelial cells (black arrow). (B) Crystallizations in casts and proximal tubular cells in a patient (black
arrows). (C) Localized inflammatory cell infiltration was present in the interstitial area (black arrow). (D) Venous thrombosis in a patient
with positive anti-phospholipid antibodies. (E–G) Scattered distribution of CD3-, CD20-, and CD8-positive lymphocytes in the in-
terstitial area (black arrows). (H) Scattered distribution of CD68-positive macrophages in the interstitial area (black arrows). HE, he-
matoxylin and eosin.

a multidisciplinary medical team that had been taking care of (rate of intubation .80%), with a relatively long disease
patients critically ill with COVID-19 in ICU since early Febru- course (median time from illness to death was 24 days).
ary, we feel obligated to share our experience with the medical Hopefully, our study can provide helpful information to
community. This study described the clinical characteristics of any physicians who are fighting against COVID-19
81 patients who were old (mean age 66.6 years) and critically ill worldwide.

2216 JASN JASN 31: 2205–2221, 2020


www.jasn.org CLINICAL RESEARCH

A B

C D

E F

Figure 4. Pathologic findings in kidney tissues from patients critically ill with COVID-19 and AKI (continued). (A) Glomerulus with
nonremarkable changes. (B) Glomerulus with swollen endothelial cells (white arrow), wrinkling of the glomerular basement membrane
with double contours (black arrows), which was consistent with chronic glomerular ischemia. (C) Arteriolar hyalinosis (black arrow). (D)
Intimal hyperplasia in interlobular artery. (E) CD4-positive cells around a glomerulus. (F) CD3 was occasionally positive within the
glomerulus (black arrows). HE, hematoxylin and eosin; PAS, Periodic acid–Schiff.

Our data showed that, in patients critically ill with COVID- of poor intake and volume loss, and most of them recovered
19 in the ICU, the incidence of AKI was 50.6%, and .40% of from AKI after fluid supplementation. Great attention
these AKIs were KDIGO stage 3. These patients had a very high should be paid to the hemodynamics and volume balance
rate of invasive mechanical ventilation and multiple critical of patients with COVID-19.
complications requiring aggressive management. Pei et al.28 A variety of antiviral drugs are being tested for their efficacy
also reported distinct incidences of AKI between patients non- against COVID-19 worldwide.32,33 All types of antibiotics
critically ill with COVID-19 and those who were critically ill were also used to manage the secondary bacterial infections.
(4.0% versus 42.9%), supporting our findings. These findings In this study, three patients had AKI after receiving antiviral
were also consistent with the incidence of AKI in patients with medications. One of them recovered from AKI after discontin-
non-COVID-19 ARDS (49.0%–68.3%).29,30 uing the suspicious drug, and the other two patients required
AKI is a common complication of patients in ICU set- CRRT support. The pathologic findings of crystallizations in
tings, which can be caused by various conditions.31 Septic the proximal tubular cells and casts highly implies drug-
shock happened in .60% of our patients and was the lead- induced AKI in one patient (Figure 3B). Another two pa-
ing cause for AKI, which was confirmed by the medical his- tients experienced AKI after antibiotic treatment. One of
tory and the pathologic findings of acute tubular injury them received vancomycin, and the other one received
(Figure 3, Table 6). Several patients with AKI had a history piperacillin-sulbactam, which are both well known renal-toxic

JASN 31: 2205–2221, 2020 AKI Outcome in Critically Ill COVID-19 2217
CLINICAL RESEARCH www.jasn.org

A B

C D

Figure 5. Immunochemistry staining of ACE2 and SARS-CoV-2. (A) ACE2 was strongly expressed in both parietal epithelial cells of
glomeruli and tubular epithelial cells (brown area). (B) ACE expression in paracarcinoma normal kidney tissue from a patient with clear
cell renal carcinoma. (C) IHC failed to detect SARS-CoV-2 in kidney tissues of patients with COVID-19. (D) Positive SARS-CoV-2 de-
tection in both alveolar pneumocytes and macrophages in lung tissues (black arrows) of a patient with AKI (patient 5 in Table 6).

antibiotics. These findings indicate that we should carefully se- were more likely caused by ischemia rather than viral
lect treatments and closely monitor the renal functions of pa- infection.
tients with COVID-19. The hypothesis that inflammatory cytokines might con-
Another concern is the possible direct attack by SARS- tribute to the immunologic disorders and multiorgan dys-
CoV-2 to the kidney because of the abundant expression of functions of patients with COVID-19 has been proposed.20,39
ACE2 in kidney tissues (Figure 5A). However, the current ev- We found significantly elevated serum IL-6, lower platelet
idence of SARS-CoV-2 infecting the kidney are not universally count, and prolonged international normalized ratio in pa-
agreed (B. Diao, C. Wang, R. Wang, Z. Feng, Y. Tan, H. Wang, tients with AKI (Tables 1 and 2). Serum IL-6 levels were also
et al.: Human kidney is a target for novel severe acute respi- associated with both AKI and death (Tables 4 and 5). It is
ratory syndrome coronavirus 2 [SARS-CoV-2] infection, believed that rigorous inflammatory reactions could activate
2020; doi:10.1101/2020.03.04.20031120).19,34–36 Our EM ex- the platelets and injure the endothelial cells, leading to clots,
aminations of kidney tissues revealed particles that greatly re- coagulopathy, and microvascular dysfunction.32,40 Antiphos-
semble the structures found in a recent report of virus-like pholipid syndrome was recently reported in COVID-19.41 We
particles.18 However, some pathologists suspected these par- also found one patient with suspected antiphospholipid syn-
ticles were clathrin-coated vesicles or multivesicular bodies drome who presented with AKI and venous thrombosis in
based on the ultrastructural features and virus morphology kidney tissues (Figure 3D). Considering potential correlations
and morphogenesis.22,23,37 Additional tools including immu- between inflammation and organ dysfunctions (Table 5), we
nogold labeling and in situ hybridization might be useful for speculated that inflammation-mediated vascular endothelial
further identification.38 Furthermore, the RT-PCR and IHC injury and a dysregulated immune response might play an
tests of kidney tissues in this study were all negative. A pre- important role in patients critically ill with COVID-19. Blood
vious study suggested the median time of virus shedding was purification therapies targeting inflammatory molecules were
20 days.3 Viral load in tissues other than the lungs were also delivered to several patients and their serologic response
reported to be either negative or much lower.19,36 Considering turned out to be promising.42
the median disease course of the ten patients who were autop- AKI served as a risk factor for mortality of patients with
sied was 34 days (IQR, 28–40), this might account for the lack COVID-19 in Cheng et al.’s12 study. Due to the limited size of
of virus detection in the kidneys. Regardless, the renal patho- samples, different Cox proportional hazards models were
logic changes were predominately acute tubular injury rather tested. The results showed that KDIGO stage 3 AKI was asso-
than glomerulopathy or interstitial nephritis. These changes ciated with death in this group of patients (Table 5).

2218 JASN JASN 31: 2205–2221, 2020


www.jasn.org CLINICAL RESEARCH

A B

C D

Figure 6. EM findings in patients with AKI. (A) EM studies showed small particles (black arrows) in the cytoplasm in renal proximal
tubular epithelium. The diameter of these particles varied from about 60 to 100 nm. (B) Particles in vesicles (black arrows) in proximal
tubular epithelium. (C) Wrinkling of the glomerular basement membrane (black arrow), subendothelial space expansion, and varying
degrees of foot process effacement were present occasionally. (D) Remnants of mitochondrial cristae at the periphery (black arrows)
were observed in proximal tubular epithelial cells.

Supporting this finding, the patients with AKI who died within DISCLOSURES
a shorter disease course also had a faster decline of renal func-
tion. Other potential risk factors for death included male sex, All authors have nothing to disclose.
elevated D-dimer, higher serum IL-6 levels, and higher SOFA
scores, which is consistent with another report.3
Our study does have several limitations. The number of
FUNDING
enrolled patients were limited. A small proportion of the pa-
tients had no baseline sCr value available, which might lead to
This work was supported by National Natural Sciences Foundation of China
misestimation of AKI, although three nephrologists from grant 81970621 (to Y. Qin); Foundation of Tongji Hospital of Tongji Medical
PUMCH reviewed the medical records independently to gen- College, Huazhong University of Science and Technology grant
erate the consensual diagnosis. The tests of tubular functions XXGZBDYJ010 (to G. Wang); and Fundamental Research Funds for the
were lacking and we failed to collect enough data from urine Central Universities grant 3332019029 (to P. Xia).
protein/creatinine results.
In conclusion, we reported the clinicopathologic charac-
teristics and prognosis of AKI in patients critically ill with ACKNOWLEDGMENTS
COVID-19 who had a long disease course. AKI was a com-
mon and multifactorial complication in the late stage of We wish to thank Ms. Cynthia S. Goldsmith, US Centers for Disease Control
and Prevention for her kind help in reviewing the EM images and providing
patients critically ill with COVID-19 in the ICU. Patients
inspiring suggestions. We thank Dr. Xiaoyin Bai, Dr. Siyuan Fan, and Dr. Hua
with AKI were of an older age, had a lower lymphocyte and Zheng from PUMCH for their advice and assistance. We wish to thank all of
platelet count, and a tendency for coagulopathy and hyper- the patients and their family members included in this study.
inflammation. Acute tubular injury was predominant in re- P. Xia, Y. Wen, W. Cao, G. Wang, and Y. Qin designed this study; S. Zhang,
nal pathology. Older age and higher serum IL-6 level were X. Yan, T. Li, B. Du, Z. Liu, X. Zhou, Y. Qin, J. Wang, J. Ma, and P. Xia cared for
associated with AKI. KDIGO stage 3 AKI independently the included patients; P. Xia, J. Ma, Y. Zhou, G. Chen, W. Jiang, and Y. Hu
extracted the clinical data; Y. Duan, S. Xu, H. Cai, H. Wu, Z. Liang, W. Cao, and
predicted death. The early involvement of nephrologists G. Wang performed the light microscope and IHC studies; H. Su and C. Zhang
might improve the outcome of AKI in critical patients performed the EM studies; and Y. Qin, G. Wang, L. Chen, C. Zhang, and X. Li
with COVID-19. critically reviewed the manuscript.

JASN 31: 2205–2221, 2020 AKI Outcome in Critically Ill COVID-19 2219
CLINICAL RESEARCH www.jasn.org

SUPPLEMENTAL MATERIAL 16. Xie C, Jiang L, Huang G, Pu H, Gong B, Lin H, et al: Comparison of
different samples for 2019 novel coronavirus detection by nucleic acid
This article contains the following supplemental material online at http:// amplification tests. Int J Infect Dis 93: 264–267, 2020
jasn.asnjournals.org/lookup/suppl/doi:10.1681/ASN.2020040426/-/ 17. Young BE, Ong SWX, Kalimuddin S, Low JG, Tan SY, Loh J, et al; Sin-
DCSupplemental. gapore 2019 Novel Coronavirus Outbreak Research Team: Epidemio-
Supplemental Table 1. Risk factors for AKI tested by univariate cox pro- logic features and clinical course of patients infected with SARS-CoV-2
portional hazards models. in Singapore. JAMA 323: 1488–1494, 2020
Supplemental Table 2. Risk factors predicting death tested by univariate cox 18. Su H, Yang M, Wan C, Yi LX, Tang F, Zhu HY, et al: Renal histopatho-
proportional hazards model. logical analysis of 26 postmortem findings of patients with COVID-19 in
Supplemental Table 3. The autopsied patients had comparable demo- China. Kidney Int 98: 219–227, 2020
graphic and clinical features with other patients. 19. Puelles VG, Lütgehetmann M, Lindenmeyer MT, Sperhake JP, Wong
Supplemental Figure 1. Compositions of secondary infections. MN, Allweiss L, et al: Multiorgan and renal tropism of SARS-CoV-2
[published online ahead of print May 13, 2020]. N Engl J Med 10.1056/
NEJMc2011400
20. Lin L, Lu L, Cao W, Li T: Hypothesis for potential pathogenesis of SARS-
CoV-2 infection-a review of immune changes in patients with viral
REFERENCES pneumonia. Emerg Microbes Infect 9: 727–732, 2020
21. Yao XH, Li TY, He ZC, Ping YF, Liu HW, Yu SC, et al: A pathological
1. Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, et al; China Novel report of three COVID-19 cases by minimally invasive autopsies.
Coronavirus Investigating and Research Team: A novel coronavirus Zhonghua Bing Li Xue Za Zhi 49: E9, 2020
from patients with pneumonia in China, 2019. N Engl J Med 382: 22. Miller SE, Brealey JK: Visualization of putative coronavirus in kidney.
727–733, 2020 Kidney Int 98: 231–232, 2020
2. World Health Organization: Coronavirus disease (COVID-19) pan- 23. Calomeni E, Satoskar A, Ayoub I, Brodsky S, Rovin BH, Nadasdy T:
demic. Available at: https://www.who.int/emergencies/diseases/ Multivesicular bodies mimicking SARS-CoV-2 in patients without
novel-coronavirus-2019. Accessed July 31, 2020 COVID-19. Kidney Int 98: 233–234, 2020
3. Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, et al.: Clinical course and risk 24. National Health Commission of China: Guideline of management of
factors for mortality of adult inpatients with COVID-19 in Wuhan, China: COVID-19 (5th trial edition). Available at: http://www.nhc.gov.cn/
A retrospective cohort study [ published correction appears in Lancet yzygj/s7653p/202002/3b09b894ac9b4204a79db5b8912d4440.
395: 1038, 2020]. Lancet 395: 1054–1062, 2020 shtml
4. Wang D, Hu B, Hu C, Zhu F, Liu X, Zhang J, et al: Clinical characteristics 25. Khwaja A: KDIGO clinical practice guidelines for acute kidney injury.
of 138 hospitalized patients with 2019 novel coronavirus-infected Nephron Clin Pract 120: c179–c184, 2012
pneumonia in Wuhan, China. JAMA 323: 1061–1069, 2020 26. Shankar-Hari M, Phillips GS, Levy ML, Seymour CW, Liu VX,
5. Wang T, Du Z, Zhu F, Cao Z, An Y, Gao Y, et al: Comorbidities and multi- Deutschman CS, et al; Sepsis Definitions Task Force: Developing a new
organ injuries in the treatment of COVID-19. Lancet 395: e52, 2020 definition and assessing new clinical criteria for septic shock: For the
6. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al: Clinical features of Third International Consensus Definitions for Sepsis and Septic Shock
patients infected with 2019 novel coronavirus in Wuhan, China. Lancet (Sepsis-3). JAMA 315: 775–787, 2016
395: 497–506, 2020 27. Fan E, Brodie D, Slutsky AS: Acute respiratory distress syndrome: Ad-
7. Guan WJ, Ni ZY, Hu Y, Liang WH, Ou CQ, He JX, et al; China Medical vances in diagnosis and treatment. JAMA 319: 698–710, 2018
Treatment Expert Group for Covid-19: Clinical characteristics of coro- 28. Pei G, Zhang Z, Peng J, Liu L, Zhang C, Yu C, et al: Renal involvement
navirus disease 2019 in China. N Engl J Med 382: 1708–1720, 2020 and early prognosis in patients with COVID-19 pneumonia. J Am Soc
8. Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, et al: Clinical course and out- Nephrol 31: 1157–1165, 2020
comes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, 29. Panitchote A, Mehkri O, Hastings A, Hanane T, Demirjian S, Torbic H,
China: A single-centered, retrospective, observational study. Lancet et al: Factors associated with acute kidney injury in acute respiratory
Respir Med 8: 475–481, 2020 distress syndrome. Ann Intensive Care 9: 74, 2019
9. Chen T, Wu D, Chen H, Yan W, Yang D, Chen G, et al: Clinical char- 30. McNicholas BA, Rezoagli E, Pham T, Madotto F, Guiard E, Fanelli V,
acteristics of 113 deceased patients with coronavirus disease 2019: et al; ESICM Trials Group and the Large observational study to UN-
Retrospective study. BMJ 368: m1091, 2020 derstand the Global impact of Severe Acute respiratory FailurE (LUNG
10. Wang L, Li X, Chen H, Yan S, Li D, Li Y, et al: Coronavirus disease 19 SAFE) Investigators: Impact of early acute kidney injury on manage-
infection does not result in acute kidney injury: An analysis of 116 hos- ment and outcome in patients with acute respiratory distress syndrome:
pitalized patients from Wuhan, China. Am J Nephrol 51: 343–348, 2020 A secondary analysis of a multicenter observational study. Crit Care
11. Chen N, Zhou M, Dong X, Qu J, Gong F, Han Y, et al: Epidemiological Med 47: 1216–1225, 2019
and clinical characteristics of 99 cases of 2019 novel coronavirus 31. Griffin BR, Liu KD, Teixeira JP: Critical care nephrology: Core curricu-
pneumonia in Wuhan, China: A descriptive study. Lancet 395: 507–513, lum 2020. Am J Kidney Dis 75: 435–452, 2020
2020 32. Li T, Lu H, Zhang W: Clinical observation and management of COVID-
12. Cheng Y, Luo R, Wang K, Zhang M, Wang Z, Dong L, et al: Kidney 19 patients. Emerg Microbes Infect 9: 687–690, 2020
disease is associated with in-hospital death of patients with COVID-19. 33. Kalil AC: Treating COVID-19–off-label drug use, compassionate
Kidney Int 97: 829–838, 2020 use, and randomized clinical trials during pandemics. JAMA 323:
13. Naicker S, Yang CW, Hwang SJ, Liu BC, Chen JH, Jha V: The novel 1897–1898, 2020
coronavirus 2019 epidemic and kidneys. Kidney Int 97: 824–828, 2020 34. Wang W, Xu Y, Gao R, Lu R, Han K, Wu G, et al: Detection of
14. Hirsch JS, Ng JH, Ross DW, Sharma P, Shah HH, Barnett RL, et al; SARS-CoV-2 in different types of clinical specimens. JAMA 323:
Northwell COVID-19 Research Consortium; Northwell Nephrology 1843–1844, 2020
COVID-19 Research Consortium: Acute kidney injury in patients hos- 35. Pan Y, Zhang D, Yang P, Poon LLM, Wang Q: Viral load of SARS-CoV-2
pitalized with COVID-19. Kidney Int 98: 209–218, 2020 in clinical samples. Lancet Infect Dis 20: 411–412, 2020
15. Ling Y, Xu SB, Lin YX, Tian D, Zhu ZQ, Dai FH, et al: Persistence and 36. Yao XH, He ZC, Li TY, Zhang HR, Wang Y, Mou H, et al: Pathological
clearance of viral RNA in 2019 novel coronavirus disease rehabilitation evidence for residual SARS-CoV-2 in pulmonary tissues of a ready-for-
patients. Chin Med J (Engl) 133: 1039–1043, 2020 discharge patient. Cell Res 30: 541–543, 2020

2220 JASN JASN 31: 2205–2221, 2020


www.jasn.org CLINICAL RESEARCH

37. Goldsmith CS, Miller SE, Martines RB, Bullock HA, Zaki SR: Electron 40. Chanchal S, Mishra A, Singh MK, Ashraf MZ: Understanding in-
microscopy of SARS-CoV-2: A challenging task. Lancet 395: e99, flammatory responses in the manifestation of prothrombotic pheno-
2020 types. Front Cell Dev Biol 8: 73, 2020
38. Goldsmith CS, Tatti KM, Ksiazek TG, Rollin PE, Comer JA, Lee WW, 41. Zhang Y, Xiao M, Zhang S, Xia P, Cao W, Jiang W, et al: Coagulopathy
et al: Ultrastructural characterization of SARS coronavirus. Emerg Infect and antiphospholipid antibodies in patients with Covid-19. N Engl
Dis 10: 320–326, 2004 J Med 382: e38, 2020
39. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ; HLH 42. Ma J, Xia P, Zhou Y, Liu Z, Zhou X, Wang J, et al: Potential effect of
Across Speciality Collaboration, UK: COVID-19: Consider cytokine storm blood purification therapy in reducing cytokine storm as a late com-
syndromes and immunosuppression. Lancet 395: 1033–1034, 2020 plication of critically ill COVID-19. Clin Immunol 214: 108408, 2020

AFFILIATIONS

1
Department of Nephrology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
2
Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
3
Department of Pathology, School of Basic Medical Science, Tongji Medical College, Huazhong University of Science and Technology,
Wuhan, China
4
Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
5
Department of Infectious Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
6
Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
7
Department of Medical ICU, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
8
Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
9
Department of Intensive Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
10
Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences,
Beijing, China
11
Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China

JASN 31: 2205–2221, 2020 AKI Outcome in Critically Ill COVID-19 2221
Supplemental material Contents

Table S1 Risk Factors for AKI Tested by Univariate Cox Proportional Hazards Models

Table S2 Risk Factors Predicting Death Tested by Univariate Cox Proportional Hazards Model

Table S3 The Autopsied AKI Patients Had Comparable Demographic and Clinical Features with Other AKI Patients

Figure S1 Compositions of Secondary Infections


Table S1 Risk Factors for AKI Tested by Univariate Cox Proportional Hazards Models

Univariate Cox proportional hazards model

β RR (95% CI) P

Demographics

Age (per 10 years) 0.529 1.697 (1.238, 2.327) 0.001

Male 0.681 1.977 (0.983, 3.974) 0.056

Past Medical History

Pre-existing Hypertension 0.273 1.314 (0.708, 2.438) 0.387

Diabetes 0.234 1.263 (0.627, 2.545) 0.513

Coronary Heart Disease -0.304 0.738 (0.340, 1.601) 0.442

Symptoms

Fever -0.688 0.502 (0.219, 1.155) 0.105

Anorexia -0.402 0.669 (0.334, 1.338) 0.256

Vomit -0.585 0.557 (0.197, 1.574) 0.269

Diarrhea -0.390 0.677 (0.323, 1.422) 0.303

Lab Tests Findings

Anemia -0.268 0.765 (0.399, 1.464) 0.418

Thrombocytopenia 0.465 1.593 (0.809, 3.134) 0.178

Serum IL-6 (ng/ml)1 0.587 1.798 (1.216, 2.661) 0.003

D-dimer Categories2 0.273 1.314 (0.905, 1.909) 0.151

hsCRP (mg/L)3 0.003 1.003 (0.998, 1.007) 0.252

Complications

Shock 0.455 1.561 (0.801, 3.042) 0.191

Cardiac Injuries 0.090 1.094 (0.504, 2.373) 0.820

Coagulopathy 0.550 1.733 (0.867, 3.466) 0.120

Secondary Infections -0.218 0.804 (0.423, 1.531) 0.507

Glucocorticoid -0.483 0.617 (0.273, 1.396) 0.246

Treatment

Anti-viral medications -0.089 0.915 (0.422, 1.983) 0.822

Antibiotics -0.044 0.957 (0.295, 3.109) 0.942

IVIG -0.542 0.581 (0.267, 1.268) 0.173

Vasopressors 0.480 3.129 (1.220, 8.024) 0.018

IVIG: Intravenous Immunoglobin; hsCRP: high-sensitive C reactive protein; IL-6: interleukin 6


1
64 patients (32 AKIs) were included in the analysis.
2
D-dimer levels were classified into 4 categories: <0.5 as category 1, 0.5-5.0 as category 2, 5.0-21.0 as category 3 and >21.0

as category 4
3
77 patients (39 AKIs) were included in the analysis.
Shock was defined hypotension requiring vasopressor therapy to maintain mean BP 65 mm Hg or greater and having a
serum lactate level greater than 2 mmol/L after adequate fluid resuscitation.
Table S2 Risk Factors Predicting Death Tested by Univariate Cox Proportional Hazards Models

Univariate Cox proportional hazards model

β RR (95% CI) P

Demographics

Age (per 10 years) 0.265 1.303 (1.009, 1.683) 0.042

Male 0.913 2.492 (1.377, 4.507) 0.003

Past Medical History

Pre-existing Hypertension 0.069 1.072 (0.645, 1.780) 0.789

Diabetes -0.146 0.864 (0.456, 1.635) 0.653

Coronary Heart Disease -0.244 0.783 (0.416, 1.476) 0.450

Lab Tests Findings

Leukocytosis 0.618 1.855 (1.019, 3.377) 0.043

Anemia -0.370 0.691 (0.401, 1.191) 0.183

Thrombocytopenia 0.497 1.644 (0.935, 2.889) 0.084

Hypernatremia 0.744 2.103 (1.252, 3.535) 0.005

Serum IL-6 (ng/ml)1 0.543 1.720 (1.238, 2.391) 0.001

D-dimer Categories2 0.791 2.205 (1.357, 3.585) 0.001

hsCRP (mg/L)3 0.001 1.001 (0.998, 1.005) 0.459

Elevated sCr 0.593 1.810 (1.048, 3.125) 0.033

eGFR (ml/min/1.73m2) -0.008 0.992 (0.985, 0.999) 0.034

Complications

ARDS -0.574 0.563 (0.173, 1.835) 0.341

Shock 0.540 1.717 (0.966, 3.050) 0.065

Cardiac Injury 0.494 1.638 (0.806, 3.330) 0.172

Coagulopathy 0.461 1.586 (0.903, 2.785) 0.108

Hypercapnia 0.015 1.015 (0.603, 1.709) 0.956

All AKI 0.365 1.441 (0.864, 2.402) 0.162

KDIGO Stage 3 AKI 0.745 2.107 (1.181, 3.760) 0.012

Clinical Evaluations

APACHE II score 0.037 1.038 (1.006, 1.072) 0.021

SOFA score 0.111 1.118 (1.053, 1.187) <0.001

Treatment
Corticosteroids 0.275 1.316 (0.566, 3.061) 0.524

Anti-viral medications -0.015 0.985 (0.511, 1.898) 0.964

Antibiotics 0.304 1.361 (0.425, 4.360) 0.604

IVIG -0.731 0.481 (0.253, 0.917) 0.026

1
64 patients (56 deaths) were included in the analysis, IL-6 was calculated in ng/ml.
2
D-dimer levels were classified into 4 categories: <0.5 as category 1, 0.5-5.0 as category 2, 5.0-21.0 as category 3 and >21.0

as category 4
3
77 patients (45 deaths) were included in the analysis.
Table S3 The Autopsied AKI Patients Had Comparable Demographic and Clinical Features with Other AKI Patients

Autopsied Patients Other AKI Patients


P Value
(n=10) (n=31)

Age (years) 68.8±10.2 69.9±9.1 0.765

Male : Female 8:2 22:9 0.700

Hypertension 5 (50.0%) 17 (54.8%) 0.655

Diabetes 2 (20.0%) 9 (29.0%) 0.700

Serum Creatinine (μmol/L) 107.0 (56.2-202.8) 104.0 (82.0-171.0) 0.867

eGFR (ml/min/1.73m2) 59.4 (26.6-97.1) 53.0 (32.8-82.5) 0.638

IL-6 (pg/ml) 150.5 (57.7-385.0) 78.8 (32.6-215.4) 0.290

Shock 6 (60.0%) 22 (71.0%) 0.698

Vasopressors 8 (80.0%) 28 (90.3%) 0.580

Corticosteroids 8 (80.0%) 26 (83.9%) 1.000

Anti-viral Medications 8 (80.0%) 25 (80.6%) 1.000

Antibiotics 10 (100.0%) 28 (90.3%) --

IVIG 9 (90.0%) 24 (77.4%) 0.653

IVIG: Intravenous Immunoglobin; IL-6: interleukin 6.


Figure S1 Compositions of Secondary Infections

47 patients experienced 49 episodes of secondary infections. Secondary bacterial pneumonia and blood stream infections were

the most common. Bacterial pneumonia was diagnosed based on the symptom of newly onset fever, yellow sputum and image

changes (Chest X-ray or CT scan). Blood stream infections was diagnosed based on the newly onset fever and positive blood

culture. One patient’s chest CT scan showed pulmonary abscess with cavities and gas-fluids levels inside. One patient who

had fever, signs of peritoneal irritations after several days of diffuse bowel obstruction was diagnosed bacterial peritonitis.

One patient who had newly onset of fever accompanied with cloudy urine and positive urine culture afterward was diagnosed

of urinary tract infection. One patient had fever and signs of cellulitis on her left foot. All these infections happened after the

hospital presentation.

Das könnte Ihnen auch gefallen