Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Nephrol Dial Transplant, 2024, 39, 26–35

https://doi.org/10.1093/ndt/gfad142
Advance access publication date: 3 July 2023

Sepsis-associated acute kidney injury—treatment


TREATMENT STANDARD

standard
Alexander Zarbock1 , Jay L. Koyner 2
, Hernando Gomez3 , Peter Pickkers4 and Lui Forni5 ,6 ; on behalf of the Acute Disease Quality
Initiative group
1
Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital of Münster, Münster, Germany and Outcomes Research Consortium,
Cleveland, OH, USA
2
Section of Nephrology, Department of Medicine, University of Chicago, Chicago, IL, USA
3
Program for Critical Care Nephrology, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA

Downloaded from https://academic.oup.com/ndt/article/39/1/26/7218568 by guest on 25 December 2023


4
Department Intensive Care Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
5
Department of Critical Care, Royal Surrey Hospital Foundation Trust, Guildford, UK
6
Faculty of Health Sciences, University of Surrey, Guildford, UK
Correspondence to: Alexander Zarbock; E-mail: zarbock@uni-muenster.de

ABSTRACT
Sepsis is a host’s deleterious response to infection, which could lead to life-threatening organ dysfunction. Sepsis-associated acute
kidney injury (SA-AKI) is the most frequent organ dysfunction and is associated with increased morbidity and mortality. Sepsis con-
tributes to ≈50% of all AKI in critically ill adult patients. A growing body of evidence has unveiled key aspects of the clinical risk
factors, pathobiology, response to treatment and elements of renal recovery that have advanced our ability to detect, prevent and
treat SA-AKI. Despite these advancements, SA-AKI remains a critical clinical condition and a major health burden, and further stud-
ies are needed to diminish the short and long-term consequences of SA-AKI. We review the current treatment standards and discuss
novel developments in the pathophysiology, diagnosis, outcome prediction and management of SA-AKI.

Keywords: acute kidney injury, biomarker, renal replacement therapy, sepsis

IN A NUTSHELL

1. The pathophysiology of sepsis-associated acute kidney injury (SA-AKI) is complex and incompletely understood.
2. Biomarkers can identify patients at high risk for AKI and predict short- and long-term adverse outcomes.
3. Implementing supportive measures (e.g. haemodynamic optimization and avoidance/minimizing nephrotoxins) in septic pa-
tients at high risk for AKI might prevent AKI development, but no specific therapies are available to treat sepsis-associated
AKI.
4. Renal replacement therapy (RRT) should be initiated in patients with sepsis who develop medically refractory complications
or persistent AKI, however, a strategy or early pre-emptive RRT is not associated with improved outcomes.
5. Preliminary data have suggested that selective subphenotypes of SA-AKI may have a better response to specific vasoactive
drugs (e.g. arginine vasopressin, angiopoietin-2), however, further confirmatory data are needed.

INTRODUCTION Here we discuss the most recent evidence supporting our cur-
rent understanding of the pathophysiology and outcome predic-
Sepsis is characterized by organ dysfunction as a consequence of
tion in patients with SA-AKI. Furthermore, based on the recent
a dysregulated response to infection. Septic shock is character-
recommendations of the Acute Disease Quality Initiative (ADQI)
ized by persistent hypotension despite adequate fluid resuscita-
group [5], we have developed a pragmatic diagnostic and treat-
tion, the need for vasoactive drugs to maintain a mean arterial
ment algorithm for SA-AKI in adults.
pressure (MAP) of at least 65 mmHg and a plasma lactate concen-
tration >2 mmol/L [1].
Up to 60% of patients with sepsis develop acute kidney in- TREATMENT STANDARDS
jury (AKI) [2]. Although the pathophysiology underlying sepsis-
From diagnosis to treatment of SA-AKI
associated AKI (SA-AKI) remains incompletely understood, mul-
Where sepsis is suspected, lactate should be measured, and if el-
tiple mechanisms, including inflammation, complement activa-
evated (>2 mmol/L), measurement should be repeated, with re-
tion, mitochondrial dysfunction and microcirculatory dysfunc-
suscitation guided by lactate levels. Once sepsis is diagnosed, the
tion, are thought to contribute to AKI development [3]. The de-
patient should be treated according to current sepsis guidelines
velopment of AKI carries a significantly increased mortality risk
[1], and before administering antibiotics, blood cultures should be
compared w sepsis alone [4].

Received: May 9, 2023; Editorial decision: June 5, 2023


© The Author(s) 2023. Published by Oxford University Press on behalf of the ERA.
All rights reserved. For permissions, please e-mail: journals.permissions@oup.com
A. Zarbock et al. | 27

Downloaded from https://academic.oup.com/ndt/article/39/1/26/7218568 by guest on 25 December 2023


Figure 1: Flow chart of the diagnostic and treatment algorithm.

obtained (Fig. 1). In the presence of sepsis-induced hypoperfusion All patients with sepsis should be considered at high risk for
(lactate levels ≥2 mmol/L) or septic shock, crystalloid fluid admin- AKI, whereas patients with certain comorbidities, such as chronic
istration should be initiated and guided based on dynamic param- kidney disease, have an even higher risk to develop AKI un-
eters of fluid responsiveness, along with vasopressors to keep the der these circumstances. Kidney function should be monitored
MAP >65 mmHg and decrease serum lactate levels. The recom- closely using serum creatinine and urine output for earlier de-
mended first-line vasopressor in septic patients is norepinephrine, tection and staging according to the Kidney Disease: Improving
as dopamine should not be used in this patient group [1]. Global Outcomes (KDIGO) guidelines [10]. However, the KDIGO
Early fluid administration is important to potentially rescue definition of AKI has several limitations. For example, a base-
the macro- and microcirculation in sepsis. The guidelines recom- line serum creatinine value is necessary to establish an increase,
mend an initial 30 ml/kg of crystalloids without adjusting this and no consensus method exists to establish a pre-AKI baseline
volume to the current fluid status of the patient [1]. As volume serum creatinine in the absence of direct data [11]. Furthermore,
overload is associated with a worse outcome [6], fluid admin- serum creatinine changes are often delayed, particularly in sep-
istration should be individualized and guided by fluid respon- sis, where the creatinine production rate may be reduced by 50%
siveness. The type of fluid used for resuscitation has resulted in and fluid resuscitation may dilute the creatinine concentration
much study and debate, with earlier evidence suggesting that [12]. Although integral to the AKI definition, urine output is non-
0.9% saline may cause hyperchloremia, resulting in AKI and worse specific and is measured accurately only in patients with an in-
patient-centred outcomes [7]. However, more recent studies have dwelling urinary catheter. Interestingly, several observational co-
demonstrated that saline may be used safely without untoward hort studies show that the same stage of AKI diagnosed by either
effects [8]. Given the contradictory evidence, this issue remains serum creatinine or urine output may confer a differential risk of
an area of contention, but based on the current evidence, the use morbidity and mortality [13, 14].
of up to 4 L of saline seems to be safe in regard to patient-centred Early detection of kidney damage is essential, as it may
outcomes [9]. increase the opportunity for successful interventions [15, 16].
28 | Nephrol Dial Transplant, 2024, Vol. 39, No. 1

Downloaded from https://academic.oup.com/ndt/article/39/1/26/7218568 by guest on 25 December 2023


Figure 2: Optimization of the volume and haemodynamic status. During the optimization, patients will receive functional haemodynamic monitoring
and optimization according to a haemodynamic algorithm: Performance of the passive leg raising test (PLRT). If the cardiac output (CO) increases by
>10%, then volume has to be given. If CO is ≤10%, then proceed with measurement of the cardiac index (CI). If the CI is <2.5 L/min/m2 , then
dobutamine or epinephrine needs to be applied. If the CI is >2.5 L/min/m2 , then proceed with MAP measurement. If the MAP is <65 mmHg, then
norepinephrine needs to be adjusted. If MAP is >65 mmHg, then the goal is achieved. This is checked every 6 hours on the first day and then twice a
day.

Combining damage and functional biomarkers to increase the epithelial cells compared with non-septic AKI. A small trial
sensitivity and specificity of AKI definitions may help [17]. demonstrated that a urinalysis score >3 was predictive of severe
Stage 1S (‘subclinical AKI’), for example, is defined by in- AKI and significantly correlated with biomarkers of tubular injury
creased stress/injury biomarker levels despite normal functional [27].
biomarkers (serum creatinine and urine output). In addition, data
from the Protocolized Care for Early Septic Shock (ProCESS) co- Patients at high risk for SA-AKI
hort demonstrated that biomarker-positive AKI is associated with
In patients at high risk for AKI, the KDIGO guidelines recommend
lower 30-day survival than biomarker-negative AKI despite the
implementing a care bundle of supportive measures [10] (see also
same functional stage of AKI defined by KDIGO criteria [18].
Fig. 2). Indeed, it has been demonstrated that implementing sup-
The concentrations of several serum biomarkers inversely cor-
portive measures in surgical patients at risk can reduce the oc-
relate with glomerular filtration rate (GFR) and may provide an
currence of AKI [15, 16, 28]. However, this has not been explicitly
advantage in detecting SA-AKI. Cystatin C and proenkephalin in-
shown in patients with sepsis. Moreover, despite these measures
crease earlier than serum creatinine in critically ill patients with
being part of routine clinical care, only a minority of patients re-
sepsis [19, 20]. Urinary tissue inhibitor of metalloproteinase-2
ceive all the guideline-recommended measures [28–31]. The bun-
(TIMP2) and insulin-like growth factor binding protein-7 (IGFBP7),
dle that has been shown to reduce AKI in patients at risk for AKI
two markers associated with cell cycle arrest, have very good per-
[15, 16] could be implemented in septic patients at high risk of
formance in predicting stage 2 or 3 AKI in critically ill patients
AKI and is outlined in Table 1. It includes avoidance of potentially
with sepsis (area under the curve 0.84) [21]. Plasma neutrophil
nephrotoxic agents (e.g. hydroxyethyl starch), close monitoring of
gelatinase-associated lipocalin has been demonstrated to be ele-
serum creatinine and urine output, avoidance of hyperglycaemia,
vated in septic patients, even in the absence of creatinine or urine
consideration of alternatives to radiocontrast agents and haemo-
output–based AKI. A higher cut-off value (454 ng/ml) provides a
dynamic monitoring with optimization of both volume status and
specificity of 74% and a sensitivity of 72% for detecting AKI [22].
haemodynamics.
Bedside Doppler ultrasound is a rapid, non-invasive and re-
peatable tool for evaluating renal perfusion and is widely used in
critically ill patients [23]. The Doppler-based renal resistive index Patients with SA-AKI
(RRI) changes before changes in serum creatinine occur during The supportive measures should be continued in patients with an
the development of AKI, and RRI has been shown to predict AKI established AKI. However, no evidence shows that the bundle can
[24]. However, it has been shown that renal Doppler does not positively influence the course of AKI.
discriminate between patterns of renal recovery nor modifies risk
stratification for AKI persistence [25]. In addition, several phys- Diuretics
iological factors, such as intra-abdominal pressure and vascular The prophylactic use of diuretics in patients with a high risk of AKI
compliance, influence RRI. Urinalysis and urine microscopy may has been shown to be unsuccessful in critically ill patients [32].
contribute to the identification of SA-AKI. Several observational Similarly, diuretics do not attenuate AKI once it is established [33].
studies evaluated a urine microscopy score in a cohort of SA-AKI Therefore, the use of diuretics for the prevention or treatment of
[26, 27]. SA-AKI shows more cast elements and renal tubule SA-AKI is not recommended. However, their use in regulating fluid
A. Zarbock et al. | 29

Table 1: Supportive measures implemented in patients at high risk for AKI.

Supportive measures Rationale

Discontinue all nephrotoxic agents when possible Nephrotoxic drugs contribute to AKI in 20–30% of patients. Agents such as antimicrobials
are used in patients already at high risk for AKI (e.g. septic patients), thus it is often
difficult to discern exactly what contribution these agents have on the overall course of
AKI. Nevertheless, limiting exposure whenever possible and weighing the risk of
developing or worsening AKI against the risk associated with not using the agent is
prudent. Recently, consensus has been obtained for the nephrotoxic potential of various
drugs in critically ill patients [65]. Nephrotoxic stewardship strategies will assist in
reducing adverse drug events and optimizing kidney health.
Optimize volume status and perfusion pressure (Fig. 2) Ensuring adequate perfusion pressure and avoiding hypo- or hypervolaemia are crucial
factors for preventing and treating AKI. In addition, fluid management should be guided
by repeated evaluations of fluid status and volume responsiveness, given a higher
cumulative positive fluid balance is associated with AKI and the need for RRT. Balanced
crystalloid solutions are the first choice for fluid replacement and are preferred over

Downloaded from https://academic.oup.com/ndt/article/39/1/26/7218568 by guest on 25 December 2023


chloride-rich solutions and synthetic colloids. Given the potentially harmful effects of
synthetic colloids, the use of albumin has been raised as a potentially preferable
alternative. However, to date, no high-quality studies have shown significant benefit to
albumin-containing regimens, so their use can only be recommended in patients who
have received large volumes of crystalloids.
Although the optimum haemodynamic targets are yet to be defined, a MAP >65 mmHg is
usually considered sufficient to maintain organ perfusion. MAP targets >65 mmHg
recommended in sepsis guidelines diminish the RRT rate in patients with pre-existing
hypertension; however, this did not translate into improved survival [35].
Norepinephrine and vasopressin remain first-line agents for the treatment of septic shock.
Venous congestion also plays a pivotal role in the development of SA-AKI. An elevated
central venous pressure (CVP) increases renal venous pressure and subsequently
decreases renal blood flow and GFR. Higher CVP in the first 24 hours of ICU admission
with septic shock was associated with an increased risk for development or persistence of
AKI over the next 5 days [66].
Consider functional haemodynamic monitoring A comprehensive haemodynamic workup is completed by preload and cardiac output
measurements since a low cardiac output state with decreased systemic oxygen delivery
is strongly associated with the development of organ dysfunction. Optimization of stroke
volume and preservation of the cardiac index >3 L/min/m2 was identified to be among the
most important measures to prevent AKI [67].
Monitor serum creatinine and urine output Close monitoring of renal function based on serum creatinine and urine output is essential
to establish an early diagnosis, classify the severity and predict the outcome of AKI.
Intensive urine output monitoring is associated with increased detection of AKI and
decreased mortality at day 30 [hazard ratio 0.85 (95% confidence interval 0.77–0.94);
P = .001], likely due to improved fluid management and less cumulative volume overload
[13].
Avoid hyperglycaemia High blood glucose levels induce inflammation, apoptosis and fibrosis pathways, cause
oxidative stress and volume depletion by osmotic diuresis and are generally
well-recognized for their contribution to kidney disease. The potential benefits of tight
glycaemic control have to be balanced against the risk of hypoglycaemia. The 2012 KDIGO
guideline recommends an intermediate corridor between tight and conventional
glycaemic control, targeting plasma glucose of 110–149 mg/dl (6.1–8.3 mmol/L) [10].
Consider alternatives to radiocontrast procedures A comparable dilemma applies to the use of iodinated radiocontrast agents and the
subsequent development of AKI. Critically ill patients with an inherently increased risk for
AKI are more likely to undergo contrast-enhanced diagnostic procedures involving
radiocontrast agents. However, if required for source detection, the use of contrast media
should not be deferred because of potential risk, which has been shown to be low in
critically ill patients [10]. Almost all studies investigating the effect of contrast agent on
the development of AKI have not shown an increased risk of AKI after contrast exposure.
Therefore the minimal risk of contrast-induced AKI has to be weighed against the benefit
of contrast-enhanced CT, especially in sepsis [68, 69].

balance and preventing fluid overload fosters their continued use metabolic acidaemia (pH <7.20) in critically ill patients with
in critical illness despite their inability to improve renal outcomes. moderate–severe AKI (Acute Kidney Injury Network stage 2 or 3)
reduced the primary composite outcome (death from any cause
Bicarbonate by day 28 and the presence of at least one organ failure at day
The subgroup analysis of the BICAR-ICU trial, a multicentre, 7) and 28-day mortality [34]. However, 24% of the control arm
open-label, controlled phase 3 clinical trial, demonstrated that received bicarbonate therapy, and these observations may be
treatment with intravenous 4.2% sodium bicarbonate for severe subject to considerable type 1 error. Also, only 60% of the trial
30 | Nephrol Dial Transplant, 2024, Vol. 39, No. 1

population had sepsis at randomization. Therefore the results Local innate immune activation, inflammation
cannot be fully extrapolated to patients with SA-AKI. and complement activation
The early interaction of damage and pathogen-associated molec-
Blood pressure and vasopressors ular patterns (DAMPs and PAMPs) with the host’s pattern recog-
Although the arterial blood pressure target in septic patients nition receptors (PRRs) induces the activation of immunity and
is unknown, one trial demonstrated that targeting a MAP complement pathways, leading to a systemic and local renal in-
>85 mmHg in patients with pre-existing hypertension reduced flammatory response. DAMPs and PAMPs can be filtered through
the serum creatinine increase and the need for renal replace- the glomerulus and engage tubular epithelial cell toll-like recep-
ment therapy (RRT) [35]. Angiotensin II, an endogenous vasocon- tors (i.e. TLR4). In the tubular system, TLR4 engagement triggers
strictor, preferentially constricts the glomerular efferent arteriole, an oxidative stress outburst [50]. PAMPs and DAMPs can also bind
thus increasing the GFR. A post hoc analysis of the ATHOS-3 trial to PRRs and promote local inflammation [51].
showed that patients with vasodilatory shock on RRT random-
ized to angiotensin II discontinued RRT earlier (38% versus 15% by Microvascular dysfunction, oxidative stress, amplification of
day 7, P = .007) and had lower 28-day mortality (30% versus 53%, the inflammatory response, intrarenal shunting and tissue
P = .012) compared with placebo [36]. Data from a post hoc anal- hypoxia

Downloaded from https://academic.oup.com/ndt/article/39/1/26/7218568 by guest on 25 December 2023


ysis suggest that patients with low angiotensin II and high renin Sepsis is known to cause widespread, systemic microvascular dys-
levels benefit from this drug [37, 38]. However, further validation function, manifested as a decrease in the proportion of capillaries
of these findings data is needed. with continuous flow, an increase in the proportion of capillaries
with intermittent or no flow and an increase in the heterogeneity
RRT of blood flow distribution [52]. This also occurs at the level of the
Several recent studies have investigated different aspects of RRT peritubular microvasculature before changes in renal blood flow
in the critically ill [39–42], specifically in SA-AKI patients. occur and creatinine increases [53]. In an animal sepsis model,
Data around the timing of RRT in SA-AKI suggest that there oxidative stress represented by increased production of reactive
is no benefit from accelerated RRT initiation [43]. One multi- oxygen and nitrogen species occurred in tubular epithelial cells
centre randomized trial in SA-AKI was stopped early for futility, adjacent to peritubular capillaries with intermittent or stop flow,
with mortality rates of 58% (138/239) in the early group and 54% suggesting a link between these two injury mechanisms [53]. Slug-
(128/238) in the delayed group [44]. However, 93 (38%) patients in gish peritubular flow can also amplify the inflammatory response
the delayed arm never received RRT. by increasing the exposure time of neighbouring epithelial cells
Regarding RRT dosing, the evidence supports the recom- to activated, cytokine-spilling leucocytes transiting through the
mended delivery dose of 20–25 ml/kg/h [40, 41], partly based peritubular capillaries [54]. This inflammatory amplification may
on studies demonstrating no benefit with higher doses or high- explain the association between capillary dysfunction and oxida-
volume haemofiltration [45–47]. tive stress in the tubular epithelium. A more direct consequence
In the setting of SA-AKI, large RCTs and meta-analyses have of microvascular dysfunction may be intrarenal shunting, lead-
demonstrated no difference in outcomes between intermittent ing to the development of areas of tissue hypoxia and to tubular
versus continuous RRT [48, 49]. However, it might be possible that injury.
haemodynamically unstable patients benefit more from contin-
uous RRT compared with intermittent RRT. Therefore, clinicians RAAS dysfunction
should start the RRT modality they are most familiar with and The RAAS modulates vascular tone, fluid and electrolyte bal-
that achieves a delivered dose of 20–25 ml/kg/h for continuous ance and GFR [55]. In critically ill patients, elevated renin levels
RRT or a Kt/V of 3.9 per week for intermittent dialysis. are associated with worse survival, major adverse kidney events
(MAKEs) and the need for RRT [56]. In addition, elevated renin
Pathophysiology levels are usually found in patients with refractory vasodila-
It is likely that SA-AKI is caused by multiple pathophysiologic tory shock [56]. Notably, the inflammatory response can cause
mechanisms that interact with host background susceptibility angiotensin-converting enzyme dysfunction, leading to a deficit
factors, the host’s capacity to withstand tissue injury or tolerance in angiotensin II [55].
capacity, direct effects related to sepsis and the effect of sepsis-
associated therapies. Future trials to test therapeutic interven- Mitochondrial dysfunction
tions to alleviate SA-AKI will need to embrace this complexity and The tubular epithelium of the proximal tubules has the daunting,
likely rely on biomarkers to select patients in whom targeting par- energetically expensive task of reabsorbing ≈70% of the sodium
ticular processes is relevant. contained in the ≈170 L/day of fluid filtered by the glomeru-
A specific pathophysiologic mechanism proved to induce tubu- lus. Unsurprisingly, the proximal tubule and thick ascending limb
lar injury, therefore AKI is defined as an endotype. In sepsis, of the loop of Henle are only second to the heart in mitochon-
rather than a single endotype, multiple endotypes likely in- drial density. Therefore, mitochondrial well-being is critical to
teract and cause injury, thereby inducing SA-AKI. Mechanisms tubular and renal function. Sepsis induces mitochondrial injury
such as local inflammation, innate immune activation, mitochon- through oxidative damage [57], TLR4-mediated inflammation [58]
drial and microvascular dysfunction, oxidative stress, hypoxia, and electron transport chain inhibition [59]. It is characterized
renin–angiotensin–aldosterone system (RAAS) dysfunction, com- by decreased mitochondrial mass, disruption of cristae and vari-
plement activation and metabolic reprogramming are all sup- able mitochondrial swelling [60]. In addition, sepsis impairs mi-
ported by the literature as prominent endotype candidates to ex- tochondrial quality control mechanisms like fission, fusion and
plain the underpinnings of SA-AKI. The evidence supporting the mitophagy, which are critical for protecting and rescuing dysfunc-
pre-eminence of these mechanisms is discussed next. tional mitochondria [60].
A. Zarbock et al. | 31

Outcome prediction FUNDING


As discussed above, AKI biomarkers have been used to improve This work was supported by the German Research Foundation (ZA
early risk stratification of patients prone to or already with SA- 428/18-2, ZA 428/26-0, ZA 428/21-1 and KFO 342/2 to AZ).
AKI. In addition, several studies have demonstrated that blood
and urinary biomarkers measured early in the clinical course of
sepsis can predict patient-oriented outcomes like AKI progression, AUTHORS’ CONTRIBUTIONS
the need for RRT and mortality.
All authors made equal contributions to the discussion of the
In samples from >2500 patients with sepsis and septic shock
content. A.Z., J.L.K, H.G., P.P. and L.G.F drafted and edited the
from the FROG-ICU and ADRENOSS studies, Dépret et al. [61]
manuscript. All authors reviewed the final manuscript before sub-
showed that in the absence of changes in serum creatinine
mission.
and urine output, an elevated proenkephalin level (>80 pmol/L)
at the time of intensive care unit (ICU) arrival was associated
with an increased risk of inpatient mortality. While this elevated DATA AVAILABILITY STATEMENT
biomarker in the absence of changes in traditional AKI mark-
No new data were generated or analysed in support of this re-
ers was only present in 6–7% of the population, the associa-

Downloaded from https://academic.oup.com/ndt/article/39/1/26/7218568 by guest on 25 December 2023


search.
tion between proenkephalin and mortality persisted after ad-
justing for age, sex, comorbidities, ICU diagnosis, creatinine and
diuresis.
CONFLICT OF INTEREST STATEMENT
In a planned substudy of the ProCESS trial (an early goal-
directed therapy in sepsis randomized control study), pa- A.Z. has received consulting fees from Astute-bioMérieux, Baxter,
tients who had persistently elevated TIMP2•IGFBP7 levels Bayer, Novartis, Guard Therapeutics, AM Pharma, Paion and Fre-
[>0.3 [ng/ml]2 /1000) after the first 6 hours of resuscitation with senius Medical Care; research funding from Astute-bioMérieux,
fluids and vasopressors were at higher risk for a composite end- Fresenius Medical Care and Baxter and speaker fees from Astute-
point of progression to severe AKI (stage 2 or 3), receipt of dialysis bioMérieux, Fresenius Medical Care and Baxter. P.P. has received
and mortality [62]. Moreover, the incidence of this composite speaker, travel and consultancy fees as a member of an advisory
endpoint was similar in patients with post-resuscitation elevated board or steering committee for Baxter, EBI, AM-Pharma, Sph-
biomarkers regardless of their pre-resuscitation biomarker sta- ingotec, Adrenomed, 4Teen4 and Paion. H.G. serves as a scien-
tus. Similarly, elevated TIMP2•IGFBP7 levels were associated with tific advisor for Novartis and Trilinear BioVentures and has re-
an increased incidence of this composite endpoint regardless of ceived research grants from bioMérieux, Baxter and TES Pharma.
AKI status based on serum creatinine and urine output [62]. M.J. has received honoraria and research support from Baxter
While these studies illustrate that a single biomarker can be Healthcare, AM-Pharma, CLS Behring, Fresenius Medical Care
used to risk-stratify patients with sepsis early in their clinical and Novartis. K.K. has received research grants from Philips Re-
course, increasingly investigators are using multiple biomarkers search North America and Google, speaker fees from Nikkiso
and advanced learning techniques (e.g. machine learning) to iden- Critical Care Medical Supplies (Shanghai) and funding from the
tify patients at risk for SA-AKI and adverse patient outcomes [5]. National Institute of Diabetes and Digestive and Kidney Dis-
In a recent study, four unique sepsis phenotypes were validated eases (grant R01DK131586) and consulting fees from Baxter (to
using two large retrospective clinical cohorts. These four pheno- the Mayo Clinic). J.L.K. has received consulting fees from Astute-
types had their unique cytokine/biomarker profiles related to dif- bioMérieux, Sphingotec, Pfizer, Baxter, Mallinckrodt, Novartis and
ferent aspects of sepsis (AKI, inflammation, coagulopathy and en- Guard Therapeutics; research funding from Astute-bioMérieux,
dothelial dysfunction) as well as their clinical characteristics [pa- BioPorto, NxStage Medical, Fresenius Medical Care and Satellite
tient demographics, baseline comorbidities and therapeutic expo- Healthcare and speaker fees from NxStage Medical. M.M. has re-
sures (e.g. the need for vasoactive medications)] [63]. Others have ceived lecture fees from bioMérieux, Fresenius Medical Care and
applied similar approaches to large data sets to identify septic pa- Baxter. T. Reis has received funding for lectures and has been a
tients at risk for AKI and death [64]. While these phenotypes are consultant or advisory board member for AstraZeneca, B. Braun,
increasingly more prevalent in the literature, their clinical utility Baxter, bioMeŕieux, Boehringer Ingelheim, Contatti Medical (Cy-
will depend heavily on demonstration that they can be identified toSorbents), Eurofarma, Fresenius Medical Care, Jafron Biomedi-
early and efficiently (i.e. using biomarkers) or that they are linked cal, Lifepharma and Nova Biomedical. T. Rimmelé serves as a sci-
to an underlying pathophysiologic process that can be targeted or entific advisor for Jafron Biomedical and Exthera; has received
with a clinically relevant response to a specific therapeutic inter- funding for lectures from B. Braun, Baxter, bioMeŕieux, Exthera,
vention. Fresenius Medical Care, Estor and Jafron Biomedical and has re-
ceived research grants from Baxter and Fresenius Medical Care.
S.M.B. is supported by a Canada Research Chair in Critical Care
Outcomes and Systems Evaluation and has received fees for sci-
entific advisory from Baxter, Novartis, Sea Star Medical, BioPorto
CONCLUSIONS and SphingoTec. R.B. has received grant money, speaker fees and
The pathophysiology of SA-AKI is very complex and still not fully advisory board fees from Baxter Acute Care, Jafron Biomedical,
understood. Currently, no specific therapy exists to prevent or CSL Behring, AM-Pharma, and Paion. V.C. has received lecture
treat this complex syndrome. Management of SA-AKI involves fees from Baxter, Estor-Toray and Aferetica-Cytosorbents. S.L.K.-
early recognition and treatment of the underlying infection, fluid G. is an elected member of the Executive Committee (or Coun-
resuscitation, inotropic or vasopressor agents, diuretics and po- cil) of the Society of Critical Care Medicine (SCCM). The views
tentially RRT. Prevention is key and includes early recognition and presented are those of the author and do not represent the
treatment of sepsis, avoidance of nephrotoxic agents and appro- views of the SCCM. R.L.M. has consulting/advisory relationships
priate dosing of medications. with Baxter, AM-Pharma, bioMeŕieux, Intercept, Mallinckrodt, GE
32 | Nephrol Dial Transplant, 2024, Vol. 39, No. 1

Healthcare, Medtronic, CHF Solutions, Sphingotec, Abiomed, Nova Kane-Gill (Department of Pharmacy and Therapeutics, School
Biomed, Sanofi, Renasym, Alexion, Fresenius Medical Care, Ab- of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA),
bott and Renibus Therapeutics. P.T.M. serves as a scientific advi- Yvelynne Kelly (Department of Critical Care, Tallaght University
sor for AM-Pharma, Novartis and Renibus Therapeutics. M.O. has Hospital, Tallaght, Dublin, Ireland; School of Medicine, Trinity
received speaker fees from Fresenius Medical Care, Baxter and College Dublin, Dublin, Ireland), Ravindra L. Mehta (Department
bioMeŕieux and research funding from Fresenius Medical Care, of Medicine, University of California San Diego, La Jolla, CA,
Baxter and bioMeŕieux. J.R.P. has received research support from USA), Patrick T. Murray (School of Medicine, University College
Jafron Biomedical and bioMérieux and consultancy or lecture fees Dublin, Dublin, Ireland), Marlies Ostermann (Department of In-
from Baxter, Nikkiso Europe, Mission Therapeutics and Paion UK. tensive Care, King’s College London, Guy’s & St Thomas’ Hospital,
A.S. has received speaker and/or consulting fees from Fresenius London, UK), John Prowle (William Harvey Research Institute,
Medical Care, CytoSorbents, Jafron Biomedical, Medtronics and B. Faculty of Medicine and Dentistry, Queen Mary University of
Braun Avitum and has received grants from the Leenaards Foun- London, London, UK), Zaccaria Ricci (Department of Anesthesia
dation and B. Braun Melsungen. A.T. has received consulting fees and Critical Care, Meyer Children’s University Hospital, Florence,
from Baxter and royalties from a 0.5% citrate patent from Baxter. Italy; Department of Health Sciences, Section of Anesthesiology
G.V. has received lecture fees from Baxter. C.R. has been on the and Intensive Care, University of Florence, Florence, Italy), Emily

Downloaded from https://academic.oup.com/ndt/article/39/1/26/7218568 by guest on 25 December 2023


advisory boards or speaker’s bureau for Asahi, Aferetica, Baxter, J. See (Department of Critical Care, University of Melbourne,
bioMérieux, Cytosorbents, B. Braun, GE, Medica, Medtronic, Jafron Parkville, Victoria, Australia; Department of Intensive Care, Royal
Biomedical and AstraZeneca. L.G.F. has received research support Melbourne Hospital, Melbourne, Victoria, Australia; Department
and lecture fees from Ortho Clinical Diagnostics, Baxter, Exthera of Nephrology, Royal Melbourne Hospital, Melbourne, Victoria,
and bioMérieux and consulting fees from La Jolla Pharmaceuticals Australia), Antoine Schneider (Adult Intensive Care Unit, Cen-
and Paion. The remaining authors declare no conflicts of interest. tre Hospitalier Universitaire Vaudois, Lausanne, Switzerland),
Danielle E. Soranno (Department of Pediatrics, Indiana Univer-
sity School of Medicine, Indianapolis, IN, USA), Ashita Tolwani
APPENDIX (Department of Medicine, University of Alabama at Birmingham,
ADQI group: Mitra K. Nadim (Division of Nephrology and Hy- Birmingham, AL, USA), Gianluca Villa (Department of Health
pertension, Department of Medicine, Keck School of Medicine, Sciences, Section of Anesthesiology, Intensive Care and Pain
University of Southern California, Los Angeles, CA, USA), Samira Medicine, University of Florence, Azienda Ospedaliero Univer-
Bell (Division of Population Health and Genomics, University of sitaria Careggi, Florence, Italy), Claudio Ronco (Department of
Dundee, Dundee, UK), Michael Joannidis (Division of Intensive Medicine, University of Padova, Padua, Italy; International Renal
Care and Emergency Medicine, Department of Internal Medicine, Research Institute of Vicenza, Vicenza, Italy; Department of
Medical University Innsbruck, Austria), Kianoush Kashani (Di- Nephrology, San Bortolo Hospital, Vicenza, Italy).
vision of Nephrology and Hypertension, Division of Pulmonary
and Critical Care Medicine, Department of Medicine, Mayo Clinic,
Rochester, MN, USA), Neesh Pannu (Department of Medicine, Uni-
versity of Alberta, Edmonton, Alberta, Canada), Melanie Meersch REFERENCES
(Department of Anesthesiology, Intensive Care and Pain Medicine, 1. Evans L, Rhodes A, Alhazzani W et al. Surviving sepsis campaign:
University Hospital of Münster, Münster, Germany), Thiago Reis International Guidelines for Management of Sepsis and Septic
(D’Or Institute for Research and Education, Division of Kidney Shock 2021. Crit Care Med 2021;49:e1063–143. https://doi.org/10.
Transplantation, DF Star Hospital, Brasiĺia, Brazil; Laboratory of 1097/CCM.0000000000005337
Molecular Pharmacology, Faculty of Health Sciences, University 2. Bagshaw SM, Lapinsky S, Dial S et al. Acute kidney injury in
of Brasiĺia, Brasiĺia, Brazil), Thomas Rimmelé (Anesthesiology septic shock: clinical outcomes and impact of duration of hy-
and Critical Care Medicine, Edouard Herriot Hospital, Hospices potension prior to initiation of antimicrobial therapy. Intensive
Civils de Lyon, Lyon, France), Sean M. Bagshaw (Department of Care Med 2009;35:871–81. https://doi.org/10.1007/s00134-008-
Critical Care Medicine, Faculty of Medicine and Dentistry, Univer- 1367-2
sity of Alberta and Alberta Health Services, Edmonton, Alberta, 3. Morrell ED, Kellum JA, Pastor-Soler NM et al. Septic acute kidney
Canada), Rinaldo Bellomo (Department of Critical Care, Univer- injury: molecular mechanisms and the importance of stratifica-
sity of Melbourne, Parkville, Victoria, Australia; Department of tion and targeting therapy. Crit Care 2014;18:501. https://doi.org/
Intensive Care, Austin Hospital, Melbourne, Victoria, Australia; 10.1186/s13054- 014- 0501- 5
Australian and New Zealand Intensive Care Research Centre, 4. Bouchard J, Acharya A, Cerda J et al. A prospective interna-
Monash University, Melbourne, Victoria, Australia; Department tional multicenter study of AKI in the intensive care unit. Clin
of Intensive Care, Royal Melbourne Hospital, Melbourne, Victoria, J Am Soc Nephrol 2015;10:1324–31. https://doi.org/10.2215/CJN.
Australia), Vicenzo Cantaluppi (Nephrology and Kidney Trans- 04360514
plantation Unit, Department of Translational Medicine, University 5. Zarbock A, Nadim MK, Pickkers P et al. Sepsis-associated acute
of Piemonte Orientale, ‘Maggiore della Carità’ University Hospital, kidney injury: consensus report of the 28th Acute Disease
Novara, Italy), Akash Deep (Paediatric Intensive Care Unit, King’s Quality Initiative Workgroup. Nat Rev Nephrol 2023;19:401–17.
College Hospital NHS Foundation Trust, London, UK), Silvia De https://doi.org/10.1038/s41581- 023- 00683- 3
Rosa (Centre for Medical Sciences, University of Trento, Trento, 6. Messmer AS, Zingg C, Muller M et al. Fluid overload and mortality
Italy; Anesthesia and Intensive Care, Santa Chiara Regional in adult critical care patients—a systematic review and meta-
Hospital, Trento, Italy), Xosé Pérez-Fernández (Servei de Medicina analysis of observational studies. Crit Care Med 2020;48:1862–70.
Intensiva, Hospital Universitari de Bellvitge, L’Hospitalet de https://doi.org/10.1097/CCM.0000000000004617
Llobregat, Barcelona, Spain), Faeq Husain-Syed (Department of 7. Semler MW, Self WH, Wanderer JP et al. Balanced crystalloids
Internal Medicine II, University Hospital Giessen and Marburg, versus saline in critically ill adults. N Engl J Med 2018;378:829–
Justus-Liebig-University Giessen, Giessen, Germany), Sandra L. 39. https://doi.org/10.1056/NEJMoa1711584
A. Zarbock et al. | 33

8. Finfer S, Micallef S, Hammond N et al. Balanced multi- Nephrology (Carlton) 2017;22:412–9. https://doi.org/10.1111/nep.
electrolyte solution versus saline in critically ill adults. 12796
N Engl J Med 2022;386:815–26. https://doi.org/10.1056/ 23. Duranteau J, Deruddre S, Vigue B et al. Doppler monitoring of
NEJMoa2114464 renal hemodynamics: why the best is yet to come. Intensive
9. Semler MW, Wanderer JP, Ehrenfeld JM et al. Balanced crys- Care Med 2008;34:1360–1. https://doi.org/10.1007/s00134-008-
talloids versus saline in the intensive care unit. The SALT 1107-7
randomized trial. Am J Respir Crit Care Med 2017;195:1362–72. 24. Yu A, Zhao Q, Qu Y et al. Renal Doppler ultrasound in the evalua-
https://doi.org/10.1164/rccm.201607-1345OC tion of renal function in patients with sepsis. Appl Bionics Biomech
10. Kidney Disease: Improving Global Outcomes Acute Kidney In- 2022;2022:3472405. https://doi.org/10.1155/2022/3472405
jury Work Group. KDIGO clinical practice guideline for acute kid- 25. Saade A, Bourmaud A, Schnell D et al. Performance of Doppler-
ney injury. Kidney Int Suppl 2012;2:1–138. based resistive index and semiquantitative renal perfusion in
11. Bernier-Jean A, Beaubien-Souligny W, Goupil R et al. Diagnosis predicting persistent acute kidney injury according to opera-
and outcomes of acute kidney injury using surrogate and im- tor experience: post hoc analysis of a prospective multicen-
putation methods for missing preadmission creatinine values. ter study. Crit Care Med 2022;50:e361–9. https://doi.org/10.1097/
BMC Nephrol 2017;18:141. https://doi.org/10.1186/s12882-017- CCM.0000000000005372

Downloaded from https://academic.oup.com/ndt/article/39/1/26/7218568 by guest on 25 December 2023


0552-3 26. Chawla LS, Dommu A, Berger A et al. Urinary sediment cast scor-
12. Doi K, Yuen PS, Eisner C et al. Reduced production of crea- ing index for acute kidney injury: a pilot study. Nephron Clin Pract
tinine limits its use as marker of kidney injury in sepsis. J 2008;110:c145–50. https://doi.org/10.1159/000166605
Am Soc Nephrol 2009;20:1217–21. https://doi.org/10.1681/ASN. 27. Bagshaw SM, Haase M, Haase-Fielitz A et al. A prospective evalu-
2008060617 ation of urine microscopy in septic and non-septic acute kidney
13. Jin K, Murugan R, Sileanu FE et al. Intensive monitoring of urine injury. Nephrol Dial Transplant 2012;27:582–8. https://doi.org/10.
output is associated with increased detection of acute kidney in- 1093/ndt/gfr331
jury and improved outcomes. Chest 2017;152:972–9. https://doi. 28. Sadjadi M, Porschen C, von Groote TC et al. Implementation
org/10.1016/j.chest.2017.05.011 of nephroprotective measures to prevent acute kidney injury
14. Kellum JA, Sileanu FE, Murugan R et al. Classifying AKI by in septic patients: a retrospective cohort study. Anesth Analg
urine output versus serum creatinine level. J Am Soc Nephrol 2023; doi: 10.1213/ANE.0000000000006495. https://doi.org/10.
2015;26:2231–8. https://doi.org/10.1681/ASN.2014070724 1213/ANE.0000000000006495
15. Meersch M, Schmidt C, Hoffmeier A et al. Prevention of car- 29. Massoth C, Kullmar M, Moncho AP et al. Implementation of
diac surgery-associated AKI by implementing the KDIGO guide- the Kidney Disease: Improving Global Outcomes guidelines for
lines in high risk patients identified by biomarkers: the PrevAKI the prevention of acute kidney injury after cardiac surgery: an
randomized controlled trial. Intensive Care Med 2017;43:1551–61. international cohort survey. Eur J Anaesthesiol 2023;40:418–24.
https://doi.org/10.1007/s00134- 016- 4670- 3 https://doi.org/10.1097/EJA.0000000000001837
16. Zarbock A, Kullmar M, Ostermann M et al. Prevention of car- 30. Kullmar M, Weiss R, Ostermann M et al. A multinational ob-
diac surgery-associated acute kidney injury by implement- servational study exploring adherence with the Kidney Dis-
ing the KDIGO guidelines in high-risk patients identified by ease: Improving Global Outcomes recommendations for preven-
biomarkers: the PrevAKI-multicenter randomized controlled tion of acute kidney injury after cardiac surgery. Anesth Analg
trial. Anesth Analg 2021;133:292–302. https://doi.org/10.1213/ 2020;130:910–6. https://doi.org/10.1213/ANE.0000000000004642
ANE.0000000000005458 31. Moledina DG, Belliveau O, Yamamoto Y et al. Variation in best
17. Ostermann M, Zarbock A, Goldstein S et al. Recommendations practice measures in patients with severe hospital-acquired
on acute kidney injury biomarkers from the Acute Disease acute kidney injury: a multicenter study. Am J Kidney Dis
Quality Initiative Consensus Conference: a consensus state- 2021;77:547–9. https://doi.org/10.1053/j.ajkd.2020.08.013
ment. JAMA Netw Open 2020;3:e2019209. https://doi.org/10.1001/ 32. Mehta RL, Pascual MT, Soroko S et al. Diuretics, mortality, and
jamanetworkopen.2020.19209 nonrecovery of renal function in acute renal failure. JAMA
18. Molinari L, Del Rio-Pertuz G, Smith A et al. Utility of 2002;288:2547–53. https://doi.org/10.1001/jama.288.20.2547
biomarkers for sepsis-associated acute kidney injury stag- 33. Ho KM, Sheridan DJ. Meta-analysis of frusemide to prevent or
ing. JAMA Netw Open 2022;5:e2212709. https://doi.org/10.1001/ treat acute renal failure. BMJ 2006;333:420. https://doi.org/10.
jamanetworkopen.2022.12709 1136/bmj.38902.605347.7C
19. Kim H, Hur M, Lee S et al. Proenkephalin, neutrophil gelatinase- 34. Jaber S, Paugam C, Futier E et al. Sodium bicarbonate therapy for
associated lipocalin, and estimated glomerular filtration rates patients with severe metabolic acidaemia in the intensive care
in patients with sepsis. Ann Lab Med 2017;37:388–97. https://doi. unit (BICAR-ICU): a multicentre, open-label, randomised con-
org/10.3343/alm.2017.37.5.388 trolled, phase 3 trial. Lancet 2018;392:31–40. https://doi.org/10.
20. Martensson J, Martling CR, Oldner A et al. Impact of sepsis on 1016/S0140- 6736(18)31080- 8
levels of plasma cystatin C in AKI and non-AKI patients. Nephrol 35. Asfar P, Meziani F, Hamel JF et al. High versus low blood-pressure
Dial Transplant 2012;27:576–81. https://doi.org/10.1093/ndt/ target in patients with septic shock. N Engl J Med 2014;370:1583–
gfr358 93. https://doi.org/10.1056/NEJMoa1312173
21. Honore PM, Nguyen HB, Gong M et al. Urinary tissue inhibitor of 36. Tumlin JA, Murugan R, Deane AM et al. Outcomes in patients
metalloproteinase-2 and insulin-like growth factor-binding pro- with vasodilatory shock and renal replacement therapy treated
tein 7 for risk stratification of acute kidney injury in patients with intravenous angiotensin II. Crit Care Med 2018;46:949–57.
with sepsis. Crit Care Med 2016;44:1851–60. https://doi.org/10. https://doi.org/10.1097/CCM.0000000000003092
1097/CCM.0000000000001827 37. Bellomo R, Forni LG, Busse LW et al. Renin and survival in
22. Md Ralib A, Mat Nor MB, Pickering JW. Plasma neutrophil patients given angiotensin II for catecholamine-resistant va-
gelatinase-associated lipocalin diagnosed acute kidney injury sodilatory shock. A clinical trial. Am J Respir Crit Care Med
in patients with systemic inflammatory disease and sepsis. 2020;202:1253–61. https://doi.org/10.1164/rccm.201911-2172OC
34 | Nephrol Dial Transplant, 2024, Vol. 39, No. 1

38. Bellomo R, Wunderink RG, Szerlip H et al. Angiotensin I and an- 53. Wu L, Tiwari MM, Messer KJ et al. Peritubular capillary dys-
giotensin II concentrations and their ratio in catecholamine- function and renal tubular epithelial cell stress following
resistant vasodilatory shock. Crit Care 2020;24:43. https://doi. lipopolysaccharide administration in mice. Am J Physiol Renal
org/10.1186/s13054- 020- 2733- x Physiol 2007;292:F261–8. https://doi.org/10.1152/ajprenal.00263.
39. Gaudry S, Ricard JD, Leclaire C et al. Acute kidney injury in 2006
critical care: experience of a conservative strategy. J Crit Care 54. Gomez H, Ince C, De Backer D et al. A unified theory of
2014;29:1022–7. https://doi.org/10.1016/j.jcrc.2014.07.014 sepsis-induced acute kidney injury: inflammation, microcircu-
40. Investigators RRTS, Bellomo R, Cass A et al. Intensity of contin- latory dysfunction, bioenergetics, and the tubular cell adapta-
uous renal-replacement therapy in critically ill patients. N Engl tion to injury. Shock 2014;41:3–11. https://doi.org/10.1097/SHK.
J Med 2009;361:1627–38. 0000000000000052
41. VA/NIH Acute Renal Failure Trial Network. Intensity of renal 55. Zarbock A, Chawla L, Bellomo R. Why the renin-angiotensin-
support in critically ill patients with acute kidney injury. N Engl aldosterone system (RAAS) in critically ill patients can no
J Med 2008;359:7–20. longer be ignored. Crit Care 2021;25:389. https://doi.org/10.1186/
42. Zarbock A, Kellum JA, Schmidt C et al. Effect of early vs delayed s13054- 021- 03816- x
initiation of renal replacement therapy on mortality in critically 56. Flannery AH, Ortiz-Soriano V, Li X et al. Serum renin and ma-

Downloaded from https://academic.oup.com/ndt/article/39/1/26/7218568 by guest on 25 December 2023


ill patients with acute kidney injury: the ELAIN randomized clin- jor adverse kidney events in critically ill patients: a multicenter
ical trial. JAMA 2016;315:2190–9. https://doi.org/10.1001/jama. prospective study. Crit Care 2021;25:294. https://doi.org/10.1186/
2016.5828 s13054- 021- 03725- z
43. STARRT-AKI Investigators; Canadian Critical Care Trials Group, 57. Frank M, Duvezin-Caubet S, Koob S et al. Mitophagy is triggered
Australian and New Zealand Intensive Care Society Clinical Tri- by mild oxidative stress in a mitochondrial fission dependent
als Group et al. Timing of initiation of renal-replacement ther- manner. Biochim Biophys Acta 2012;1823:2297–310. https://doi.
apy in acute kidney injury. N Engl J Med 2020;383:240–51. https: org/10.1016/j.bbamcr.2012.08.007
//doi.org/10.1056/NEJMoa2000741 58. Waltz P, Carchman EH, Young AC et al. Lipopolysaccaride
44. Barbar SD, Clere-Jehl R, Bourredjem A et al. Timing of renal- induces autophagic signaling in macrophages via a TLR4,
replacement therapy in patients with acute kidney injury and heme oxygenase-1 dependent pathway. Autophagy 2011;7:315–
sepsis. N Engl J Med 2018;379:1431–42. https://doi.org/10.1056/ 20. https://doi.org/10.4161/auto.7.3.14044
NEJMoa1803213 59. Brealey D, Karyampudi S, Jacques TS et al. Mitochondrial dys-
45. Park JT, Lee H, Kee YK et al. High-dose versus conventional- function in a long-term rodent model of sepsis and organ fail-
dose continuous venovenous hemodiafiltration and patient ure. Am J Physiol Regul Integr Comp Physiol 2004;286:R491–7. https:
and kidney survival and cytokine removal in sepsis-associated //doi.org/10.1152/ajpregu.00432.2003
acute kidney injury: a randomized controlled trial. Am J Kid- 60. Liu JX, Yang C, Zhang WH et al. Disturbance of mitochondrial
ney Dis 2016;68:599–608. https://doi.org/10.1053/j.ajkd.2016.02. dynamics and mitophagy in sepsis-induced acute kidney in-
049 jury. Life Sci 2019;235:116828. https://doi.org/10.1016/j.lfs.2019.
46. Chung KK, Coates EC, Smith DJ, Jr et al. High-volume hemofil- 116828
tration in adult burn patients with septic shock and acute kid- 61. Dépret F, Hollinger A, Cariou A et al. Incidence and outcome of
ney injury: a multicenter randomized controlled trial. Crit Care subclinical acute kidney injury using penKid in critically ill pa-
2017;21:289. https://doi.org/10.1186/s13054- 017- 1878- 8 tients. Am J Respir Crit Care Med 2020;202:822–9. https://doi.org/
47. Joannes-Boyau O, Honore PM, Perez P et al. High-volume versus 10.1164/rccm.201910-1950OC
standard-volume haemofiltration for septic shock patients with 62. Fiorentino M, Xu Z, Smith A et al. Serial measurement of cell-
acute kidney injury (IVOIRE study): a multicentre randomized cycle arrest biomarkers [TIMP-2]·[IGFBP7] and risk for progres-
controlled trial. Intensive Care Med 2013;39:1535–46. https://doi. sion to death, dialysis, or severe acute kidney injury in patients
org/10.1007/s00134- 013- 2967- z with septic shock. Am J Respir Crit Care Med 2020;202:1262–70.
48. Schefold JC, von Haehling S, Pschowski R et al. The effect of https://doi.org/10.1164/rccm.201906-1197OC
continuous versus intermittent renal replacement therapy on 63. Seymour CW, Kennedy JN, Wang S et al. Derivation, valida-
the outcome of critically ill patients with acute renal failure tion, and potential treatment implications of novel clinical phe-
(CONVINT): a prospective randomized controlled trial. Crit Care notypes for sepsis. JAMA 2019;321:2003–17. https://doi.org/10.
2014;18:R11. https://doi.org/10.1186/cc13188 1001/jama.2019.5791
49. Bagshaw SM, Berthiaume LR, Delaney A et al. Continu- 64. Chaudhary K, Vaid A, Duffy A et al. Utilization of deep learning
ous versus intermittent renal replacement therapy for criti- for subphenotype identification in sepsis-associated acute kid-
cally ill patients with acute kidney injury: a meta-analysis. ney injury. Clin J Am Soc Nephrol 2020;15:1557–65. https://doi.org/
Crit Care Med 2008;36:610–7. https://doi.org/10.1097/01.CCM. 10.2215/CJN.09330819
0B013E3181611F552 65. Gray MP, Barreto EF, Schreier DJ et al. Consensus obtained for the
50. Kalakeche R, Hato T, Rhodes G et al. Endotoxin uptake by S1 nephrotoxic potential of 167 drugs in adult critically ill patients
proximal tubular segment causes oxidative stress in the down- using a modified Delphi method. Drug Saf 2022;45:389–98. https:
stream S2 segment. J Am Soc Nephrol 2011;22:1505–16. https: //doi.org/10.1007/s40264- 022- 01173- 4
//doi.org/10.1681/ASN.2011020203 66. Legrand M, Dupuis C, Simon C et al. Association between sys-
51. Kuwabara S, Goggins E, Okusa MD. The pathophysiology of temic hemodynamics and septic acute kidney injury in criti-
sepsis-associated AKI. Clin J Am Soc Nephrol 2022;17:1050–69. cally ill patients: a retrospective observational study. Crit Care
https://doi.org/10.2215/CJN.00850122 2013;17:R278. https://doi.org/10.1186/cc13133
52. De Backer D, Orbegozo Cortes D, Donadello K et al. Pathophys- 67. von Groote TC, Ostermann M, Forni LG et al. The AKI care bun-
iology of microcirculatory dysfunction and the pathogenesis dle: all bundle components are created equal-are they? Intensive
of septic shock. Virulence 2014;5:73–79. https://doi.org/10.4161/ Care Med 2022;48:242–5. https://doi.org/10.1007/s00134-021-
viru.26482 06601-0
A. Zarbock et al. | 35

68. Schleder S, Luerken L, Dendl LM et al. Impact of multide- 69. Pohlan J, Witham D, Farkic L et al. Body computed tomography in
tector computed tomography on the diagnosis and treatment sepsis: predictors of CT findings and patient outcomes in a retro-
of patients with systemic inflammatory response syndrome spective medical ICU cohort study. Emerg Radiol 2022;29:979–85.
or sepsis. Eur Radiol 2017;27:4544–51. https://doi.org/10.1007/ https://doi.org/10.1007/s10140- 022- 02083- 9
s00330- 017- 4897- 6

Downloaded from https://academic.oup.com/ndt/article/39/1/26/7218568 by guest on 25 December 2023

Received: May 9, 2023; Editorial decision: June 5, 2023


© The Author(s) 2023. Published by Oxford University Press on behalf of the ERA.
All rights reserved. For permissions, please e-mail: journals.permissions@oup.com

You might also like