Skip to main content

Open Access 12.01.2025 | Review Article

Kidney Injury Following Cardiac Surgery: A Review of Our Current Understanding

verfasst von: Christine-Elena Kamla, Melanie Meersch-Dini, Lilian Monteiro Pereira Palma

Erschienen in: American Journal of Cardiovascular Drugs

Abstract

Around one-quarter of all patients undergoing cardiac procedures, particularly those on cardiopulmonary bypass, develop cardiac surgery-associated acute kidney injury (CSA-AKI). This complication increases the risk of several serious morbidities and of mortality, representing a significant burden for both patients and the healthcare system. Patients with diminished kidney function before surgery, such as those with chronic kidney disease, are at heightened risk of developing CSA-AKI and have poorer outcomes than patients without preexisting kidney injury who develop CSA-AKI. Several mechanisms are involved in the development of CSA-AKI; injury is primarily thought to result from an amplification loop of inflammation and cell death, with complement and immune system activation, cardiopulmonary bypass, and ischemia-reperfusion injury all contributing to pathogenesis. At present there are no effective, targeted pharmacological therapies for the prevention or treatment of CSA-AKI, although several preclinical trials have shown promise, and clinical trials are under way. Progress in the understanding of the complex pathophysiology of CSA-AKI is needed to improve the development of successful strategies for its prevention, management, and treatment. In this review, we outline our current understanding of CSA-AKI development and management strategies and discuss potential future therapeutic targets under investigation.
Key Points
Cardiac surgery-associated acute kidney injury is a common postoperative complication associated with considerable morbidity and mortality, and its incidence is increased in patients with impaired preoperative kidney function.
Several mechanistic triggers during cardiac surgery can result in a cycle of uncontrolled complement activation, inflammation, and kidney injury.
Currently, there are no targeted, effective pharmacological interventions for cardiac surgery-associated acute kidney injury. This review discusses the rationale behind several upcoming and ongoing clinical trials.

1 Introduction

1.1 What is Cardiac Surgery-Associated Acute Kidney Injury (CSA-AKI)?

Cardiac surgery-associated acute kidney injury (CSA-AKI) is a common perioperative complication associated with substantial morbidity and mortality. Patients with CSA-AKI are more likely to experience associated postoperative morbidities such as an increased risk of infection, prolonged stay in the intensive care unit (ICU)—including a prolonged need for mechanical ventilation—and further cardiovascular events (including myocardial infarctions, strokes, and heart failure) than are surgical patients with no postoperative AKI [13]. In addition, CSA-AKI is associated with poorer long-term kidney function; patients undergoing cardiac surgery who develop AKI are significantly more likely to experience major adverse kidney events (MAKE) [4, 5]. The umbrella term MAKE includes progression from AKI to chronic kidney disease (CKD) and end-stage kidney disease, as well as an increased risk of mortality in the years following surgery [6]. Although it has been demonstrated that the risk of MAKE – including progression to CKD—rises significantly with the length and severity of CSA-AKI, it is important to highlight that any-stage AKI is associated with an increased risk of MAKE and that permanent injury can occur even when recovery is early [2, 5, 714].
Another major clinical consequence, occurring in 2–5% of patients with CSA-AKI, is the need for life-saving kidney replacement therapy (KRT) [15, 16]. The need for prolonged KRT presents its own difficulties, many of which are long term, and is associated with significantly increased healthcare costs [2, 17, 18]. It has also been shown that both operative and long-term mortality risks are increased, in some cases by up to eight times, in patients who develop CSA-AKI compared with those who do not [2, 8, 9, 17, 1922]. The substantial morbidity and mortality burden, alongside the requirement for longer hospitalization, increased time in the ICU, and greater need for further interventions, emphasizes the lasting impacts of CSA-AKI on both patients and the healthcare system [23].
Although there is some awareness of this complication and its impact, greater understanding of how to identify and manage patients at risk of CSA-AKI, and how to optimally manage patients who do present with CSA-AKI, is needed to reduce the risk of its development and improve outcomes. In this review, we detail the incidence of known risk factors, the underlying pathophysiology of CSA-AKI, and the current and developing management strategies. We also explore the existing evidence supporting the role of inflammation and immunity, particularly the complement system, in this serious post-surgical complication.

2 CSA-AKI: How Important is This Complication?

2.1 Which Patients are At Risk?

Historically, it has been difficult to assess the true incidence of CSA-AKI because of differing global diagnostic criteria, pooled data combining a variety of cardiac procedures, and minimal data from low-income countries being included in incidence studies [20]. Recently, a large prospective observational study using pooled incidence rates from 30 countries reported that, globally, 25.9% of patients undergoing cardiac surgery subsequently developed CSA-AKI; this is in line with previous estimates from the literature [20, 24, 25]. Cardiac surgery has also been identified as the second most common cause of AKI in the ICU, after sepsis [26, 27]. Secondary analysis of this study demonstrated that around 13% of patients undergoing cardiac surgery developed prolonged AKI (> 7 days to < 3 months duration)—also known as acute kidney disease. These patients were more likely to have had an early occurrence of postoperative AKI than patients who did not develop acute kidney disease [28].
The emergence of CSA-AKI is multifactorial, and a wide range of risk factors and pathophysiological mechanisms contribute to its development and progression. Several patient characteristics known to be risk factors for CSA-AKI include female sex, non-white race, advanced age, preoperative hypertension, and preoperative anemia [2, 15, 29]. A range of procedure-related risk factors also increase the risk posed by surgery performed on cardiopulmonary bypass (CPB), including complex surgeries requiring prolonged CPB and aortic cross clamp durations, hypoperfusion during surgery caused by phases of low flow when on CPB, and direct contact of blood with the CPB circuitry [2, 15, 30].
Impaired preoperative kidney function is an important and often overlooked risk factor; lower preoperative estimated glomerular filtration rate (eGFR) has been demonstrated to be an accurate independent predictor of mortality following cardiac surgery [14, 31]. Existing CKD also increases the risk of CSA-AKI, from ~ 25% in patients without CKD to ~ 50% in those with CKD [25, 32]. This is especially significant as it has been demonstrated that patients with preoperative CKD who develop CSA-AKI have poorer long-term outcomes and a significantly higher risk of dialysis and/or mortality than patients with CSA-AKI who have no previous kidney disease [33].

2.2 How is CSA-AKI Identified in Patients?

It has been suggested that the detection and management of CSA-AKI varies widely because of sub-optimal use of available clinical guidance [14, 34, 35]. Several definitions of AKI are used in clinical practice, and the Kidney Disease: Improving Global Outcomes (KDIGO) definition—noted for its improved diagnostic sensitivity over alternative criteria—is used most routinely [34, 36]. The KDIGO guidelines define serum creatinine (sCr) and urine output thresholds for AKI, with criteria allowing staging from mild to severe (Table 1). Once kidney injury has been identified, its impact is routinely assessed by monitoring eGFR, a surrogate readout for kidney function. Currently, eGFR is calculated using the creatinine-based CKD Epidemiology Collaboration (CKD-EPI) equation for adults, or the CKD in children study (CKiD) equation in pediatric patients [37, 38].
Table 1.
Kidney Disease: Improving Global Outcomes serum creatinine and urine output criteria for staging of acute kidney injury [Adapted from 40, 44, 45]
AKI stage (severity)
sCr
Urine output
I (mild)
≥ 0.3 mg/dl increase within 48 h or 1.5–1.9 × baseline for 7 days
< 0.5 ml/kg/h for 6–12 h
II (moderate)
2–2.9 × baseline
< 0.5 ml/kg/h for 12 h
III (severe)
≥ 3 × baseline, or increase to ≥4 mg/dl, or initiation of KRT
< 0.3 ml/kg/h for 24 h or anuria for 12 h
AKI acute kidney injury; KRT kidney replacement therapy; sCr serum creatinine.
However, reductions in eGFR occur only after significant insult and can return to baseline despite latent injury masked by the renal function reserve (RFR), namely the functional capacity of the kidneys above the resting filtration rate [3941]. This buffer function acts to protect the kidney from injury during times of stress but can, in effect, mask early injury and delay diagnosis [39]. Therefore, the standard diagnostic tools such as sCr, eGFR, and urine output may lack sensitivity for detecting mild, but nonetheless clinically relevant, kidney damage [42, 43].

2.3 How Can Diagnosis of CSA-AKI be Improved?

It has been posited that the extent of previous injury to the kidneys, and the remaining RFR, may have a substantial role in determining the extent of injury following subsequent insults; indeed, patients with higher preoperative RFR have been shown to be somewhat protected against developing severe CSA-AKI [46]. Preexisting CKD leads to a reduced RFR, impairing the ability of the kidney to compensate for subsequent kidney injuries such as CSA-AKI and possibly contributing to the development of undetected, but still impactful, AKI following limited kidney insult [32].
Currently, there are no sensitive assays in routine clinical use to detect early kidney damage before changes in markers such as sCr can be detected [47, 48]. Preliminary evidence indicates that changes in the levels of a selection of urinary biomarkers (e.g., NGAL, TIMP-2, and IGFBP7) and plasma biomarkers (e.g., interleukin-8 and tumor necrosis factor-α) may be detectable following early stress and damage, before injury to the kidney is substantial enough to meet the currently defined AKI criteria [47, 49, 50]. Several recent trials—such as PrevAKI1, PrevAKI2, and BigpAK—used the biomarkers urinary TIMP-2 and IGFBP7 to identify patients at high risk of AKI [5052]. Following these studies, the Acute Disease Quality Initiative Consensus Conference recommended integration of biomarkers into routine clinical practice, with the aim of improving implementation of measures to prevent progressive kidney damage and subsequent AKI [43]. The assessment of CSA-AKI biomarkers specifically in pediatric patients undergoing cardiac surgery identified urinary NGAL, alongside other proteins, including pre-and post-surgical serum FGF23 levels, as markers of post-surgery AKI [53, 54]. Biomarkers with increased sensitivity for kidney injury will further broaden the definition of AKI, allowing identification of subclinical AKI where there is initial stress or damage following surgery [34]. Biomarkers are evaluated for their effectiveness in the prediction, identification, and improved management of patients with AKI. Their use in the preoperative setting needs to be standardized to integrate them into day-to-day practice [43].
Risk following cardiac surgery can be predicted with tools such as the Society of Thoracic Surgeons operative risk calculator. Other scores and tools include the Cleveland Clinic scoring system and the Leicester scoring system [55, 56]. Although these tools are useful for personalized shared decision-making, they do have limitations; for example, the presence of preoperative CKD is a known risk factor for CSA-AKI development but is not recorded in the Society of Thoracic Surgeons operative risk calculator [57, 58]. Additionally, studies to assess the impact of approaches such as real-time AKI risk stratification-biomarker-directed fluid management and the use of the renal angina index to predict severe AKI are ongoing [59, 60]. It is unclear how consistently these tools are used across different countries and regions. Continuing refinement of these tools, as well as initiatives to ensure their consistent adoption in routine clinical practice worldwide, is key [5658].

3 Mechanism of Injury in CSA-AKI

The pathophysiology of CSA-AKI is multifactorial, which contributes to the difficulty in its identification and management; however, tissue pathologies in CSA-AKI are thought to result in large part from ischemia-reperfusion injury (IRI) [61]. During CPB, the conversion from physiological pulsatile blood flow to laminar flow triggers an intense inflammatory response and reflexive kidney vasoconstriction [62]. Kidney hypoperfusion leads to ischemia, which causes the kidney to be starved of oxygen and adenosine triphosphate, leading to the death of tubular epithelial and endothelial cells [63]. Cellular damage, apoptosis, and/or necrosis occurring during ischemia releases damage-associated molecular patterns (DAMPs), and detection of these by kidney cells and resident immune cells leads to the release of proinflammatory cytokines, alongside activation of complement proteins [64, 65]. At the termination of CPB, reperfusion occurs, which can itself cause substantial damage to the kidney tissue, due in part to infiltration of circulating leukocytes, which migrate to the injury site. Here, they become activated, releasing more proinflammatory cytokines and producing reactive oxygen species, causing further cell death, release of DAMPs, and activation of the complement system, perpetuating and amplifying a cycle of inflammation and injury [66]. Combined, this process of IRI appears to be a major driving force behind CSA-AKI [64].
Interaction between the blood and the extra-corporeal circuit during CPB can also contribute to CSA-AKI, likely due to both the nephrotoxic effect of hemolysis and an uncontrolled activation of circulating complement [67]. Hemolysis results in the generation of free heme, which induces oxidative stress; levels of heme and markers of oxidative stress have been shown to correlate with the risk of CSA-AKI [68]. Laminar blood flow through CPB circuitry further exacerbates hemolysis and damage to the endothelium through shear stress, contributing to kidney injury [69]. The process of CPB has also been shown to contribute to IRI via hemodilution and decreased oxygen tension in the kidneys, suggesting that CPB may contribute to CSA-AKI development via numerous mechanisms [70, 71].
Fluid overload is a risk factor for the development of AKI and is associated with increased severity of AKI and higher mortality rates [72]. The pathophysiological mechanisms underlying this relationship include increased intra-abdominal pressure, venous congestion, and impaired kidney perfusion, all of which contribute to the development of AKI [73].
The relative benefit of off-pump surgeries to avoid CPB is debated: some studies have reported a reduced incidence of CSA-AKI when CPB is avoided, and others have concluded that off-pump surgery does not reduce the need for KRT or mortality [74, 75]. However, in practice, only a small number of cardiac procedures, such as coronary artery bypass graft surgery, can be performed off-pump, and appropriate patient selection for off-pump procedures is crucial, meaning this approach may not be suitable for most patients at risk of CSA-AKI [76].
A schematic detailing the key pathophysiological mechanisms underlying the development of CSA-AKI is presented in Fig. 1.

3.1 CSA-AKI and the Complement System

The complement system is an integral component of the innate immune system, providing defense from microbial invasion, clearance of injured cells, and removal of apoptotic cells [77]. The complement system has also been implicated as a pathophysiological driver of several other conditions as well as CSA-AKI [7880]. Briefly, the complement system is composed of circulating soluble proteins, membrane-bound receptors, and a variety of regulatory proteins that contribute to signaling via three pathways: classical, lectin, and alternative. All three pathways result in activation of the terminal complement pathway, causing inflammation through release of anaphylatoxins (C3a and C5a), and cell lysis via formation of the membrane attack complex (MAC, C5b-9) (Fig. 2) [77].
Several studies have demonstrated the role of complement in IRI, particularly components of the terminal complement cascade. Anaphylatoxins, such as C3a and C5a, are proinflammatory mediators, acting as both chemo-attractants for and activators of leukocytes; they are also detectable by non-immune cells, which express their receptors [81]. Mouse models have demonstrated that deletions in terminal complement components, or their receptors, are protective against kidney damage in the context of IRI [82, 83]. The role of complement in kidney IRI has been well reviewed previously, and—although all complement pathways are involved to some extent—injury seems primarily mediated by terminal complement, namely generation of C5a, MAC formation, and cell lysis [64].
Increasing evidence indicates that the complement system is also activated throughout and after CPB. First, activation markers of both the classical and the alternative complement pathways have been shown to increase during CPB [84]. Activation of these pathways results in terminal complement activation, which can cause secondary damage to kidney cells through a variety of mechanisms, including kidney cell lysis [77]. It is noteworthy that, although complement activation can cause hemolysis, the presence of free heme triggered by CPB can in turn activate the complement system, highlighting a possible feedback loop of worsening activation and damage [80]. It has also been demonstrated that alternative pathway activation during tubular necrosis leads to localized deposition of C3d in the kidney tubules and increased levels of the complement fragment Ba in the urine of patients with AKI; C3d and Ba levels both correlate with increases in sCr [85, 86]. These data indicate a role for complement activation in the mechanism of ischemic injury and direct cellular damage in the kidneys, following cardiac surgery requiring CPB (Fig. 3).

4 Current Management and Future Targets

Current management of CSA-AKI focuses on risk reduction and prevention, because effective pharmacological therapies for the treatment of emergent CSA-AKI are lacking. Several proposed treatments, such as fenoldopam (arterial vasodilator), levosimendan (inotrope), and spironolactone (aldosterone antagonist), have failed to demonstrate any significant efficacy in reducing AKI development and/or any substantial effect on patient outcomes following cardiac surgery [2, 8791]. Although severe AKI is often managed with KRT, the optimal timing of such treatments is still debated, and more targeted management options for CSA-AKI, especially those that can be administered earlier in the disease course, are still required [92]. Specifically in pediatric patients, a meta-analysis published in 2021 assessed data from a total of 2339 patients undergoing cardiac surgery from 20 randomized controlled trials (RCTs) and found that the effectiveness of most strategies currently employed to prevent CSA-AKI in children are not supported by current clinical data; there is some limited support for the use of dexmedetomidine and remote ischemic preconditioning (RIPC) [93].
Currently, the KDIGO guidelines recommend a perioperative bundle of care in patients at high risk to prevent and reduce the severity of AKI following major surgical procedures. This bundle includes optimization of perfusion, pressure and fluid management, halting nephrotoxic medication, avoiding contrast agents, and close monitoring of postoperative kidney function [2, 52, 94]. However, only around 5% of patients receive the full bundle of care in clinical practice, despite evidence from the PrevAKI trials demonstrating that biomarker-guided use of the KDIGO bundle of interventions was more effective in reducing the severity of CSA-AKI than standard ICU care [52, 95]. Although the severity of CSA-AKI correlates with the severity of patient outcomes, any occurrence of post-surgical AKI—including mild or transient injury—confers an increased risk of MAKE, highlighting the importance of wider implementation of preventive strategies [5].
Two organizations, Enhanced Recovery After Surgery and the PeriOperative Quality Initiative, have collaborated to develop expert-led guidance for the treatment of cardiac surgery patients. The outcome of this collaboration was an established protocol for the perioperative care of cardiac patients, with a recommendation for multidisciplinary team involvement throughout to improve patient outcomes, including in reducing the incidence of CSA-AKI [96]. This multi-modal approach aims to allow specialist management of each aspect of patient health; for example, ongoing discussions between clinical specialists such as surgeons, intensivists (preferably with critical care nephrology expertise), and perfusionists when managing cardiac surgery patients may aid in the recognition of individuals at high risk of CSA-AKI, support earlier diagnosis of postoperative kidney injury, and reduce delays in the implementation of appropriate management approaches. This may ultimately lead to a reduction in the risk of CSA-AKI and the associated morbidity and mortality [97].
Outside the preventive KDIGO bundle of care, there are currently two non-pharmacological, surgical measures that may reduce the risk of mild CSA-AKI. First, the Enhanced Recovery After Surgery and PeriOperative Quality Initiative consensus report strongly recommends goal-directed perfusion as standard of care—whereby oxygen delivery is maintained above a critical level throughout surgery, although its effectiveness in moderate and severe cases of CSA-AKI remains to be demonstrated [96, 98, 99] and a meta-analysis of two RCTs found a very low level of evidence for its use [99]. Second, several studies have explored the efficacy of RIPC for reducing the emergence of CSA-AKI, as discussed. Induction of RIPC consists of repeated, short instances of ischemia–reperfusion induced in a remote location (often a distant limb) to promote protection against subsequent IRI [100]. A meta-analysis of 21 RCTs supported the use of RIPC in reducing the incidence of CSA-AKI, the requirement for ventilation, and the need for ICU admission, particularly in younger patients undergoing less complex surgeries [101]. However, this meta-analysis did not account for method of anesthesia, despite previous evidence demonstrating that the effect of RIPC was eliminated with the administration of propofol [102]. A further meta-analysis of 31 RCTs found a moderate level of evidence for the use of RIPC [99]. Despite this, the use of RIPC is likely to reduce the risk of CSA-AKI, kidney injury markers, and the need for KRT in high-risk patients not anesthetized with propofol [103].
Although several clinical trials have evaluated pharmacological interventions for the prevention of CSA-AKI, most of these have failed to meet their primary endpoints. Despite promising tolerability and demonstrable renoprotective effects observed in a phase Ib study using recombinant alpha-1-microglobulin (RMC-035) to target free heme [104], the subsequent phase II trial (NCT05126303) was terminated prematurely because of early futility. Further, a phase III trial of a p53-mediated cell death inhibitor (QPI-1002) was terminated because of low efficacy (NCT03510897). Lastly, although prophylactic delivery of recombinant human erythropoietin was initially shown to reduce the incidence of postoperative AKI in one RCT [105], later meta-analyses revealed that, although it was efficacious if given before anesthesia, the effect was limited to low-risk patients and therefore had no effect on the overall incidence of CSA-AKI [106, 107]. These trials demonstrate a continuing pattern of pharmacological therapies that either fail to target the root causes of CSA-AKI or neglect to show benefit in the most high-risk populations, including patients with preexisting CKD.
Despite many trials failing to meet their primary endpoints, several ongoing studies are evaluating pharmacological agents for the prevention of CSA-AKI. One pilot trial is using low-dose lithium, a well-tolerated glycogen synthase kinase-3β inhibitor, within the perioperative period to prevent kidney injury in patients at high risk of AKI (NCT03056248). Upregulation of glycogen synthase kinase-3β has been observed following kidney injury, and this trial aims to dampen immune-mediated injury by inhibiting upstream triggers [108]. Another recent phase III trial, investigating the renoprotective effect of ilofotase alfa (human recombinant alkaline phosphatase) in patients with sepsis-associated AKI, was terminated early because of futility in the primary endpoint [109]. However, the observation of a reduction in the occurrence of MAKE led to the initiation of a phase II trial of ilofotase alfa in patients undergoing cardiac surgery (NCT06168799). A further trial, studying the effects of LSALT peptide in patients undergoing cardiac surgery with CPB, is currently recruiting (NCT05879432); the LSALT peptide is an inhibitor of dipeptidase-1, an endothelial adhesion molecule involved in the recruitment of innate immune cells.
More recently, positive results from the PROTECTION study (NCT03709264) have demonstrated the benefits of intravenous amino acid administration in protecting adult patients from the development of CSA-AKI. The trial found that, in patients scheduled to undergo cardiac surgery, amino acid infusions given for up to 3 days were well tolerated and associated with a significant reduction in the risk of developing any-stage CSA-AKI compared with placebo (relative risk 0.85; 95% confidence interval 0.77–0.94; p = 0.002) [110]. This study was limited to using only sCr to diagnose AKI, and most patients had less severe injury than is highlighted in our review. However, the promising benefits observed in this study raise the possibility that amino acid depletion during cardiac surgery may also play a pathogenic role in the development of CSA-AKI [110, 111].
It is also important to note that the clearance and distribution of drugs is altered in patients with kidney impairment, so calculations such as CKD-EPI and CKiD should be used to aid rational dose adjustments in patients with preexisting kidney damage; consideration of differences in drug metabolism and pharmacokinetics in this population may help further improve the design of upcoming clinical trials [112]. Further understanding of the pathophysiological mechanisms underpinning CSA-AKI development, and treatments targeting these, are clearly required, and the results of the ongoing trials will be of great interest to the field.

4.1 Targeting Complement-Mediated Kidney Injury

Inhibition of the terminal complement component C5 with the monoclonal antibody pexelizumab was demonstrated to be safe in patients undergoing cardiac surgery and was associated with reduced mortality rates in early clinical trials [113, 114]. Unfortunately, these promising findings were not fully validated in later cohorts, and results failed to meet statistical significance [115]. However, following this initial proof of concept, a randomized, double-blind, placebo-controlled, global phase III study of ravulizumab in adults with CKD undergoing surgery with CPB was designed and is currently recruiting (ARTEMIS, NCT05746559). Ravulizumab is a humanized monoclonal antibody against complement C5 that inhibits C5 cleavage to C5a and C5b and thus prevents formation of the MAC [116, 117]. Complement C5 inhibition has been safely and effectively used in the treatment of patients with complement-mediated illnesses—such as atypical hemolytic uremic syndrome, generalized myasthenia gravis, and paroxysmal nocturnal hemoglobinuria—for almost 20 years [118, 119]. Furthermore, recent results from phase II and phase III studies have confirmed that ravulizumab is also well tolerated; long-term follow-up data from the ravulizumab 301 study demonstrated positive efficacy and tolerability following up to 6 years of treatment in patients with paroxysmal nocturnal hemoglobinuria [117, 120122]. One major risk of complement inhibition is increased susceptibility to infection with encapsulated bacteria, especially Neisseria meningitidis. Vaccination (and additional antibiotic prophylaxis as appropriate) is a requirement for all patients using complement inhibitors. The risk of such infections is elevated following complement inhibition, but recent real-world data have shown that this can be successfully mitigated at the population level and should not be a barrier to treatment [123].
The ARTEMIS study will assess the occurrence of MAKE in participants with stage 3 or 4 CKD following non-emergent cardiac surgery with and without inhibition of the terminal complement pathway. The results of ARTEMIS will indicate whether complement activation during and after surgery is a key driving force in CSA-AKI, as suggested by the available preclinical and patient-level data [86, 124]. Beyond the potential kidney-specific benefits, recent evidence also suggests that complement inhibition may have an additional therapeutic effect during cardiac surgery: a positive correlation has been observed between serum levels of both MAC components and anaphylatoxins with blood loss following surgery with CPB [84]. Therefore, in addition to providing renoprotection via complement inhibition, blood loss—another factor known to directly influence patient outcomes following surgery—may also be reduced.

5 Conclusion

CSA-AKI is an underappreciated perioperative complication that significantly increases morbidity and mortality in patients undergoing cardiac surgery, particularly in high-risk populations such as those with preexisting CKD. The pathophysiology of CSA-AKI is multifactorial, encompassing patients’ preoperative clinical characteristics, surgical factors, and postoperative management. Substantial evidence indicates that activation of immune and inflammatory cascades is key to the pathophysiology of CSA-AKI, likely beginning with contact-mediated complement activation, alongside IRI. This creates a feedback loop whereby complement-mediated kidney injury causes the release of proinflammatory mediators, propagating further activation of complement in the kidneys and perpetuating kidney damage. The KDIGO bundle has been suggested to be efficacious in reducing the overall incidence and severity of CSA-AKI, including in patients at high risk for postoperative AKI. However, implementation of the full bundle is not recommended in all patients, and it is uncommon for patients to receive the complete bundle of interventions. It also remains unclear whether all components of the bundle are equally as effective (or necessary) to control the risk of developing CSA-AKI; adequately powered global trials to interrogate this further are ongoing [125]. Patients with preexisting CKD are at a significantly increased risk of developing CSA-AKI and suffer more severely in both the short and the long term, highlighting an unmet need for thorough assessment of kidney function before surgery. It also emphasizes the need for more effective therapeutic options for reducing the risk of CSA-AKI development and for the treatment of CSA-AKI in patients who develop this complication. Although there are some promising preclinical studies and therapeutics in development, further research is required to improve our understanding of the pathophysiology of CSA-AKI and allow the development of more effective and targeted strategies for the prevention, management, and treatment of this complex post-surgical complication.

Acknowledgements

Medical writing support was funded by Alexion, AstraZeneca Rare Disease, Boston, Massachusetts, USA, and was provided by Gillian May Mackie, PhD, and Matthew D Badham, PhD, of Bioscript Group, Macclesfield, UK. The authors would like to acknowledge Åsa Lommelé, PhD, and Thainá Jehá, MD, of Alexion, AstraZeneca Rare Disease, for their critical review of the manuscript.

Declarations

Conflict of interest

Dr Kamla is the site investigator for the ARTEMIS trial at the University Hospital LMU Munich. Dr Meersch-Dini has received lecture fees from Fresenius Medical Care and Baxter and an unrestricted research grant from Baxter. Dr Palma has received honoraria from Alexion, AstraZeneca Rare Disease Brazil, and consultancy fees from Novartis and Apellis.

Author contributions

All authors contributed to writing and reviewing the article. All authors have read and approved the final version of the manuscript.

Ethics approval

Not applicable.
Not applicable.

Data availability

Not applicable.

Compliance with ethical standards

Not applicable.
Not applicable.

Code availability

Not applicable.

Ethics statement

This is a review article, with no new experimental data generated in either humans or animal models. Therefore, no ethical approval was required.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial 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://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

© Springer Medizin

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

© Springer Medizin

e.Dent – Das Online-Abo der Zahnmedizin

Online-Abonnement

Mit e.Dent erhalten Sie Zugang zu allen zahnmedizinischen Fortbildungen und unseren zahnmedizinischen und ausgesuchten medizinischen Zeitschriften.

Literatur
1.
Zurück zum Zitat Hansen MK, Gammelager H, Jacobsen CJ, Hjortdal VE, Layton JB, Rasmussen BS, et al. Acute kidney injury and long-term risk of cardiovascular events after cardiac surgery: a population-based cohort study. J Cardiothorac Vasc Anesth. 2015;29(3):617–25.PubMedCrossRef Hansen MK, Gammelager H, Jacobsen CJ, Hjortdal VE, Layton JB, Rasmussen BS, et al. Acute kidney injury and long-term risk of cardiovascular events after cardiac surgery: a population-based cohort study. J Cardiothorac Vasc Anesth. 2015;29(3):617–25.PubMedCrossRef
2.
Zurück zum Zitat Schurle A, Koyner JL. CSA-AKI: incidence, epidemiology, clinical outcomes, and economic impact. J Clin Med. 2021;10(24):5746. Schurle A, Koyner JL. CSA-AKI: incidence, epidemiology, clinical outcomes, and economic impact. J Clin Med. 2021;10(24):5746.
3.
Zurück zum Zitat Hobson C, Ozrazgat-Baslanti T, Kuxhausen A, Thottakkara P, Efron PA, Moore FA, et al. Cost and mortality associated with postoperative acute kidney injury. Ann Surg. 2015;261(6):1207–14.PubMedCrossRef Hobson C, Ozrazgat-Baslanti T, Kuxhausen A, Thottakkara P, Efron PA, Moore FA, et al. Cost and mortality associated with postoperative acute kidney injury. Ann Surg. 2015;261(6):1207–14.PubMedCrossRef
4.
Zurück zum Zitat See EJ, Toussaint ND, Bailey M, Johnson DW, Polkinghorne KR, Robbins R, et al. Risk factors for major adverse kidney events in the first year after acute kidney injury. Clin Kidney J. 2021;14(2):556–63.PubMedCrossRef See EJ, Toussaint ND, Bailey M, Johnson DW, Polkinghorne KR, Robbins R, et al. Risk factors for major adverse kidney events in the first year after acute kidney injury. Clin Kidney J. 2021;14(2):556–63.PubMedCrossRef
5.
Zurück zum Zitat Molina Andujar A, Escudero VJ, Pineiro GJ, Lucas A, Rovira I, Matute P, et al. Impact of cardiac surgery associated acute kidney injury on 1-year major adverse kidney events. Front Nephrol. 2023;3:1059668.PubMedPubMedCentralCrossRef Molina Andujar A, Escudero VJ, Pineiro GJ, Lucas A, Rovira I, Matute P, et al. Impact of cardiac surgery associated acute kidney injury on 1-year major adverse kidney events. Front Nephrol. 2023;3:1059668.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat McKown AC, Wang L, Wanderer JP, Ehrenfeld J, Rice TW, Bernard GR, et al. Predicting major adverse kidney events among critically ill adults using the electronic health record. J Med Syst. 2017;41(10):156.PubMedPubMedCentralCrossRef McKown AC, Wang L, Wanderer JP, Ehrenfeld J, Rice TW, Bernard GR, et al. Predicting major adverse kidney events among critically ill adults using the electronic health record. J Med Syst. 2017;41(10):156.PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Bhatraju PK, Zelnick LR, Chinchilli VM, Moledina DG, Coca SG, Parikh CR, et al. Association between early recovery of kidney function after acute kidney injury and long-term clinical outcomes. JAMA Netw Open. 2020;3(4): e202682.PubMedPubMedCentralCrossRef Bhatraju PK, Zelnick LR, Chinchilli VM, Moledina DG, Coca SG, Parikh CR, et al. Association between early recovery of kidney function after acute kidney injury and long-term clinical outcomes. JAMA Netw Open. 2020;3(4): e202682.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Brown JR, Kramer RS, Coca SG, Parikh CR. Duration of acute kidney injury impacts long-term survival after cardiac surgery. Ann Thorac Surg. 2010;90(4):1142–8.PubMedCrossRef Brown JR, Kramer RS, Coca SG, Parikh CR. Duration of acute kidney injury impacts long-term survival after cardiac surgery. Ann Thorac Surg. 2010;90(4):1142–8.PubMedCrossRef
9.
Zurück zum Zitat Xu JR, Zhu JM, Jiang J, Ding XQ, Fang Y, Shen B, et al. Risk factors for long-term mortality and progressive chronic kidney disease associated with acute kidney injury after cardiac surgery. Medicine (Baltimore). 2015;94(45): e2025.PubMedCrossRef Xu JR, Zhu JM, Jiang J, Ding XQ, Fang Y, Shen B, et al. Risk factors for long-term mortality and progressive chronic kidney disease associated with acute kidney injury after cardiac surgery. Medicine (Baltimore). 2015;94(45): e2025.PubMedCrossRef
10.
Zurück zum Zitat Coca SG, Singanamala S, Parikh CR. Chronic kidney disease after acute kidney injury: a systematic review and meta-analysis. Kidney Int. 2012;81(5):442–8.PubMedCrossRef Coca SG, Singanamala S, Parikh CR. Chronic kidney disease after acute kidney injury: a systematic review and meta-analysis. Kidney Int. 2012;81(5):442–8.PubMedCrossRef
11.
Zurück zum Zitat Ryden L, Sartipy U, Evans M, Holzmann MJ. Acute kidney injury after coronary artery bypass grafting and long-term risk of end-stage renal disease. Circulation. 2014;130(23):2005–11.PubMedCrossRef Ryden L, Sartipy U, Evans M, Holzmann MJ. Acute kidney injury after coronary artery bypass grafting and long-term risk of end-stage renal disease. Circulation. 2014;130(23):2005–11.PubMedCrossRef
12.
Zurück zum Zitat Menez S, Moledina DG, Garg AX, Thiessen-Philbrook H, McArthur E, Jia Y, et al. Results from the TRIBE-AKI Study found associations between post-operative blood biomarkers and risk of chronic kidney disease after cardiac surgery. Kidney Int. 2021;99(3):716–24.PubMedCrossRef Menez S, Moledina DG, Garg AX, Thiessen-Philbrook H, McArthur E, Jia Y, et al. Results from the TRIBE-AKI Study found associations between post-operative blood biomarkers and risk of chronic kidney disease after cardiac surgery. Kidney Int. 2021;99(3):716–24.PubMedCrossRef
13.
Zurück zum Zitat Menez S, Ju W, Menon R, Moledina DG, Thiessen Philbrook H, McArthur E, et al. Urinary EGF and MCP-1 and risk of CKD after cardiac surgery. JCI Insight. 2021;6(11):e147464. Menez S, Ju W, Menon R, Moledina DG, Thiessen Philbrook H, McArthur E, et al. Urinary EGF and MCP-1 and risk of CKD after cardiac surgery. JCI Insight. 2021;6(11):e147464.
14.
Zurück zum Zitat Hougardy JM, Revercez P, Pourcelet A, Oumeiri BE, Racape J, Le Moine A, et al. Chronic kidney disease as major determinant of the renal risk related to on-pump cardiac surgery: a single-center cohort study. Acta Chir Belg. 2016;116(4):217–24.PubMedCrossRef Hougardy JM, Revercez P, Pourcelet A, Oumeiri BE, Racape J, Le Moine A, et al. Chronic kidney disease as major determinant of the renal risk related to on-pump cardiac surgery: a single-center cohort study. Acta Chir Belg. 2016;116(4):217–24.PubMedCrossRef
15.
Zurück zum Zitat O'Neal JB, Shaw AD, Billings FT. Acute kidney injury following cardiac surgery: current understanding and future directions. Crit Care. 2016;20(1):187. O'Neal JB, Shaw AD, Billings FT. Acute kidney injury following cardiac surgery: current understanding and future directions. Crit Care. 2016;20(1):187.
16.
Zurück zum Zitat Machado MN, Nakazone MA, Maia LN. Prognostic value of acute kidney injury after cardiac surgery according to kidney disease: improving global outcomes definition and staging (KDIGO) criteria. PLoS ONE. 2014;9(5): e98028.PubMedPubMedCentralCrossRef Machado MN, Nakazone MA, Maia LN. Prognostic value of acute kidney injury after cardiac surgery according to kidney disease: improving global outcomes definition and staging (KDIGO) criteria. PLoS ONE. 2014;9(5): e98028.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Lysak N, Bihorac A, Hobson C. Mortality and cost of acute and chronic kidney disease after cardiac surgery. Curr Opin Anaesthesiol. 2017;30(1):113–7.PubMedPubMedCentralCrossRef Lysak N, Bihorac A, Hobson C. Mortality and cost of acute and chronic kidney disease after cardiac surgery. Curr Opin Anaesthesiol. 2017;30(1):113–7.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Alshaikh HN, Katz NM, Gani F, Nagarajan N, Canner JK, Kacker S, et al. Financial impact of acute kidney injury after cardiac operations in the United States. Ann Thorac Surg. 2018;105(2):469–75.PubMedCrossRef Alshaikh HN, Katz NM, Gani F, Nagarajan N, Canner JK, Kacker S, et al. Financial impact of acute kidney injury after cardiac operations in the United States. Ann Thorac Surg. 2018;105(2):469–75.PubMedCrossRef
19.
Zurück zum Zitat Ortega-Loubon C, Fernandez-Molina M, Carrascal-Hinojal Y, Fulquet-Carreras E. Cardiac surgery-associated acute kidney injury. Ann Card Anaesth. 2016;19(4):687–98.PubMedPubMedCentralCrossRef Ortega-Loubon C, Fernandez-Molina M, Carrascal-Hinojal Y, Fulquet-Carreras E. Cardiac surgery-associated acute kidney injury. Ann Card Anaesth. 2016;19(4):687–98.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Hu J, Chen R, Liu S, Yu X, Zou J, Ding X. Global incidence and outcomes of adult patients with acute kidney injury after cardiac surgery: a systematic review and meta-analysis. J Cardiothorac Vasc Anesth. 2016;30(1):82–9.PubMedCrossRef Hu J, Chen R, Liu S, Yu X, Zou J, Ding X. Global incidence and outcomes of adult patients with acute kidney injury after cardiac surgery: a systematic review and meta-analysis. J Cardiothorac Vasc Anesth. 2016;30(1):82–9.PubMedCrossRef
21.
Zurück zum Zitat Lassnigg A, Schmidlin D, Mouhieddine M, Bachmann LM, Druml W, Bauer P, et al. Minimal changes of serum creatinine predict prognosis in patients after cardiothoracic surgery: a prospective cohort study. J Am Soc Nephrol. 2004;15(6):1597–605.PubMedCrossRef Lassnigg A, Schmidlin D, Mouhieddine M, Bachmann LM, Druml W, Bauer P, et al. Minimal changes of serum creatinine predict prognosis in patients after cardiothoracic surgery: a prospective cohort study. J Am Soc Nephrol. 2004;15(6):1597–605.PubMedCrossRef
22.
Zurück zum Zitat Gaffney AM, Sladen RN. Acute kidney injury in cardiac surgery. Curr Opin Anaesthesiol. 2015;28(1):50–9.PubMedCrossRef Gaffney AM, Sladen RN. Acute kidney injury in cardiac surgery. Curr Opin Anaesthesiol. 2015;28(1):50–9.PubMedCrossRef
23.
Zurück zum Zitat Brown JR, Baker RA, Shore-Lesserson L, Fox AA, Mongero LB, Lobdell KW, et al. The Society of Thoracic Surgeons/Society of Cardiovascular Anesthesiologists/American Society of Extracorporeal Technology clinical practice guidelines for the prevention of adult cardiac surgery-associated acute kidney injury. Ann Thorac Surg. 2023;115(1):34–42.PubMedCrossRef Brown JR, Baker RA, Shore-Lesserson L, Fox AA, Mongero LB, Lobdell KW, et al. The Society of Thoracic Surgeons/Society of Cardiovascular Anesthesiologists/American Society of Extracorporeal Technology clinical practice guidelines for the prevention of adult cardiac surgery-associated acute kidney injury. Ann Thorac Surg. 2023;115(1):34–42.PubMedCrossRef
24.
Zurück zum Zitat Mao H, Katz N, Ariyanon W, Blanca-Martos L, Adybelli Z, Giuliani A, et al. Cardiac surgery-associated acute kidney injury. Cardiorenal Med. 2013;3(3):178–99.PubMedPubMedCentralCrossRef Mao H, Katz N, Ariyanon W, Blanca-Martos L, Adybelli Z, Giuliani A, et al. Cardiac surgery-associated acute kidney injury. Cardiorenal Med. 2013;3(3):178–99.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Zarbock A, Weiss R, Albert F, Rutledge K, Kellum JA, Bellomo R, et al. Epidemiology of surgery associated acute kidney injury (EPIS-AKI): a prospective international observational multi-center clinical study. Intensive Care Med. 2023;49(12):1441–55.PubMedPubMedCentralCrossRef Zarbock A, Weiss R, Albert F, Rutledge K, Kellum JA, Bellomo R, et al. Epidemiology of surgery associated acute kidney injury (EPIS-AKI): a prospective international observational multi-center clinical study. Intensive Care Med. 2023;49(12):1441–55.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Vives M, Hernandez A, Parramon F, Estanyol N, Pardina B, Munoz A, et al. Acute kidney injury after cardiac surgery: prevalence, impact and management challenges. Int J Nephrol Renovasc Dis. 2019;12:153–66.PubMedPubMedCentralCrossRef Vives M, Hernandez A, Parramon F, Estanyol N, Pardina B, Munoz A, et al. Acute kidney injury after cardiac surgery: prevalence, impact and management challenges. Int J Nephrol Renovasc Dis. 2019;12:153–66.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Uchino S, Kellum JA, Bellomo R, Doig GS, Morimatsu H, Morgera S, et al. Acute renal failure in critically ill patients: a multinational, multicenter study. JAMA. 2005;294(7):813–8.PubMedCrossRef Uchino S, Kellum JA, Bellomo R, Doig GS, Morimatsu H, Morgera S, et al. Acute renal failure in critically ill patients: a multinational, multicenter study. JAMA. 2005;294(7):813–8.PubMedCrossRef
28.
Zurück zum Zitat Meersch M, Weiss R, Strauss C, Albert F, Booke H, Forni L, et al. Acute kidney disease beyond day 7 after major surgery: a secondary analysis of the EPIS-AKI trial. Intensive Care Med. 2024;50(2):247–57.PubMedPubMedCentralCrossRef Meersch M, Weiss R, Strauss C, Albert F, Booke H, Forni L, et al. Acute kidney disease beyond day 7 after major surgery: a secondary analysis of the EPIS-AKI trial. Intensive Care Med. 2024;50(2):247–57.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Parolari A, Pesce LL, Pacini D, Mazzanti V, Salis S, Sciacovelli C, et al. Risk factors for perioperative acute kidney injury after adult cardiac surgery: role of perioperative management. Ann Thorac Surg. 2012;93(2):584–91.PubMedCrossRef Parolari A, Pesce LL, Pacini D, Mazzanti V, Salis S, Sciacovelli C, et al. Risk factors for perioperative acute kidney injury after adult cardiac surgery: role of perioperative management. Ann Thorac Surg. 2012;93(2):584–91.PubMedCrossRef
30.
Zurück zum Zitat Li Z, Fan G, Zheng X, Gong X, Chen T, Liu X, et al. Risk factors and clinical significance of acute kidney injury after on-pump or off-pump coronary artery bypass grafting: a propensity score-matched study. Interact Cardiovasc Thorac Surg. 2019;28(6):893–9.PubMedCrossRef Li Z, Fan G, Zheng X, Gong X, Chen T, Liu X, et al. Risk factors and clinical significance of acute kidney injury after on-pump or off-pump coronary artery bypass grafting: a propensity score-matched study. Interact Cardiovasc Thorac Surg. 2019;28(6):893–9.PubMedCrossRef
31.
Zurück zum Zitat Leballo G, Moutlana HJ, Muteba MK, Chakane PM. Factors associated with acute kidney injury and mortality during cardiac surgery. Cardiovasc J Afr. 2021;32(6):308–13.PubMedPubMedCentralCrossRef Leballo G, Moutlana HJ, Muteba MK, Chakane PM. Factors associated with acute kidney injury and mortality during cardiac surgery. Cardiovasc J Afr. 2021;32(6):308–13.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Zhang D, Teng J, Luo Z, Ding X, Jiang W. Risk factors and prognosis of acute kidney injury after cardiac surgery in patients with chronic kidney disease. Blood Purif. 2023;52(2):166–73.PubMedCrossRef Zhang D, Teng J, Luo Z, Ding X, Jiang W. Risk factors and prognosis of acute kidney injury after cardiac surgery in patients with chronic kidney disease. Blood Purif. 2023;52(2):166–73.PubMedCrossRef
33.
Zurück zum Zitat Wu VC, Huang TM, Lai CF, Shiao CC, Lin YF, Chu TS, et al. Acute-on-chronic kidney injury at hospital discharge is associated with long-term dialysis and mortality. Kidney Int. 2011;80(11):1222–30.PubMedCrossRef Wu VC, Huang TM, Lai CF, Shiao CC, Lin YF, Chu TS, et al. Acute-on-chronic kidney injury at hospital discharge is associated with long-term dialysis and mortality. Kidney Int. 2011;80(11):1222–30.PubMedCrossRef
34.
Zurück zum Zitat Casanova AG, Sancho-Martinez SM, Vicente-Vicente L, Ruiz Bueno P, Jorge-Monjas P, Tamayo E, et al. Diagnosis of cardiac surgery-associated acute kidney injury: state of the art and perspectives. J Clin Med. 2022;11(15):4576. Casanova AG, Sancho-Martinez SM, Vicente-Vicente L, Ruiz Bueno P, Jorge-Monjas P, Tamayo E, et al. Diagnosis of cardiac surgery-associated acute kidney injury: state of the art and perspectives. J Clin Med. 2022;11(15):4576.
35.
36.
Zurück zum Zitat Roy AK, Mc Gorrian C, Treacy C, Kavanaugh E, Brennan A, Mahon NG, et al. A comparison of traditional and novel definitions (RIFLE, AKIN, and KDIGO) of acute kidney injury for the prediction of outcomes in acute decompensated heart failure. Cardiorenal Med. 2013;3(1):26–37.PubMedPubMedCentralCrossRef Roy AK, Mc Gorrian C, Treacy C, Kavanaugh E, Brennan A, Mahon NG, et al. A comparison of traditional and novel definitions (RIFLE, AKIN, and KDIGO) of acute kidney injury for the prediction of outcomes in acute decompensated heart failure. Cardiorenal Med. 2013;3(1):26–37.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Atkinson MA, Ng DK, Warady BA, Furth SL, Flynn JT. The CKiD study: overview and summary of findings related to kidney disease progression. Pediatr Nephrol. 2021;36(3):527–38.PubMedCrossRef Atkinson MA, Ng DK, Warady BA, Furth SL, Flynn JT. The CKiD study: overview and summary of findings related to kidney disease progression. Pediatr Nephrol. 2021;36(3):527–38.PubMedCrossRef
38.
Zurück zum Zitat Inker LA, Eneanya ND, Coresh J, Tighiouart H, Wang D, Sang Y, et al. New creatinine- and cystatin C-based equations to estimate GFR without race. N Engl J Med. 2021;385(19):1737–49.PubMedPubMedCentralCrossRef Inker LA, Eneanya ND, Coresh J, Tighiouart H, Wang D, Sang Y, et al. New creatinine- and cystatin C-based equations to estimate GFR without race. N Engl J Med. 2021;385(19):1737–49.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Sharma A, Mucino MJ, Ronco C. Renal functional reserve and renal recovery after acute kidney injury. Nephron Clin Pract. 2014;127(1–4):94–100.PubMedCrossRef Sharma A, Mucino MJ, Ronco C. Renal functional reserve and renal recovery after acute kidney injury. Nephron Clin Pract. 2014;127(1–4):94–100.PubMedCrossRef
40.
Zurück zum Zitat Kellum JA, Romagnani P, Ashuntantang G, Ronco C, Zarbock A, Anders HJ. Acute kidney injury. Nat Rev Dis Primers. 2021;7(1):52.PubMedCrossRef Kellum JA, Romagnani P, Ashuntantang G, Ronco C, Zarbock A, Anders HJ. Acute kidney injury. Nat Rev Dis Primers. 2021;7(1):52.PubMedCrossRef
41.
Zurück zum Zitat Gunes-Altan M, Bosch A, Striepe K, Bramlage P, Schiffer M, Schmieder RE, et al. Is GFR decline induced by SGLT2 inhibitor of clinical importance? Cardiovasc Diabetol. 2024;23(1):184.PubMedPubMedCentralCrossRef Gunes-Altan M, Bosch A, Striepe K, Bramlage P, Schiffer M, Schmieder RE, et al. Is GFR decline induced by SGLT2 inhibitor of clinical importance? Cardiovasc Diabetol. 2024;23(1):184.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Ostermann M, Kunst G, Baker E, Weerapolchai K, Lumlertgul N. Cardiac surgery associated AKI prevention strategies and medical treatment for CSA-AKI. J Clin Med. 2021;10(22):5285. Ostermann M, Kunst G, Baker E, Weerapolchai K, Lumlertgul N. Cardiac surgery associated AKI prevention strategies and medical treatment for CSA-AKI. J Clin Med. 2021;10(22):5285.
43.
Zurück zum Zitat Ostermann M, Zarbock A, Goldstein S, Kashani K, Macedo E, Murugan R, et al. Recommendations on acute kidney injury biomarkers from the Acute Disease Quality Initiative Consensus Conference: a consensus statement. JAMA Netw Open. 2020;3(10): e2019209.PubMedCrossRef Ostermann M, Zarbock A, Goldstein S, Kashani K, Macedo E, Murugan R, et al. Recommendations on acute kidney injury biomarkers from the Acute Disease Quality Initiative Consensus Conference: a consensus statement. JAMA Netw Open. 2020;3(10): e2019209.PubMedCrossRef
44.
Zurück zum Zitat Kellum JA, Lameire N, Aspelin P, et al. KDIGO clinical practice guideline for acute kidney injury. Kidney Int Suppl. 2012;2(1):1–138. Kellum JA, Lameire N, Aspelin P, et al. KDIGO clinical practice guideline for acute kidney injury. Kidney Int Suppl. 2012;2(1):1–138.
45.
Zurück zum Zitat Okusa MD, Davenport A. Reading between the (guide)lines—the KDIGO practice guideline on acute kidney injury in the individual patient. Kidney Int. 2014;85(1):39–48.PubMedCrossRef Okusa MD, Davenport A. Reading between the (guide)lines—the KDIGO practice guideline on acute kidney injury in the individual patient. Kidney Int. 2014;85(1):39–48.PubMedCrossRef
46.
Zurück zum Zitat Husain-Syed F, Ferrari F, Sharma A, Danesi TH, Bezerra P, Lopez-Giacoman S, et al. Preoperative renal functional reserve predicts risk of acute kidney injury after cardiac operation. Ann Thorac Surg. 2018;105(4):1094–101.PubMedCrossRef Husain-Syed F, Ferrari F, Sharma A, Danesi TH, Bezerra P, Lopez-Giacoman S, et al. Preoperative renal functional reserve predicts risk of acute kidney injury after cardiac operation. Ann Thorac Surg. 2018;105(4):1094–101.PubMedCrossRef
47.
Zurück zum Zitat Cottam D, Azzopardi G, Forni LG. Biomarkers for early detection and predicting outcomes in acute kidney injury. Br J Hosp Med (Lond). 2022;83(8):1–11.PubMedCrossRef Cottam D, Azzopardi G, Forni LG. Biomarkers for early detection and predicting outcomes in acute kidney injury. Br J Hosp Med (Lond). 2022;83(8):1–11.PubMedCrossRef
48.
Zurück zum Zitat Macedo E, Bouchard J, Soroko SH, Chertow GM, Himmelfarb J, Ikizler TA, et al. Fluid accumulation, recognition and staging of acute kidney injury in critically-ill patients. Crit Care. 2010;14(3):R82.PubMedPubMedCentralCrossRef Macedo E, Bouchard J, Soroko SH, Chertow GM, Himmelfarb J, Ikizler TA, et al. Fluid accumulation, recognition and staging of acute kidney injury in critically-ill patients. Crit Care. 2010;14(3):R82.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Guzzi LM, Bergler T, Binnall B, Engelman DT, Forni L, Germain MJ, et al. Clinical use of [TIMP-2]*[IGFBP7] biomarker testing to assess risk of acute kidney injury in critical care: guidance from an expert panel. Crit Care. 2019;23(1):225.PubMedPubMedCentralCrossRef Guzzi LM, Bergler T, Binnall B, Engelman DT, Forni L, Germain MJ, et al. Clinical use of [TIMP-2]*[IGFBP7] biomarker testing to assess risk of acute kidney injury in critical care: guidance from an expert panel. Crit Care. 2019;23(1):225.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Gocze I, Jauch D, Gotz M, Kennedy P, Jung B, Zeman F, et al. Biomarker-guided intervention to prevent acute kidney injury after major surgery: the prospective randomized BigpAK study. Ann Surg. 2018;267(6):1013–20.PubMedCrossRef Gocze I, Jauch D, Gotz M, Kennedy P, Jung B, Zeman F, et al. Biomarker-guided intervention to prevent acute kidney injury after major surgery: the prospective randomized BigpAK study. Ann Surg. 2018;267(6):1013–20.PubMedCrossRef
51.
Zurück zum Zitat Meersch M, Schmidt C, Hoffmeier A, Van Aken H, Wempe C, Gerss J, et al. Prevention of cardiac surgery-associated AKI by implementing the KDIGO guidelines in high risk patients identified by biomarkers: the PrevAKI randomized controlled trial. Intensive Care Med. 2017;43(11):1551–61.PubMedPubMedCentralCrossRef Meersch M, Schmidt C, Hoffmeier A, Van Aken H, Wempe C, Gerss J, et al. Prevention of cardiac surgery-associated AKI by implementing the KDIGO guidelines in high risk patients identified by biomarkers: the PrevAKI randomized controlled trial. Intensive Care Med. 2017;43(11):1551–61.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Zarbock A, Kullmar M, Ostermann M, Lucchese G, Baig K, Cennamo A, et al. Prevention of cardiac surgery-associated acute kidney injury by implementing the KDIGO guidelines in high-risk patients identified by biomarkers: the PrevAKI-multicenter randomized controlled trial. Anesth Analg. 2021;133(2):292–302.PubMed Zarbock A, Kullmar M, Ostermann M, Lucchese G, Baig K, Cennamo A, et al. Prevention of cardiac surgery-associated acute kidney injury by implementing the KDIGO guidelines in high-risk patients identified by biomarkers: the PrevAKI-multicenter randomized controlled trial. Anesth Analg. 2021;133(2):292–302.PubMed
53.
Zurück zum Zitat Goldstein SL, Akcan-Arikan A, Afonso N, Askenazi DJ, Basalely AM, Basu RK, et al. Derivation and validation of an optimal neutrophil gelatinase-associated lipocalin cutoff to predict stage 2/3 acute kidney injury (AKI) in critically ill children. Kidney Int Rep. 2024;9(8):2443–52.PubMedPubMedCentralCrossRef Goldstein SL, Akcan-Arikan A, Afonso N, Askenazi DJ, Basalely AM, Basu RK, et al. Derivation and validation of an optimal neutrophil gelatinase-associated lipocalin cutoff to predict stage 2/3 acute kidney injury (AKI) in critically ill children. Kidney Int Rep. 2024;9(8):2443–52.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Volovelsky O, Terrell TC, Swain H, Bennett MR, Cooper DS, Goldstein SL. Pre-operative level of FGF23 predicts severe acute kidney injury after heart surgery in children. Pediatr Nephrol. 2018;33(12):2363–70.PubMedCrossRef Volovelsky O, Terrell TC, Swain H, Bennett MR, Cooper DS, Goldstein SL. Pre-operative level of FGF23 predicts severe acute kidney injury after heart surgery in children. Pediatr Nephrol. 2018;33(12):2363–70.PubMedCrossRef
55.
Zurück zum Zitat Birnie K, Verheyden V, Pagano D, Bhabra M, Tilling K, Sterne JA, et al. Predictive models for kidney disease: improving global outcomes (KDIGO) defined acute kidney injury in UK cardiac surgery. Crit Care. 2014;18(6):606.PubMedPubMedCentralCrossRef Birnie K, Verheyden V, Pagano D, Bhabra M, Tilling K, Sterne JA, et al. Predictive models for kidney disease: improving global outcomes (KDIGO) defined acute kidney injury in UK cardiac surgery. Crit Care. 2014;18(6):606.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Vives M, Candela A, Monedero P, Tamayo E, Hernandez A, Wijeysundera DN, et al. Improving the performance of the Cleveland Clinic Score for predicting acute kidney injury after cardiac surgery: a prospective multicenter cohort study. Minerva Anestesiol. 2024;90(4):245–53.PubMedCrossRef Vives M, Candela A, Monedero P, Tamayo E, Hernandez A, Wijeysundera DN, et al. Improving the performance of the Cleveland Clinic Score for predicting acute kidney injury after cardiac surgery: a prospective multicenter cohort study. Minerva Anestesiol. 2024;90(4):245–53.PubMedCrossRef
57.
Zurück zum Zitat Shahian DM, Jacobs JP, Badhwar V, Kurlansky PA, Furnary AP, Cleveland JC Jr, et al. The Society of Thoracic Surgeons 2018 adult cardiac surgery risk models: part 1-background, design considerations, and model development. Ann Thorac Surg. 2018;105(5):1411–8.PubMedCrossRef Shahian DM, Jacobs JP, Badhwar V, Kurlansky PA, Furnary AP, Cleveland JC Jr, et al. The Society of Thoracic Surgeons 2018 adult cardiac surgery risk models: part 1-background, design considerations, and model development. Ann Thorac Surg. 2018;105(5):1411–8.PubMedCrossRef
58.
Zurück zum Zitat O’Brien SM, Feng L, He X, Xian Y, Jacobs JP, Badhwar V, et al. The Society of Thoracic Surgeons 2018 adult cardiac surgery risk models: part 2-statistical methods and results. Ann Thorac Surg. 2018;105(5):1419–28.PubMed O’Brien SM, Feng L, He X, Xian Y, Jacobs JP, Badhwar V, et al. The Society of Thoracic Surgeons 2018 adult cardiac surgery risk models: part 2-statistical methods and results. Ann Thorac Surg. 2018;105(5):1419–28.PubMed
59.
Zurück zum Zitat Basu RK, Kaddourah A, Goldstein SL, Investigators AS. Assessment of a renal angina index for prediction of severe acute kidney injury in critically ill children: a multicentre, multinational, prospective observational study. Lancet Child Adolesc Health. 2018;2(2):112–20.PubMedPubMedCentralCrossRef Basu RK, Kaddourah A, Goldstein SL, Investigators AS. Assessment of a renal angina index for prediction of severe acute kidney injury in critically ill children: a multicentre, multinational, prospective observational study. Lancet Child Adolesc Health. 2018;2(2):112–20.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Goldstein SL, Krallman KA, Roy JP, Collins M, Chima RS, Basu RK, et al. Real-time acute kidney injury risk stratification-biomarker directed fluid management improves outcomes in critically ill children and young adults. Kidney Int Rep. 2023;8(12):2690–700.PubMedPubMedCentralCrossRef Goldstein SL, Krallman KA, Roy JP, Collins M, Chima RS, Basu RK, et al. Real-time acute kidney injury risk stratification-biomarker directed fluid management improves outcomes in critically ill children and young adults. Kidney Int Rep. 2023;8(12):2690–700.PubMedPubMedCentralCrossRef
61.
Zurück zum Zitat Yu Y, Li C, Zhu S, Jin L, Hu Y, Ling X, et al. Diagnosis, pathophysiology and preventive strategies for cardiac surgery-associated acute kidney injury: a narrative review. Eur J Med Res. 2023;28(1):45.PubMedPubMedCentralCrossRef Yu Y, Li C, Zhu S, Jin L, Hu Y, Ling X, et al. Diagnosis, pathophysiology and preventive strategies for cardiac surgery-associated acute kidney injury: a narrative review. Eur J Med Res. 2023;28(1):45.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Lannemyr L, Bragadottir G, Krumbholz V, Redfors B, Sellgren J, Ricksten S-E. Effects of cardiopulmonary bypass on renal perfusion, filtration, and oxygenation in patients undergoing cardiac surgery. Anesthesiology. 2017;126(2):205–13.PubMedCrossRef Lannemyr L, Bragadottir G, Krumbholz V, Redfors B, Sellgren J, Ricksten S-E. Effects of cardiopulmonary bypass on renal perfusion, filtration, and oxygenation in patients undergoing cardiac surgery. Anesthesiology. 2017;126(2):205–13.PubMedCrossRef
63.
Zurück zum Zitat Sharfuddin AA, Molitoris BA. Pathophysiology of ischemic acute kidney injury. Nat Rev Nephrol. 2011;7(4):189–200.PubMedCrossRef Sharfuddin AA, Molitoris BA. Pathophysiology of ischemic acute kidney injury. Nat Rev Nephrol. 2011;7(4):189–200.PubMedCrossRef
64.
Zurück zum Zitat Danobeitia JS, Djamali A, Fernandez LA. The role of complement in the pathogenesis of renal ischemia-reperfusion injury and fibrosis. Fibrogenesis Tissue Repair. 2014;7:16.PubMedPubMedCentralCrossRef Danobeitia JS, Djamali A, Fernandez LA. The role of complement in the pathogenesis of renal ischemia-reperfusion injury and fibrosis. Fibrogenesis Tissue Repair. 2014;7:16.PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Rabb H, Griffin MD, McKay DB, Swaminathan S, Pickkers P, Rosner MH, et al. Inflammation in AKI: current understanding, key questions, and knowledge gaps. J Am Soc Nephrol. 2016;27(2):371–9.PubMedCrossRef Rabb H, Griffin MD, McKay DB, Swaminathan S, Pickkers P, Rosner MH, et al. Inflammation in AKI: current understanding, key questions, and knowledge gaps. J Am Soc Nephrol. 2016;27(2):371–9.PubMedCrossRef
67.
Zurück zum Zitat Rosner MH, Okusa MD. Acute kidney injury associated with cardiac surgery. Clin J Am Soc Nephrol. 2006;1(1):19–32.PubMedCrossRef Rosner MH, Okusa MD. Acute kidney injury associated with cardiac surgery. Clin J Am Soc Nephrol. 2006;1(1):19–32.PubMedCrossRef
68.
Zurück zum Zitat Billings FT, Ball SK, Roberts LJ 2nd, Pretorius M. Postoperative acute kidney injury is associated with hemoglobinemia and an enhanced oxidative stress response. Free Radic Biol Med. 2011;50(11):1480–7.PubMedPubMedCentralCrossRef Billings FT, Ball SK, Roberts LJ 2nd, Pretorius M. Postoperative acute kidney injury is associated with hemoglobinemia and an enhanced oxidative stress response. Free Radic Biol Med. 2011;50(11):1480–7.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Bhirowo YP, Raksawardana YK, Setianto BY, Sudadi S, Tandean TN, Zaharo AF, et al. Hemolysis and cardiopulmonary bypass: meta-analysis and systematic review of contributing factors. J Cardiothorac Surg. 2023;18(1):291.PubMedPubMedCentralCrossRef Bhirowo YP, Raksawardana YK, Setianto BY, Sudadi S, Tandean TN, Zaharo AF, et al. Hemolysis and cardiopulmonary bypass: meta-analysis and systematic review of contributing factors. J Cardiothorac Surg. 2023;18(1):291.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Kumar AB, Suneja M. Cardiopulmonary bypass-associated acute kidney injury. Anesthesiology. 2011;114(4):964–70.PubMedCrossRef Kumar AB, Suneja M. Cardiopulmonary bypass-associated acute kidney injury. Anesthesiology. 2011;114(4):964–70.PubMedCrossRef
71.
Zurück zum Zitat Milne B, Gilbey T, De Somer F, Kunst G. Adverse renal effects associated with cardiopulmonary bypass. Perfusion. 2024;39(3):452–68.PubMedCrossRef Milne B, Gilbey T, De Somer F, Kunst G. Adverse renal effects associated with cardiopulmonary bypass. Perfusion. 2024;39(3):452–68.PubMedCrossRef
72.
Zurück zum Zitat Kanbay M, Copur S, Mizrak B, Ortiz A, Soler MJ. Intravenous fluid therapy in accordance with kidney injury risk: when to prescribe what volume of which solution. Clin Kidney J. 2023;16(4):684–92.PubMedCrossRef Kanbay M, Copur S, Mizrak B, Ortiz A, Soler MJ. Intravenous fluid therapy in accordance with kidney injury risk: when to prescribe what volume of which solution. Clin Kidney J. 2023;16(4):684–92.PubMedCrossRef
73.
Zurück zum Zitat Salahuddin N, Sammani M, Hamdan A, Joseph M, Al-Nemary Y, Alquaiz R, et al. Fluid overload is an independent risk factor for acute kidney injury in critically Ill patients: results of a cohort study. BMC Nephrol. 2017;18(1):45.PubMedPubMedCentralCrossRef Salahuddin N, Sammani M, Hamdan A, Joseph M, Al-Nemary Y, Alquaiz R, et al. Fluid overload is an independent risk factor for acute kidney injury in critically Ill patients: results of a cohort study. BMC Nephrol. 2017;18(1):45.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Lamy A, Devereaux PJ, Prabhakaran D, Taggart DP, Hu S, Paolasso E, et al. Off-pump or on-pump coronary-artery bypass grafting at 30 days. N Engl J Med. 2012;366(16):1489–97.PubMedCrossRef Lamy A, Devereaux PJ, Prabhakaran D, Taggart DP, Hu S, Paolasso E, et al. Off-pump or on-pump coronary-artery bypass grafting at 30 days. N Engl J Med. 2012;366(16):1489–97.PubMedCrossRef
75.
Zurück zum Zitat Cheungpasitporn W, Thongprayoon C, Kittanamongkolchai W, Srivali N, O’Corragain OA, Edmonds PJ, et al. Comparison of renal outcomes in off-pump versus on-pump coronary artery bypass grafting: a systematic review and meta-analysis of randomized controlled trials. Nephrology (Carlton). 2015;20(10):727–35.PubMedCrossRef Cheungpasitporn W, Thongprayoon C, Kittanamongkolchai W, Srivali N, O’Corragain OA, Edmonds PJ, et al. Comparison of renal outcomes in off-pump versus on-pump coronary artery bypass grafting: a systematic review and meta-analysis of randomized controlled trials. Nephrology (Carlton). 2015;20(10):727–35.PubMedCrossRef
76.
Zurück zum Zitat Puskas JD, Martin J, Cheng DC, Benussi S, Bonatti JO, Diegeler A, et al. ISMICS consensus conference and statements of randomized controlled trials of off-pump versus conventional coronary artery bypass surgery. Innovations (Phila). 2015;10(4):219–29.PubMedCrossRef Puskas JD, Martin J, Cheng DC, Benussi S, Bonatti JO, Diegeler A, et al. ISMICS consensus conference and statements of randomized controlled trials of off-pump versus conventional coronary artery bypass surgery. Innovations (Phila). 2015;10(4):219–29.PubMedCrossRef
77.
Zurück zum Zitat Merle NS, Church SE, Fremeaux-Bacchi V, Roumenina LT. Complement system part I—molecular mechanisms of activation and regulation. Front Immunol. 2015;6:262.PubMedPubMedCentralCrossRef Merle NS, Church SE, Fremeaux-Bacchi V, Roumenina LT. Complement system part I—molecular mechanisms of activation and regulation. Front Immunol. 2015;6:262.PubMedPubMedCentralCrossRef
78.
79.
80.
Zurück zum Zitat Merle NS, Boudhabhay I, Leon J, Fremeaux-Bacchi V, Roumenina LT. Complement activation during intravascular hemolysis: implication for sickle cell disease and hemolytic transfusion reactions. Transfus Clin Biol. 2019;26(2):116–24.PubMedCrossRef Merle NS, Boudhabhay I, Leon J, Fremeaux-Bacchi V, Roumenina LT. Complement activation during intravascular hemolysis: implication for sickle cell disease and hemolytic transfusion reactions. Transfus Clin Biol. 2019;26(2):116–24.PubMedCrossRef
81.
Zurück zum Zitat Klos A, Tenner AJ, Johswich KO, Ager RR, Reis ES, Kohl J. The role of the anaphylatoxins in health and disease. Mol Immunol. 2009;46(14):2753–66.PubMedPubMedCentralCrossRef Klos A, Tenner AJ, Johswich KO, Ager RR, Reis ES, Kohl J. The role of the anaphylatoxins in health and disease. Mol Immunol. 2009;46(14):2753–66.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Thurman JM, Ljubanovic D, Edelstein CL, Gilkeson GS, Holers VM. Lack of a functional alternative complement pathway ameliorates ischemic acute renal failure in mice. J Immunol. 2003;170(3):1517–23.PubMedCrossRef Thurman JM, Ljubanovic D, Edelstein CL, Gilkeson GS, Holers VM. Lack of a functional alternative complement pathway ameliorates ischemic acute renal failure in mice. J Immunol. 2003;170(3):1517–23.PubMedCrossRef
83.
Zurück zum Zitat Peng Q, Li K, Smyth LA, Xing G, Wang N, Meader L, et al. C3a and C5a promote renal ischemia-reperfusion injury. J Am Soc Nephrol. 2012;23(9):1474–85.PubMedPubMedCentralCrossRef Peng Q, Li K, Smyth LA, Xing G, Wang N, Meader L, et al. C3a and C5a promote renal ischemia-reperfusion injury. J Am Soc Nephrol. 2012;23(9):1474–85.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Kefalogianni R, Kamani F, Gaspar M, Aw TC, Donovan J, Laffan M, et al. Complement activation during cardiopulmonary bypass and association with clinical outcomes. EJHaem. 2022;3(1):86–96.PubMedPubMedCentralCrossRef Kefalogianni R, Kamani F, Gaspar M, Aw TC, Donovan J, Laffan M, et al. Complement activation during cardiopulmonary bypass and association with clinical outcomes. EJHaem. 2022;3(1):86–96.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Thurman JM, Lucia MS, Ljubanovic D, Holers VM. Acute tubular necrosis is characterized by activation of the alternative pathway of complement. Kidney Int. 2005;67(2):524–30.PubMedCrossRef Thurman JM, Lucia MS, Ljubanovic D, Holers VM. Acute tubular necrosis is characterized by activation of the alternative pathway of complement. Kidney Int. 2005;67(2):524–30.PubMedCrossRef
86.
Zurück zum Zitat Stenson EK, Kendrick J, Dixon B, Thurman JM. The complement system in pediatric acute kidney injury. Pediatr Nephrol. 2023;38(5):1411–25.PubMedCrossRef Stenson EK, Kendrick J, Dixon B, Thurman JM. The complement system in pediatric acute kidney injury. Pediatr Nephrol. 2023;38(5):1411–25.PubMedCrossRef
87.
Zurück zum Zitat Sun H, Xie Q, Peng Z. Does fenoldopam protect kidney in cardiac surgery? A systemic review and meta-analysis with trial sequential analysis. Shock. 2019;52(3):326–33.PubMedCrossRef Sun H, Xie Q, Peng Z. Does fenoldopam protect kidney in cardiac surgery? A systemic review and meta-analysis with trial sequential analysis. Shock. 2019;52(3):326–33.PubMedCrossRef
88.
Zurück zum Zitat Mehta RH, Leimberger JD, van Diepen S, Meza J, Wang A, Jankowich R, et al. Levosimendan in patients with left ventricular dysfunction undergoing cardiac surgery. N Engl J Med. 2017;376(21):2032–42.PubMedCrossRef Mehta RH, Leimberger JD, van Diepen S, Meza J, Wang A, Jankowich R, et al. Levosimendan in patients with left ventricular dysfunction undergoing cardiac surgery. N Engl J Med. 2017;376(21):2032–42.PubMedCrossRef
89.
Zurück zum Zitat Wang B, He X, Gong Y, Cheng B. Levosimendan in patients with left ventricular dysfunction undergoing cardiac surgery: an update meta-analysis and trial sequential analysis. Biomed Res Int. 2018;2018:7563083.PubMedPubMedCentral Wang B, He X, Gong Y, Cheng B. Levosimendan in patients with left ventricular dysfunction undergoing cardiac surgery: an update meta-analysis and trial sequential analysis. Biomed Res Int. 2018;2018:7563083.PubMedPubMedCentral
90.
Zurück zum Zitat Landoni G, Lomivorotov VV, Alvaro G, Lobreglio R, Pisano A, Guarracino F, et al. Levosimendan for hemodynamic support after cardiac surgery. N Engl J Med. 2017;376(21):2021–31.PubMedCrossRef Landoni G, Lomivorotov VV, Alvaro G, Lobreglio R, Pisano A, Guarracino F, et al. Levosimendan for hemodynamic support after cardiac surgery. N Engl J Med. 2017;376(21):2021–31.PubMedCrossRef
91.
Zurück zum Zitat Barba-Navarro R, Tapia-Silva M, Garza-Garcia C, Lopez-Giacoman S, Melgoza-Toral I, Vazquez-Rangel A, et al. The effect of spironolactone on acute kidney injury after cardiac surgery: a randomized, placebo-controlled trial. Am J Kidney Dis. 2017;69(2):192–9.PubMedCrossRef Barba-Navarro R, Tapia-Silva M, Garza-Garcia C, Lopez-Giacoman S, Melgoza-Toral I, Vazquez-Rangel A, et al. The effect of spironolactone on acute kidney injury after cardiac surgery: a randomized, placebo-controlled trial. Am J Kidney Dis. 2017;69(2):192–9.PubMedCrossRef
92.
Zurück zum Zitat Tamargo C, Hanouneh M, Cervantes CE. Treatment of acute kidney injury: a review of current approaches and emerging innovations. J Clin Med. 2024;13(9):2455. Tamargo C, Hanouneh M, Cervantes CE. Treatment of acute kidney injury: a review of current approaches and emerging innovations. J Clin Med. 2024;13(9):2455.
93.
Zurück zum Zitat Van den Eynde J, Cloet N, Van Lerberghe R, Sa M, Vlasselaers D, Toelen J, et al. Strategies to prevent acute kidney injury after pediatric cardiac surgery: a network meta-analysis. Clin J Am Soc Nephrol. 2021;16(10):1480–90.PubMedPubMedCentralCrossRef Van den Eynde J, Cloet N, Van Lerberghe R, Sa M, Vlasselaers D, Toelen J, et al. Strategies to prevent acute kidney injury after pediatric cardiac surgery: a network meta-analysis. Clin J Am Soc Nephrol. 2021;16(10):1480–90.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Khwaja A. KDIGO clinical practice guidelines for acute kidney injury. Nephron Clin Pract. 2012;120(4):c179–84.PubMedCrossRef Khwaja A. KDIGO clinical practice guidelines for acute kidney injury. Nephron Clin Pract. 2012;120(4):c179–84.PubMedCrossRef
95.
Zurück zum Zitat Kullmar M, Weiss R, Ostermann M, Campos S, Grau Novellas N, Thomson G, et al. A multinational observational study exploring adherence with the kidney disease: improving global outcomes recommendations for prevention of acute kidney injury after cardiac surgery. Anesth Analg. 2020;130(4):910–6.PubMedCrossRef Kullmar M, Weiss R, Ostermann M, Campos S, Grau Novellas N, Thomson G, et al. A multinational observational study exploring adherence with the kidney disease: improving global outcomes recommendations for prevention of acute kidney injury after cardiac surgery. Anesth Analg. 2020;130(4):910–6.PubMedCrossRef
96.
Zurück zum Zitat Brown JK, Shaw AD, Mythen MG, Guzzi L, Reddy VS, Crisafi C, et al. Adult cardiac surgery-associated acute kidney injury: joint consensus report. J Cardiothorac Vasc Anesth. 2023;37(9):1579–90.PubMedCrossRef Brown JK, Shaw AD, Mythen MG, Guzzi L, Reddy VS, Crisafi C, et al. Adult cardiac surgery-associated acute kidney injury: joint consensus report. J Cardiothorac Vasc Anesth. 2023;37(9):1579–90.PubMedCrossRef
97.
Zurück zum Zitat Rizo-Topete LM, Rosner MH, Ronco C. Acute kidney injury risk assessment and the nephrology rapid response team. Blood Purif. 2017;43(1–3):82–8.PubMedCrossRef Rizo-Topete LM, Rosner MH, Ronco C. Acute kidney injury risk assessment and the nephrology rapid response team. Blood Purif. 2017;43(1–3):82–8.PubMedCrossRef
98.
Zurück zum Zitat Ranucci M, Johnson I, Willcox T, Baker RA, Boer C, Baumann A, et al. Goal-directed perfusion to reduce acute kidney injury: A randomized trial. J Thorac Cardiovasc Surg. 2018;156(5):1918-27 e2.PubMedCrossRef Ranucci M, Johnson I, Willcox T, Baker RA, Boer C, Baumann A, et al. Goal-directed perfusion to reduce acute kidney injury: A randomized trial. J Thorac Cardiovasc Surg. 2018;156(5):1918-27 e2.PubMedCrossRef
99.
Zurück zum Zitat Hariri G, Collet L, Duarte L, Martin GL, Resche-Rigon M, Lebreton G, et al. Prevention of cardiac surgery-associated acute kidney injury: a systematic review and meta-analysis of non-pharmacological interventions. Crit Care. 2023;27(1):354.PubMedPubMedCentralCrossRef Hariri G, Collet L, Duarte L, Martin GL, Resche-Rigon M, Lebreton G, et al. Prevention of cardiac surgery-associated acute kidney injury: a systematic review and meta-analysis of non-pharmacological interventions. Crit Care. 2023;27(1):354.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Wever KE, Menting TP, Rovers M, van der Vliet JA, Rongen GA, Masereeuw R, et al. Ischemic preconditioning in the animal kidney, a systematic review and meta-analysis. PLoS ONE. 2012;7(2): e32296.PubMedPubMedCentralCrossRef Wever KE, Menting TP, Rovers M, van der Vliet JA, Rongen GA, Masereeuw R, et al. Ischemic preconditioning in the animal kidney, a systematic review and meta-analysis. PLoS ONE. 2012;7(2): e32296.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Zhou C, Bulluck H, Fang N, Li L, Hausenloy DJ. Age and surgical complexity impact on renoprotection by remote ischemic preconditioning during adult cardiac surgery: a meta analysis. Sci Rep. 2017;7(1):215.PubMedPubMedCentralCrossRef Zhou C, Bulluck H, Fang N, Li L, Hausenloy DJ. Age and surgical complexity impact on renoprotection by remote ischemic preconditioning during adult cardiac surgery: a meta analysis. Sci Rep. 2017;7(1):215.PubMedPubMedCentralCrossRef
102.
Zurück zum Zitat Kottenberg E, Thielmann M, Bergmann L, Heine T, Jakob H, Heusch G, et al. Protection by remote ischemic preconditioning during coronary artery bypass graft surgery with isoflurane but not propofol—a clinical trial. Acta Anaesthesiol Scand. 2012;56(1):30–8.PubMedCrossRef Kottenberg E, Thielmann M, Bergmann L, Heine T, Jakob H, Heusch G, et al. Protection by remote ischemic preconditioning during coronary artery bypass graft surgery with isoflurane but not propofol—a clinical trial. Acta Anaesthesiol Scand. 2012;56(1):30–8.PubMedCrossRef
103.
Zurück zum Zitat Zarbock A, Schmidt C, Van Aken H, Wempe C, Martens S, Zahn PK, et al. Effect of remote ischemic preconditioning on kidney injury among high-risk patients undergoing cardiac surgery: a randomized clinical trial. JAMA. 2015;313(21):2133–41.PubMedCrossRef Zarbock A, Schmidt C, Van Aken H, Wempe C, Martens S, Zahn PK, et al. Effect of remote ischemic preconditioning on kidney injury among high-risk patients undergoing cardiac surgery: a randomized clinical trial. JAMA. 2015;313(21):2133–41.PubMedCrossRef
104.
Zurück zum Zitat Weiss R, Meersch M, Wempe C, von Groote T, Agervald T, Zarbock A. Recombinant alpha-1-microglobulin (RMC-035) to prevent acute kidney injury in cardiac surgery patients: phase 1b evaluation of safety and pharmacokinetics. Kidney Int Rep. 2023;8(5):980–8.PubMedPubMedCentralCrossRef Weiss R, Meersch M, Wempe C, von Groote T, Agervald T, Zarbock A. Recombinant alpha-1-microglobulin (RMC-035) to prevent acute kidney injury in cardiac surgery patients: phase 1b evaluation of safety and pharmacokinetics. Kidney Int Rep. 2023;8(5):980–8.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Tasanarong A, Duangchana S, Sumransurp S, Homvises B, Satdhabudha O. Prophylaxis with erythropoietin versus placebo reduces acute kidney injury and neutrophil gelatinase-associated lipocalin in patients undergoing cardiac surgery: a randomized, double-blind controlled trial. BMC Nephrol. 2013;14:136.PubMedPubMedCentralCrossRef Tasanarong A, Duangchana S, Sumransurp S, Homvises B, Satdhabudha O. Prophylaxis with erythropoietin versus placebo reduces acute kidney injury and neutrophil gelatinase-associated lipocalin in patients undergoing cardiac surgery: a randomized, double-blind controlled trial. BMC Nephrol. 2013;14:136.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Tie HT, Luo MZ, Lin D, Zhang M, Wan JY, Wu QC. Erythropoietin administration for prevention of cardiac surgery-associated acute kidney injury: a meta-analysis of randomized controlled trials. Eur J Cardiothorac Surg. 2015;48(1):32–9.PubMedCrossRef Tie HT, Luo MZ, Lin D, Zhang M, Wan JY, Wu QC. Erythropoietin administration for prevention of cardiac surgery-associated acute kidney injury: a meta-analysis of randomized controlled trials. Eur J Cardiothorac Surg. 2015;48(1):32–9.PubMedCrossRef
107.
Zurück zum Zitat Penny-Dimri JC, Cochrane AD, Perry LA, Smith JA. characterising the role of perioperative erythropoietin for preventing acute kidney injury after cardiac surgery: systematic review and meta-analysis. Heart Lung Circ. 2016;25(11):1067–76.PubMedCrossRef Penny-Dimri JC, Cochrane AD, Perry LA, Smith JA. characterising the role of perioperative erythropoietin for preventing acute kidney injury after cardiac surgery: systematic review and meta-analysis. Heart Lung Circ. 2016;25(11):1067–76.PubMedCrossRef
108.
Zurück zum Zitat Sharif S, Chen B, Brewster P, Chen T, Dworkin L, Gong R. Rationale and design of assessing the effectiveness of short-term low-dose lithium therapy in averting cardiac surgery-associated acute kidney injury: a randomized, double blinded, placebo controlled pilot trial. Front Med (Lausanne). 2021;8: 639402.PubMedPubMedCentralCrossRef Sharif S, Chen B, Brewster P, Chen T, Dworkin L, Gong R. Rationale and design of assessing the effectiveness of short-term low-dose lithium therapy in averting cardiac surgery-associated acute kidney injury: a randomized, double blinded, placebo controlled pilot trial. Front Med (Lausanne). 2021;8: 639402.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Pickkers P, Angus DC, Bass K, Bellomo R, van den Berg E, Bernholz J, et al. Phase-3 trial of recombinant human alkaline phosphatase for patients with sepsis-associated acute kidney injury (REVIVAL). Intensive Care Med. 2024;50(1):68–78.PubMedPubMedCentralCrossRef Pickkers P, Angus DC, Bass K, Bellomo R, van den Berg E, Bernholz J, et al. Phase-3 trial of recombinant human alkaline phosphatase for patients with sepsis-associated acute kidney injury (REVIVAL). Intensive Care Med. 2024;50(1):68–78.PubMedPubMedCentralCrossRef
110.
Zurück zum Zitat Landoni G, Monaco F, Ti LK, Baiardo Redaelli M, Bradic N, Comis M, et al. A randomized trial of intravenous amino acids for kidney protection. N Engl J Med. 2024;391(8):687–98.PubMedCrossRef Landoni G, Monaco F, Ti LK, Baiardo Redaelli M, Bradic N, Comis M, et al. A randomized trial of intravenous amino acids for kidney protection. N Engl J Med. 2024;391(8):687–98.PubMedCrossRef
111.
Zurück zum Zitat Navaei AH, Shekerdemian LS, Mohammad MA, Coss-Bu JA, Bastero P, Ettinger NA, et al. Derangement of arginine and related amino acids in children undergoing surgery for congenital heart disease with cardiopulmonary bypass. Crit Care Explor. 2020;2(7): e0150.PubMedPubMedCentralCrossRef Navaei AH, Shekerdemian LS, Mohammad MA, Coss-Bu JA, Bastero P, Ettinger NA, et al. Derangement of arginine and related amino acids in children undergoing surgery for congenital heart disease with cardiopulmonary bypass. Crit Care Explor. 2020;2(7): e0150.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Roberts DM, Sevastos J, Carland JE, Stocker SL, Lea-Henry TN. Clinical pharmacokinetics in kidney disease: application to rational design of dosing regimens. Clin J Am Soc Nephrol. 2018;13(8):1254–63.PubMedPubMedCentralCrossRef Roberts DM, Sevastos J, Carland JE, Stocker SL, Lea-Henry TN. Clinical pharmacokinetics in kidney disease: application to rational design of dosing regimens. Clin J Am Soc Nephrol. 2018;13(8):1254–63.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Smith PK, Carrier M, Chen JC, Haverich A, Levy JH, Menasche P, et al. Effect of pexelizumab in coronary artery bypass graft surgery with extended aortic cross-clamp time. Ann Thorac Surg. 2006;82(3):781–8 (discussion 8–9).PubMedCrossRef Smith PK, Carrier M, Chen JC, Haverich A, Levy JH, Menasche P, et al. Effect of pexelizumab in coronary artery bypass graft surgery with extended aortic cross-clamp time. Ann Thorac Surg. 2006;82(3):781–8 (discussion 8–9).PubMedCrossRef
114.
Zurück zum Zitat Haverich A, Shernan SK, Levy JH, Chen JC, Carrier M, Taylor KM, et al. Pexelizumab reduces death and myocardial infarction in higher risk cardiac surgical patients. Ann Thorac Surg. 2006;82(2):486–92.PubMedCrossRef Haverich A, Shernan SK, Levy JH, Chen JC, Carrier M, Taylor KM, et al. Pexelizumab reduces death and myocardial infarction in higher risk cardiac surgical patients. Ann Thorac Surg. 2006;82(2):486–92.PubMedCrossRef
115.
Zurück zum Zitat Smith PK, Shernan SK, Chen JC, Carrier M, Verrier ED, Adams PX, et al. Effects of C5 complement inhibitor pexelizumab on outcome in high-risk coronary artery bypass grafting: combined results from the PRIMO-CABG I and II trials. J Thorac Cardiovasc Surg. 2011;142(1):89–98.PubMedCrossRef Smith PK, Shernan SK, Chen JC, Carrier M, Verrier ED, Adams PX, et al. Effects of C5 complement inhibitor pexelizumab on outcome in high-risk coronary artery bypass grafting: combined results from the PRIMO-CABG I and II trials. J Thorac Cardiovasc Surg. 2011;142(1):89–98.PubMedCrossRef
116.
Zurück zum Zitat Kulasekararaj AG, Hill A, Rottinghaus ST, Langemeijer S, Wells R, Gonzalez-Fernandez FA, et al. Ravulizumab (ALXN1210) vs eculizumab in C5-inhibitor-experienced adult patients with PNH: the 302 study. Blood. 2019;133(6):540–9.PubMedPubMedCentralCrossRef Kulasekararaj AG, Hill A, Rottinghaus ST, Langemeijer S, Wells R, Gonzalez-Fernandez FA, et al. Ravulizumab (ALXN1210) vs eculizumab in C5-inhibitor-experienced adult patients with PNH: the 302 study. Blood. 2019;133(6):540–9.PubMedPubMedCentralCrossRef
117.
Zurück zum Zitat Lee JW, Sicre de Fontbrune F, Wong Lee Lee L, Pessoa V, Gualandro S, Fureder W, et al. Ravulizumab (ALXN1210) vs eculizumab in adult patients with PNH naive to complement inhibitors: the 301 study. Blood. 2019;133(6):530–9. Lee JW, Sicre de Fontbrune F, Wong Lee Lee L, Pessoa V, Gualandro S, Fureder W, et al. Ravulizumab (ALXN1210) vs eculizumab in adult patients with PNH naive to complement inhibitors: the 301 study. Blood. 2019;133(6):530–9.
118.
Zurück zum Zitat Nishimura JI, Kawaguchi T, Ito S, Murai H, Shimono A, Matsuda T, et al. Real-world safety profile of eculizumab in patients with paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, or generalized myasthenia gravis: an integrated analysis of post-marketing surveillance in Japan. Int J Hematol. 2023;118(4):419–31.PubMedCrossRef Nishimura JI, Kawaguchi T, Ito S, Murai H, Shimono A, Matsuda T, et al. Real-world safety profile of eculizumab in patients with paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, or generalized myasthenia gravis: an integrated analysis of post-marketing surveillance in Japan. Int J Hematol. 2023;118(4):419–31.PubMedCrossRef
119.
Zurück zum Zitat Hillmen P, Young NS, Schubert J, Brodsky RA, Socie G, Muus P, et al. The complement inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med. 2006;355(12):1233–43.PubMedCrossRef Hillmen P, Young NS, Schubert J, Brodsky RA, Socie G, Muus P, et al. The complement inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med. 2006;355(12):1233–43.PubMedCrossRef
120.
Zurück zum Zitat Rondeau E, Scully M, Ariceta G, Barbour T, Cataland S, Heyne N, et al. The long-acting C5 inhibitor, ravulizumab, is effective and safe in adult patients with atypical hemolytic uremic syndrome naive to complement inhibitor treatment. Kidney Int. 2020;97(6):1287–96.PubMedCrossRef Rondeau E, Scully M, Ariceta G, Barbour T, Cataland S, Heyne N, et al. The long-acting C5 inhibitor, ravulizumab, is effective and safe in adult patients with atypical hemolytic uremic syndrome naive to complement inhibitor treatment. Kidney Int. 2020;97(6):1287–96.PubMedCrossRef
121.
Zurück zum Zitat Kulasekararaj AG, Griffin M, Langemeijer S, Usuki K, Kulagin A, Ogawa M, et al. Long-term safety and efficacy of ravulizumab in patients with paroxysmal nocturnal hemoglobinuria: 2-year results from two pivotal phase 3 studies. Eur J Haematol. 2022;109(3):205–14.PubMedPubMedCentralCrossRef Kulasekararaj AG, Griffin M, Langemeijer S, Usuki K, Kulagin A, Ogawa M, et al. Long-term safety and efficacy of ravulizumab in patients with paroxysmal nocturnal hemoglobinuria: 2-year results from two pivotal phase 3 studies. Eur J Haematol. 2022;109(3):205–14.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Kulasekararaj A, Schrezenmeier H, Usuki K, Kulagin A, Gualandro SF, Notaro R, et al. Ravulizumab provides durable control of intravascular hemolysis and improves survival in patients with paroxysmal nocturnal hemoglobinuria: long-term follow-up of study 301 and comparisons with patients of the international PNH registry. Blood. 2023;142(Suppl 1):2714.CrossRef Kulasekararaj A, Schrezenmeier H, Usuki K, Kulagin A, Gualandro SF, Notaro R, et al. Ravulizumab provides durable control of intravascular hemolysis and improves survival in patients with paroxysmal nocturnal hemoglobinuria: long-term follow-up of study 301 and comparisons with patients of the international PNH registry. Blood. 2023;142(Suppl 1):2714.CrossRef
123.
Zurück zum Zitat Fam S, Werneburg B, Pandya S, et al. Clinical and real-world pharmacovigilance data of meningococcal infections in eculizumab- or ravulizumab-treated patients. J Neurol Sci. 2023;455: 121883.CrossRef Fam S, Werneburg B, Pandya S, et al. Clinical and real-world pharmacovigilance data of meningococcal infections in eculizumab- or ravulizumab-treated patients. J Neurol Sci. 2023;455: 121883.CrossRef
124.
Zurück zum Zitat McCullough JW, Renner B, Thurman JM. The role of the complement system in acute kidney injury. Semin Nephrol. 2013;33(6):543–56.PubMedCrossRef McCullough JW, Renner B, Thurman JM. The role of the complement system in acute kidney injury. Semin Nephrol. 2013;33(6):543–56.PubMedCrossRef
125.
Zurück zum Zitat von Groote T, Meersch M, Romagnoli S, Ostermann M, Ripolles-Melchor J, Schneider AG, et al. Biomarker-guided intervention to prevent acute kidney injury after major surgery (BigpAK-2 trial): study protocol for an international, prospective, randomised controlled multicentre trial. BMJ Open. 2023;13(3): e070240.CrossRef von Groote T, Meersch M, Romagnoli S, Ostermann M, Ripolles-Melchor J, Schneider AG, et al. Biomarker-guided intervention to prevent acute kidney injury after major surgery (BigpAK-2 trial): study protocol for an international, prospective, randomised controlled multicentre trial. BMJ Open. 2023;13(3): e070240.CrossRef
Metadaten
Titel
Kidney Injury Following Cardiac Surgery: A Review of Our Current Understanding
verfasst von
Christine-Elena Kamla
Melanie Meersch-Dini
Lilian Monteiro Pereira Palma
Publikationsdatum
12.01.2025
Verlag
Springer International Publishing
Erschienen in
American Journal of Cardiovascular Drugs
Print ISSN: 1175-3277
Elektronische ISSN: 1179-187X
DOI
https://doi.org/10.1007/s40256-024-00715-8

Kompaktes Leitlinien-Wissen Innere Medizin (Link öffnet in neuem Fenster)

Mit medbee Pocketcards schnell und sicher entscheiden.
Leitlinien-Wissen kostenlos und immer griffbereit auf ihrem Desktop, Handy oder Tablet.

Neu im Fachgebiet Kardiologie

Blutdruck am Belastungsende zeigt kardiovaskuläres Risiko

Diverse Blutdruckparameter während der Ergometrie sind schon auf ihre Aussagekraft, Mortalität und kardiovaskuläres Risiko betreffend, hin abgeklopft worden. Nun hat auch das Druckverhalten am Ende der Belastung im Fokus einer Studie gestanden.

Neue US-Leitlinie zum Management bei akutem Koronarsyndrom

In den USA ist die Leitlinie zum Management bei akutem Koronarsyndrom aktualisiert worden. In Europa erfolgte das neueste Guideline-Update dazu bereits 2023. Gehen beide Leitlinien konform oder gibt es nennenswerte Differenzen? 

Mechanische Herzklappe beschert jüngeren Betroffenen längeres Leben

Patienten und Patientinnen bevorzugen bioprothetische Herzklappen gegenüber mechanischen Klappenprothesen. Diese Wahl könnte sich zumindest für jüngere Patienten nachteilig auswirken: Ihnen bietet eine mechanische Klappe anscheinend einen Überlebensvorteil.

Lp(a) wird zu selten gemessen

Laut europäischer Dyslipidämie-Leitlinie sollte die Bestimmung der Serumkonzentration von Lipoprotein (a) mindestens einmal im Leben jedes Erwachsenen erwogen werden. Tatsächlich wird die Messung selbst kardiovaskulär gefährdeten Personen nur selten zuteil.

EKG Essentials: EKG befunden mit System (Link öffnet in neuem Fenster)

In diesem CME-Kurs können Sie Ihr Wissen zur EKG-Befundung anhand von zwölf Video-Tutorials auffrischen und 10 CME-Punkte sammeln.
Praxisnah, relevant und mit vielen Tipps & Tricks vom Profi.

Update Kardiologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.