Acessibilidade / Reportar erro

Does dexmedetomidine reduce the risk of acute kidney injury after cardiac surgery? A meta-analysis of randomized controlled trials

Abstract

Background:

Acute Kidney Injury (AKI) is a common complication after cardiac surgery and has been associated with poor outcomes. Dexmedetomidine (DEX) has been shown to confer direct renoprotection based on some animal and clinical studies, but data from other trials came to the opposite conclusion following cardiac surgery. This meta-analysis was conducted to evaluate the effects of perioperative DEX administration on the occurrence of AKI and the outcomes after cardiac surgery.

Methods:

We searched databases including EMBASE, PubMed, and Cochrane CENTRAL for Randomized Controlled Trials (RCTs) focused on DEX for AKI in adult patients after cardiac surgery. The primary outcome was incidence of AKI. Secondary outcomes were Mechanical Ventilation (MV) duration, Intensive Care Unit (ICU) Length Of Stay (LOS), hospital LOS and mortality.

Results:

Fifteen trials enrolling 2907 study patients were collected in the meta-analyses. Compared with controls, DEX reduced the incidence of postoperative AKI (Odds Ratio [OR = 0.66]; 95% Confidence Interval [95% CI 0.48–0.91]; p = 0.01), and there was no significant difference between groups in postoperative mortality (OR = 0.63; 95% CI 0.32–1.26; p = 0.19), MV duration (Weighted Mean Difference [WMD = -0.44]; 95% CI -1.50–0.63; p = 0.42), ICU LOS (WMD = -1.19; 95% CI -2.89-0.51; p = 0.17), and hospital LOS (WMD = -0.31; 95% CI -0.76–0.15; p = 0.19).

Conclusions:

Perioperative DEX reduced the incidence of postoperative AKI in adult patients undergoing cardiac surgery. No significant decrease existed in mortality, MV duration, ICU LOS and hospital LOS owing to DEX administration.

KEYWORDS
Acute kidney injury; Cardiac surgery; Dexmedetomidine; Meta-analysis

Introduction

Acute Kidney Injury (AKI) is a recognized complication following cardiac surgery with a reported incidence between 5% and 42%.11 Wang Y, Bellomo R. Cardiac surgery-associated acute kidney injury: risk factors, pathophysiology and treatment. Nat Rev Nephrol. 2017;13:697-711. Postoperative AKI results in poor outcomes, prolonged hospital Length of Stay (LOS), increased hospital costs and mortality.22 Ortega-Loubon C, Fernández-Molina M, Carrascal-Hinojal Y, et al. Cardiac surgery-associated acute kidney injury. Ann Card Anaesth. 2016;19:687–98. The mechanism of AKI after cardiac surgery is tightly associated with the hemodynamic instability and sympathetic activity during Cardiopulmonary Bypass (CPB).33 Guan C, Li C, Xu L, et al. Risk factors of cardiac surgery-associated acute kidney injury: development and validation of a perioperative predictive nomogram. J Nephrol. 2019;32:937–45.,44 Ranucci M, Ambrogi F, Pistuddi V. Cardiac surgery associated acute kidney injury and the role of cardiopulmonary bypass technique. J Thorac Cardiovasc Surg. 2019;157:301.,55 Liu D, Liu B, Liang Z, et al. Acute Kidney Injury following Cardiopulmonary Bypass: A Challenging Picture. Oxid Med Cell Longev. 2021;2021:8873581. Although numerous trials attempted to identify strategies to prevent AKI, the incidence is still around 40% and no definite strategy exists yet.66 Meersch M, Schmidt C, Hoffmeier A, 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;43:1551–61.,77 Küllmar M, Zarbock A, Engelman DT, et al. Prevention of acute kidney injury. Crit Care Clin. 2020;36:691–704.,88 Couturier C, Maillard N, Mariat C, et al. Prevention of cardiac surgery-associated acute kidney injury by risk stratification using (TIMP-2)*(IGFBP7). Biomark Med. 2021;15:1201–10.,99 Presta P, Bolignano D, Coppolino G, et al. Antecedent ACE-inhibition, inflammatory response, and cardiac surgery associated acute kidney injury. Rev Cardiovasc Med. 2021 ;22:207–13.,1010 Li S, Fu S, Xiao Y, et al. Recent Perioperative Pharmacological Prevention of Acute Kidney Injury after Cardiac Surgery: A Narrative Review. Am J Cardiovasc Drugs. 2017;17:17–25.

Dexmedetomidine (DEX) is a highly selective α2 adrenore-ceptor agonist and has been widely used for sedation during cardiac surgery. DEX differs from other sedatives by the properties of anti-inflammatory and sympatholytics.1111 Weerink MAS, Struys M, Hannivoort LN, et al. Clinical Pharmacokinetics and Pharmacodynamics of Dexmedetomidine. Clin Pharmacokinet. 2017;56:893–913.,1212 Gallego-Ligorit L, Vives M, Vallés-Torres J, et al. Use of Dexmedetomidine in Cardiothoracic and Vascular Anesthesia. J Cardiothorac Vasc Anesth. 2018;32:1426–38. These properties offer a hypothesis that DEX might reduce the incidence of postoperative AKI. Preclinical studies indicated the renoprotective effect of DEX in various animal models.1313 Zhao Y, Feng X, Li B, et al. Dexmedetomidine Protects Against Lipopolysaccharide-lnduced Acute Kidney Injury by Enhancing Autophagy Through Inhibition of the PI3K/AKT/mTOR Pathway. Front Pharmacol. 2020;11:128.,1414 Feng X, Guan W, Zhao Y, et al. Dexmedetomidine ameliorates lipopolysaccharide-induced acute kidney injury in rats by inhibiting inflammation and oxidative stress via the GSK-3β/Nrf2 signaling pathway. J Cell Physiol. 2019;234:18994–9009.,1515 Wang Z, Wu J, Hu Z, et al. Dexmedetomidine Alleviates Lipopolysaccharide-lnduced Acute Kidney Injury by Inhibiting p75NTR-Mediated Oxidative Stress and Apoptosis. Oxid Med Cell Longev. 2020;2020:5454210. Several single-center Randomized Controlled Trials (RCTs) have also addressed this question and the results are controversial.1616 Tang C, Hu Y, Gao J, et al. Dexmedetomidine pretreatment attenuates myocardial ischemia reperfusion induced acute kidney injury and endoplasmic reticulum stress in human and rat. Life Sci. 2020;257:118004.,1717 Cho JS, Shim JK, Soh S, et al. Perioperative dexmedetomidine reduces the incidence and severity of acute kidney injury following valvular heart surgery. Kidney Int. 2016;89:693–700.,1818 Balkanay OO, Goksedef D, Omeroglu SN, et al. The dose-related effects of dexmedetomidine on renal functions and serum neutrophil gelatinase-associated lipocalin values after coronary artery bypass grafting: a randomized, triple-blind, placebo-controlled study. Interact Cardiovasc Thorac Surg. 2015;20: 209–14.,1919 Zhai M, Kang F, Han M, et al. The effect of dexmedetomidine on renal function in patients undergoing cardiac valve replacement under cardiopulmonary bypass: A double-blind randomized controlled trial. J Clin Anesth. 2017;40:33–8. Previous meta-analyses had evaluated the effect of DEX in cardiac surgery and showed a reduced risk of postoperative AKI.2020 Peng K, Li D, Applegate 2nd RL, et al. Effect of dexmedetomidine on cardiac surgery-associated acute kidney injury: a metaanalysis with trial sequential analysis of randomized controlled trials. J Cardiothorac Vasc Anesth. 2020;34:603–13.,2121 Liu Y, Sheng B, Wang S, et al. Dexmedetomidine prevents acute kidney injury after adult cardiac surgery: a meta-analysis of randomized controlled trials. BMC Anesthesiol. 2018;18:7.,2222 Chen X, Huang T, Cao X, et al. Comparative efficacy of drugs for preventing acute kidney injury after cardiac surgery: a network meta-analysis. Am J Cardiovasc Drugs. 2018;18:49–58. However, the studies were limited by high heterogeneity and relatively small sample size. Moreover, some strengthened studies focused on this issue were published in recent years.2323 Turan A, Duncan A, Leung S, et al. Dexmedetomidine for reduction of atrial fibrillation and delirium after cardiac surgery (DECADE): a randomised placebo-controlled trial. Lancet. 2020;396:177–85.,2424 Subramaniam B, Shankar P, Shaefi S, et al. Effect of intravenous acetaminophen vs placebo combined with propofol or dexmedetomidine on postoperative delirium among older patients following cardiac surgery: The DEXACET Randomized Clinical Trial. Jama. 2019;321:686–96. Therefore, we conducted this meta-analysis to assess if DEX is associated with a protective effect of AKI after cardiac surgery.

Methods

Search strategy and study criteria

This meta-analysis was performed according to the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-analyses) guidelines2525 Moher D, Liberati A, Tetzlaff J, et al. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6:e1000097. and three electronic databases including MEDLINE (through PubMed), Embase (through OVID) and Cochrane Library were searched to identify relevant studies. The search strategy for PubMed was performed using the keywords “dexmedetomidine”, “cardiac surgery”, “heart surgery”, “kidney”, and “renal”. Various combinations of key words and different search strategies were developed for another two databases. The search encompassed the period between January 1997 and November 2022. All eligible studies met the following conditions: 1) Randomized controlled trials only, and as an original article, 2) Studies published in English, 3) Adult patients undergoing cardiac surgery with or without cardiopulmonary bypass, including coronary artery bypass graft or cardiac valve replacement or coronary artery bypass graft combined with cardiac valve replacement; 4) Intervention: DEX; 5) Comparison: placebo or control (other therapy); 6) Outcome measure: the incidence of postoperative AKI. Exclusion criteria were as follows: retrospective study, observational study, conference abstracts, expert opinion, review articles, case reports, abstracts, editorials, and letters to the editor, animal studies, studies involving pediatric population, and studies lacking clinical outcome data, and failure to contact the authors. Furthermore, the references of relevant studies were also assessed.

Literature review and data extraction

The literature review and data extraction were independently completed by 2 investigators. In the case of duplicate records pertaining to a single study, we considered the PubMed database to take precedence. Disagreements were handled by discussion to reach consensus. Quality assessment was completed using the Cochrane risk of bias tool: randomization, allocation concealment, blinding, withdrawals and dropouts, and intention-to-treat analysis. Data extraction included characteristics of included studies and patients.

Postoperative outcomes

The primary end point was incidence of AKI defined based on three definitions, consisting of KDIGO (Kidney Disease: Improving Global Outcome), RIFLE (Risk, Injury, Failure, Loss of kidney function, and End-stage kidney disease), AKIN (Acute Kidney Injury Network) and undergoing RRT (Renal Replacement Therapy) for new onset of AKI after cardiac surgery. Secondary outcomes included mortality, Mechanical Ventilation (MV) duration, ICU LOS, and hospital LOS.

Statistical analysis

For dichotomous outcomes (reported with incidence), we calculated the Odds Ratio (OR) with 95% Confidence Interval (95% CI). For continuous outcomes (reported as mean ± standard deviation, median and interquartile range, or median and range), we calculated mean differences for each study according to the statistical method of Hozo et al.2626 Hozo SP, Djulbegovic B, Hozo I. Estimating the mean and variance from the median, range, and the size of a sample. BMC Med Res Methodol. 2005;5:13. and used weights to pool the estimate (Weighted Mean Difference – WMD) with 95% CI. Random-effect models were used to analyze the data in light of the heterogeneity. Heterogeneity was assessed with Inconsistency statistic (I22 Ortega-Loubon C, Fernández-Molina M, Carrascal-Hinojal Y, et al. Cardiac surgery-associated acute kidney injury. Ann Card Anaesth. 2016;19:687–98.). Publication bias was assessed by Begg’s test, Egger’s test and Macaskill test. Meta-regression and subgroup analysis were conducted to explore the potential sources of significant heterogeneity. Sensitivity analyses were used to assess the robustness of our results by removing each included study at one time to obtain and evaluate the remaining overall estimates: p < 0.05 (2 sided) was considered to be statistically significant for hypothesis testing. All statistical analyses were performed in REVMAN (version 5.0; Cochrane Collaboration, Oxford, UK) and Stata (version 15.0; Stata-Corp LP).

Results

Study characteristics

Figure 1 shows the flow chart for the study screening and selection process in this meta-analysis. Fifteen trials with sixteen groups of data ultimately met our criteria.1616 Tang C, Hu Y, Gao J, et al. Dexmedetomidine pretreatment attenuates myocardial ischemia reperfusion induced acute kidney injury and endoplasmic reticulum stress in human and rat. Life Sci. 2020;257:118004.,1818 Balkanay OO, Goksedef D, Omeroglu SN, et al. The dose-related effects of dexmedetomidine on renal functions and serum neutrophil gelatinase-associated lipocalin values after coronary artery bypass grafting: a randomized, triple-blind, placebo-controlled study. Interact Cardiovasc Thorac Surg. 2015;20: 209–14.,1919 Zhai M, Kang F, Han M, et al. The effect of dexmedetomidine on renal function in patients undergoing cardiac valve replacement under cardiopulmonary bypass: A double-blind randomized controlled trial. J Clin Anesth. 2017;40:33–8.,2323 Turan A, Duncan A, Leung S, et al. Dexmedetomidine for reduction of atrial fibrillation and delirium after cardiac surgery (DECADE): a randomised placebo-controlled trial. Lancet. 2020;396:177–85.,2424 Subramaniam B, Shankar P, Shaefi S, et al. Effect of intravenous acetaminophen vs placebo combined with propofol or dexmedetomidine on postoperative delirium among older patients following cardiac surgery: The DEXACET Randomized Clinical Trial. Jama. 2019;321:686–96.,2727 Djaiani G, Silverton N, Fedorko L, et al. Dexmedetomidine versus Propofol Sedation Reduces Delirium after Cardiac Surgery: A Randomized Controlled Trial. Anesthesiology. 2016;124:362–8.,2828 Li X, Yang J, Nie XL, et al. Impact of dexmedetomidine on the incidence of delirium in elderly patients after cardiac surgery: A randomized controlled trial. PLoS One. 2017;12:e0170757.,2929 Liu X, Zhang K, Wang W, et al. Dexmedetomidine sedation reduces atrial fibrillation after cardiac surgery compared to propofol: a randomized controlled trial. Crit Care. 2016;20:298.,3030 Park JB, Bang SH, Chee HK, et al. Efficacy and safety of dexmedetomidine for postoperative delirium in adult cardiac surgery on cardiopulmonary bypass. Korean J Thorac Cardiovasc Surg. 2014;47:249–54.,3131 Shehabi Y, Grant P, Wolfenden H, et al. Prevalence of delirium with dexmedetomidine compared with morphine-based therapy after cardiac surgery: a randomized controlled trial (DEXmedetomidine COmpared to Morphine-DEXCOM Study). Anesthesiology. 2009; 111:1075–84.,3232 Shi C, Jin J, Qiao L, et al. Effect of perioperative administration of dexmedetomidine on delirium after cardiac surgery in elderly patients: a double-blinded, multi-center, randomized study. Clin Interv Aging. 2019;14:571–5.,3333 Soliman R, Zohry G. The myocardial protective effect of dexmedetomidine in high-risk patients undergoing aortic vascular surgery. Ann Card Anaesth. 2016;19:606–13.,3434 Zi J, Fan Y, Dong C, et al. Anxiety Administrated by dexmedetomidine to prevent new-onset of postoperative atrial fibrillation in patients undergoing off-pump coronary artery bypass graft. Int Heart J. 2020;61:263–72.,3535 Likhvantsev VV, Landoni G, Grebenchikov OA, et al. Perioperative dexmedetomidine supplement decreases delirium incidence after adult cardiac surgery: a randomized, double-blind, controlled study. J Cardiothorac Vasc Anesth. 2021 ;35:449–57. Two studies were for coronary artery bypass grafting, nine were for combined cardiac surgery, two for valve replacement surgery, and two for aortic vascular surgery. Nine trials used placebo as control, whereas four used propofol, one used morphine or remifentanil. DEX was continuously infused at a rate of approximately 0.2 to 0.8 mcg.kg–1.h–1 for 24 hours after a loading dose (0.4–1 mcg.kg–1) in six studies or infused at a rate of approximately 0.04 to 1.5 mcg.kg–1.h–1 without a loading dose in nine. DEX was used intraoperatively in eleven studies and postoperatively in four.

Figure 1
Flow diagram of studies included into meta-analyses.

For primary outcomes, AKI incidence was reported in fifteen trials, including two showing the number of patients needed for dialysis owing to the new onset of AKI after cardiac surgery; For second endpoint, mortality, in seven; mechanical ventilation duration, in twelve; ICU LOS, in thirteen; and hospital LOS, in ten.

Study design and patient characteristics are summarized in Tables 1 and 2. The quality assessment is listed in Figure 2 and Table 3.

Figure 2
Quality assessment of studies included into metaanalyses.

Table 1
Summarized study design of included randomized trials.
Table 2
Summarized patient characteristics of the included randomized trials.
Table 3
Summarized quality assessment of included randomized trials.

Effect of DEX on incidence of AKI, and mortality

The outcome of AKI was reported in 2907 study participants, and the overall incidence was 7.95% (DEX group, 6.52%; control group, 9.37%). The postoperative incidence of AKI was significantly reduced by DEX (fifteen studies; OR = 0.66; 95% CI 0.48–0.91; p = 0.01; I2 = 6%); (Fig. 3). There was no evidence of publication bias (Begg’s test p = 0.96; Egger’s test p = 0.55).

Figure 3
DEX reduced the incidence of AKI.

Subgroup analyses revealed similar trends to those of postoperative AKI outcome based on different characteristics such as age (≥ 62.5 vs. < 62.5 years), male proportion (≥ 62% vs. < 62%), diabetes proportion (≥ 25% vs. < 25%), hypertension proportion (≥ 25% vs. < 25%), previous Myocardial Infarction (Ml) proportion (≥ 15% vs. < 15%), Left Ventricular Ejection Fraction (LVEF) (≥ 60% vs. < 60%), Cardiopulmonary Bypass (CPB) duration (≥ 100 vs. < 100 min), β-blocker (≥ 50% vs. < 50%), Statin (≥ 65% vs. < 65%), loading dose (use or not), type of control (placebo vs. others), administration timing (pre/intraoperative vs. postoperative) and surgery type (combined surgery vs. others) (Supplementary Table 1).

Meta-regression analyses performed for the potential sources of significant heterogeneity are listed in Supplementary Table 2, and there were no significant differences for postoperative AKI in all the subgroups.

Sensitivity analyses excluding each included study at a time revealed that all the studies were consistent with the direction and size of the overall AKI-reducing effect of DEX (p < 0.05 for all) except Cho.

The outcome mortality was reported in 1883 study participants, and the overall incidence was 1.86% (DEX group, 1.38%; control group, 2.34%). There was no significant difference between DEX and the risk of mortality (Seven studies; OR = 0.63; 95% CI 0.32–1.26; p = 0.19; I2 = 0%); (Supplementary Figure 1).

Effect of DEX on MV duration, ICU LOS, and hospital LOS

Postoperative MV duration was reported in twelve studies, and no statistically significant reduction by DEX was found (eleven studies; WMD = -0.44; 95% CI -1.50–0.63; p = 0.42; I2 = 73%); (Supplementary Fig. 2). There was no significant difference in ICU LOS (thirteen studies; WMD = -1.19; 95% CI -2.89–0.51; p = 0.17; l2=74%); (Supplementary Fig. 3), as well as in hospital LOS (ten studies; WMD = -0.31; 95% CI -0.76–0.15; p = 0.19; I2 = 76%); (Supplementary Fig. 4).

Discussion

In this meta-analysis of fifteen RCTs involving 2907 adult patients undergoing cardiac surgery, we found that perioperative DEX was associated with a decrease in postoperative AKI. However, postoperative parameters including MV duration, ICU, hospital LOS and mortality did not seem to present a significant reduction as a result of the DEX.

AKI is common after cardiac surgery and small increases in postoperative serum creatinine levels have been reported to be related with worse outcome, even when renal function returns to normal ultimately.3636 Lau G, Wald R, Sladen R, et al. Acute kidney injury in cardiac surgery and cardiac intensive care. Semin Cardiothorac Vasc Anesth. 2015;19:270–87.,3737 Lassnigg A, Schmid ER, Hiesmayr M, et al. Impact of minimal increases in serum creatinine on outcome in patients after cardiothoracic surgery: do we have to revise current definitions of acute renal failure? Crit Care Med. 2008;36:1129–37. The reason that cardiac surgery can cause AKI is always accompanied by renal Ischemia-Reperfusion Injury (I/RI), elevated sympathetic activity, and hemodynamic instability. For this reason, pharmacologic or other prophylaxis which have these properties may reduce AKI after cardiac surgery and this is an important research area to clinicians.3838 Brown JR, Shore-Lesserson L, Fox AA, 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. J Extra CorporTechnol. 2022;54:267–90.,3939 Yu Y, Li C, Zhu S, et al. Diagnosis, pathophysiology, and preventive strategies for cardiac surgery-associated acute kidney injury: a narrative review. Eur J Med Res. 2023;28:45.,4040 Peng K, Mcllroy DR, Bollen BA, et al. Society of Cardiovascular Anesthesiologists Clinical Practice Update for Management of Acute Kidney Injury Associated with Cardiac Surgery. Anesth Analg. 2022;135:744–56.

DEX has been widely used in anesthesia procedures and has shown organ protection by stabilizing the sympathetic system, exerting anti-inflammatory effects, and attenuating Ischemia/Reperfusion (I/R) injury in vivo and vitro studies.4141 Ueki M, Kawasaki T, Habe K, et al. The effects of dexmedetomidine on inflammatory mediators after cardiopulmonary bypass. Anaesthesia. 2014;69:693–700.,4545 Yang SJ, Fan CN, Wang MJ, et al. Effects of dexmedetomidine on renal microcirculation in ischemia/reperfusion-induced acute kidney injury in rats. Sci Rep. 2021;11:2026. There is a hypothesis that the incidence of AKI may be reduced owing to the use of DEX in cardiac surgery.4646 Honore PM, De Bels D, Preseau T, Spapen HD. Dexmedetomidine: the first new kid on the block for preventing cardiac surgery-associated acute kidney injury? Crit Care. 2018;22:151.,4747 Li S, Fu S, Xiao Y, Xu G. Recent Perioperative Pharmacological Prevention of Acute Kidney Injury after Cardiac Surgery: A Narrative Review [published correction appears in Am J Cardiovasc Drugs. 2018 Feb 26] Am J Cardiovasc Drugs. 2017;17:17–25. Several studies have compared the efficacy of DEX at enhancing urine output and at decreasing the concentration of blood urea nitrogen and creatinine after surgery,1919 Zhai M, Kang F, Han M, et al. The effect of dexmedetomidine on renal function in patients undergoing cardiac valve replacement under cardiopulmonary bypass: A double-blind randomized controlled trial. J Clin Anesth. 2017;40:33–8.,4848 Wu S, Yao H, Cheng N, Guo N, Chen J, Ge M, Cai J. Determining whether dexmedetomidine provides a reno-protective effect in patients receiving laparoscopic radical prostatectomy: a pilot study. Int Urol Nephrol. 2019;51:1553–61.,4949 Wu F, Duan H, Xie Y. Preventive effects of dexmedetomidine on renal dysfunction and hemodynamic stability in malignant obstructive jaundice patients during peri-operative period. Med Sci Monit. 2019;25:6782–7. and other randomized controlled trials have reported a lower rate of kidney injury.1717 Cho JS, Shim JK, Soh S, et al. Perioperative dexmedetomidine reduces the incidence and severity of acute kidney injury following valvular heart surgery. Kidney Int. 2016;89:693–700.,5050 Soh S, Shim JK, Song JW, Bae JC, Kwak YL. Effect of dexmedetomidine on acute kidney injury after aortic surgery: a single-centre, placebo-controlled, randomised controlled trial. Br J Anaesth. 2020. https://doi.org/10.1016/j.bja.2019.12.036. Online ahead of print.
https://doi.org/10.1016/j.bja.2019.12.03...
,5151 Bayram A, Ulgey A, Baykan A, Narin N, Narin F, Esmaoglu A, Boyaci A. The effects of dexmedetomidine on early stage renal functions in pediatric patients undergoing cardiac angiography using non-ionic contrast media: a double-blind, randomized clinical trial. Paediatr Anaesth. 2014;24:426–32. No general consensus was reached on the effect of DEX for AKI.5252 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;10:5285.,5353 Massoth C, Zarbock A, Meersch M. Acute Kidney Injury in Cardiac Surgery. Crit Care Clin. 2021 ;37:267–78.,5454 Küllmar M, Zarbock A, Engelman DT, Chatterjee S, Wagner NM. Prevention of acute kidney injury. Crit Care Clin. 2020;36:691–704. A few meta-analyses have been conducted to address this issue. However, a meta-analysis performed by Peng,2020 Peng K, Li D, Applegate 2nd RL, et al. Effect of dexmedetomidine on cardiac surgery-associated acute kidney injury: a metaanalysis with trial sequential analysis of randomized controlled trials. J Cardiothorac Vasc Anesth. 2020;34:603–13. which included nine RCTs with a total of 1308 patients, showed low heterogeneity (I2 = 30%). Another meta-analysis by Liu[21] including ten RCTs with a total of 1575 patients showed only eight groups of data from seven studies on the main outcome. Our study with an almost two times larger sample size collected some high-quality research published in recent years and provided a more convincing conclusion.

Based on our literature review, positive reno-protective effects were reported in two studies. Moreover, in our data analysis, the combined results with a random-effects model revealed lower AKI incidence in patients with DEX, and the pooled OR succeeded to reach statistical significance. However, this benefit did not translate into the second outcomes, such as MV duration, ICU LOS, hospital LOS and mortality. A possible explanation is that our meta-analysis with a relatively small sample size may account for such differences. Another is that heterogeneity for the MV duration, ICU and hospital LOS is almost over 50%. In fact, there are trends toward lower MV duration, ICU LOS, hospital LOS and mortality. Further randomized studies with large sample sizes are encouraged to verify the current findings.

Our analysis has several limitations. Firstly, many factors could influence AKI after cardiac surgery, such as age, degree of hypertension, and drugs used for treating hypertension and diabetes mellitus. We were unable to access individual patient data, so the influences of confounding factors may be underestimated. Secondly, we only included English language trials and published studies, which may lead to publication bias. Thirdly, many design differences among these studies made it difficult to reduce clinical heterogeneity. Subgroup analyses and meta-regression were performed for the potential sources of heterogeneity. Finally, based on the included data, there are four different definitions of AKI, including RIFLE, AKIN, KDIGO, and need for RRT. Six studies did not mention the definition of AKI. According to previous studies,5555 Zeng X, McMahon GM, Brunelli SM, Bates DW, Waikar SS. Incidence, outcomes, and comparisons across definitions of AKI in hospitalized individuals. Clin J Am Soc Nephrol. 2014;9:12–20.,5656 Koeze J, Keus F, Dieperink W, van der Horst IC, Zijlstra JG, van Meurs M. Incidence, timing and outcome of AKI in critically ill patients varies with the definition used and the addition of urine output criteria. BMC Nephrol. 2017;18:70. the incidence of AKI can vary greatly according to the definition used, and our study might draw a misleading conclusion. Given only three or less studies were included, a subgroup analysis based on AKI definition was not performed.

Conclusion

In summary, our meta-analysis indicated that perioperative DEX use reduced postoperative AKI in patients receiving cardiac surgery. However, DEX use is not associated with MV duration, ICU LOS, hospital LOS and mortality. Future, much larger trials are needed to verify the current findings.

Data availability statement

The data used to support the findings of this study are included within the supplementary information file.

  • Funding statement
    This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
  • Ethical statement
    Since this was a meta-analysis, ethical approval was not required under the arrangements of the Institutional Review Board in our hospital.

Supplementary materials

Supplementary material associated with this article can be found in the online version at doi:10.1016/j.bjane.2023.07.003.

References

  • 1
    Wang Y, Bellomo R. Cardiac surgery-associated acute kidney injury: risk factors, pathophysiology and treatment. Nat Rev Nephrol. 2017;13:697-711.
  • 2
    Ortega-Loubon C, Fernández-Molina M, Carrascal-Hinojal Y, et al. Cardiac surgery-associated acute kidney injury. Ann Card Anaesth. 2016;19:687–98.
  • 3
    Guan C, Li C, Xu L, et al. Risk factors of cardiac surgery-associated acute kidney injury: development and validation of a perioperative predictive nomogram. J Nephrol. 2019;32:937–45.
  • 4
    Ranucci M, Ambrogi F, Pistuddi V. Cardiac surgery associated acute kidney injury and the role of cardiopulmonary bypass technique. J Thorac Cardiovasc Surg. 2019;157:301.
  • 5
    Liu D, Liu B, Liang Z, et al. Acute Kidney Injury following Cardiopulmonary Bypass: A Challenging Picture. Oxid Med Cell Longev. 2021;2021:8873581.
  • 6
    Meersch M, Schmidt C, Hoffmeier A, 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;43:1551–61.
  • 7
    Küllmar M, Zarbock A, Engelman DT, et al. Prevention of acute kidney injury. Crit Care Clin. 2020;36:691–704.
  • 8
    Couturier C, Maillard N, Mariat C, et al. Prevention of cardiac surgery-associated acute kidney injury by risk stratification using (TIMP-2)*(IGFBP7). Biomark Med. 2021;15:1201–10.
  • 9
    Presta P, Bolignano D, Coppolino G, et al. Antecedent ACE-inhibition, inflammatory response, and cardiac surgery associated acute kidney injury. Rev Cardiovasc Med. 2021 ;22:207–13.
  • 10
    Li S, Fu S, Xiao Y, et al. Recent Perioperative Pharmacological Prevention of Acute Kidney Injury after Cardiac Surgery: A Narrative Review. Am J Cardiovasc Drugs. 2017;17:17–25.
  • 11
    Weerink MAS, Struys M, Hannivoort LN, et al. Clinical Pharmacokinetics and Pharmacodynamics of Dexmedetomidine. Clin Pharmacokinet. 2017;56:893–913.
  • 12
    Gallego-Ligorit L, Vives M, Vallés-Torres J, et al. Use of Dexmedetomidine in Cardiothoracic and Vascular Anesthesia. J Cardiothorac Vasc Anesth. 2018;32:1426–38.
  • 13
    Zhao Y, Feng X, Li B, et al. Dexmedetomidine Protects Against Lipopolysaccharide-lnduced Acute Kidney Injury by Enhancing Autophagy Through Inhibition of the PI3K/AKT/mTOR Pathway. Front Pharmacol. 2020;11:128.
  • 14
    Feng X, Guan W, Zhao Y, et al. Dexmedetomidine ameliorates lipopolysaccharide-induced acute kidney injury in rats by inhibiting inflammation and oxidative stress via the GSK-3β/Nrf2 signaling pathway. J Cell Physiol. 2019;234:18994–9009.
  • 15
    Wang Z, Wu J, Hu Z, et al. Dexmedetomidine Alleviates Lipopolysaccharide-lnduced Acute Kidney Injury by Inhibiting p75NTR-Mediated Oxidative Stress and Apoptosis. Oxid Med Cell Longev. 2020;2020:5454210.
  • 16
    Tang C, Hu Y, Gao J, et al. Dexmedetomidine pretreatment attenuates myocardial ischemia reperfusion induced acute kidney injury and endoplasmic reticulum stress in human and rat. Life Sci. 2020;257:118004.
  • 17
    Cho JS, Shim JK, Soh S, et al. Perioperative dexmedetomidine reduces the incidence and severity of acute kidney injury following valvular heart surgery. Kidney Int. 2016;89:693–700.
  • 18
    Balkanay OO, Goksedef D, Omeroglu SN, et al. The dose-related effects of dexmedetomidine on renal functions and serum neutrophil gelatinase-associated lipocalin values after coronary artery bypass grafting: a randomized, triple-blind, placebo-controlled study. Interact Cardiovasc Thorac Surg. 2015;20: 209–14.
  • 19
    Zhai M, Kang F, Han M, et al. The effect of dexmedetomidine on renal function in patients undergoing cardiac valve replacement under cardiopulmonary bypass: A double-blind randomized controlled trial. J Clin Anesth. 2017;40:33–8.
  • 20
    Peng K, Li D, Applegate 2nd RL, et al. Effect of dexmedetomidine on cardiac surgery-associated acute kidney injury: a metaanalysis with trial sequential analysis of randomized controlled trials. J Cardiothorac Vasc Anesth. 2020;34:603–13.
  • 21
    Liu Y, Sheng B, Wang S, et al. Dexmedetomidine prevents acute kidney injury after adult cardiac surgery: a meta-analysis of randomized controlled trials. BMC Anesthesiol. 2018;18:7.
  • 22
    Chen X, Huang T, Cao X, et al. Comparative efficacy of drugs for preventing acute kidney injury after cardiac surgery: a network meta-analysis. Am J Cardiovasc Drugs. 2018;18:49–58.
  • 23
    Turan A, Duncan A, Leung S, et al. Dexmedetomidine for reduction of atrial fibrillation and delirium after cardiac surgery (DECADE): a randomised placebo-controlled trial. Lancet. 2020;396:177–85.
  • 24
    Subramaniam B, Shankar P, Shaefi S, et al. Effect of intravenous acetaminophen vs placebo combined with propofol or dexmedetomidine on postoperative delirium among older patients following cardiac surgery: The DEXACET Randomized Clinical Trial. Jama. 2019;321:686–96.
  • 25
    Moher D, Liberati A, Tetzlaff J, et al. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6:e1000097.
  • 26
    Hozo SP, Djulbegovic B, Hozo I. Estimating the mean and variance from the median, range, and the size of a sample. BMC Med Res Methodol. 2005;5:13.
  • 27
    Djaiani G, Silverton N, Fedorko L, et al. Dexmedetomidine versus Propofol Sedation Reduces Delirium after Cardiac Surgery: A Randomized Controlled Trial. Anesthesiology. 2016;124:362–8.
  • 28
    Li X, Yang J, Nie XL, et al. Impact of dexmedetomidine on the incidence of delirium in elderly patients after cardiac surgery: A randomized controlled trial. PLoS One. 2017;12:e0170757.
  • 29
    Liu X, Zhang K, Wang W, et al. Dexmedetomidine sedation reduces atrial fibrillation after cardiac surgery compared to propofol: a randomized controlled trial. Crit Care. 2016;20:298.
  • 30
    Park JB, Bang SH, Chee HK, et al. Efficacy and safety of dexmedetomidine for postoperative delirium in adult cardiac surgery on cardiopulmonary bypass. Korean J Thorac Cardiovasc Surg. 2014;47:249–54.
  • 31
    Shehabi Y, Grant P, Wolfenden H, et al. Prevalence of delirium with dexmedetomidine compared with morphine-based therapy after cardiac surgery: a randomized controlled trial (DEXmedetomidine COmpared to Morphine-DEXCOM Study). Anesthesiology. 2009; 111:1075–84.
  • 32
    Shi C, Jin J, Qiao L, et al. Effect of perioperative administration of dexmedetomidine on delirium after cardiac surgery in elderly patients: a double-blinded, multi-center, randomized study. Clin Interv Aging. 2019;14:571–5.
  • 33
    Soliman R, Zohry G. The myocardial protective effect of dexmedetomidine in high-risk patients undergoing aortic vascular surgery. Ann Card Anaesth. 2016;19:606–13.
  • 34
    Zi J, Fan Y, Dong C, et al. Anxiety Administrated by dexmedetomidine to prevent new-onset of postoperative atrial fibrillation in patients undergoing off-pump coronary artery bypass graft. Int Heart J. 2020;61:263–72.
  • 35
    Likhvantsev VV, Landoni G, Grebenchikov OA, et al. Perioperative dexmedetomidine supplement decreases delirium incidence after adult cardiac surgery: a randomized, double-blind, controlled study. J Cardiothorac Vasc Anesth. 2021 ;35:449–57.
  • 36
    Lau G, Wald R, Sladen R, et al. Acute kidney injury in cardiac surgery and cardiac intensive care. Semin Cardiothorac Vasc Anesth. 2015;19:270–87.
  • 37
    Lassnigg A, Schmid ER, Hiesmayr M, et al. Impact of minimal increases in serum creatinine on outcome in patients after cardiothoracic surgery: do we have to revise current definitions of acute renal failure? Crit Care Med. 2008;36:1129–37.
  • 38
    Brown JR, Shore-Lesserson L, Fox AA, 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. J Extra CorporTechnol. 2022;54:267–90.
  • 39
    Yu Y, Li C, Zhu S, et al. Diagnosis, pathophysiology, and preventive strategies for cardiac surgery-associated acute kidney injury: a narrative review. Eur J Med Res. 2023;28:45.
  • 40
    Peng K, Mcllroy DR, Bollen BA, et al. Society of Cardiovascular Anesthesiologists Clinical Practice Update for Management of Acute Kidney Injury Associated with Cardiac Surgery. Anesth Analg. 2022;135:744–56.
  • 41
    Ueki M, Kawasaki T, Habe K, et al. The effects of dexmedetomidine on inflammatory mediators after cardiopulmonary bypass. Anaesthesia. 2014;69:693–700.
  • 42
    Bao N, Tang B. Organ-protective effects and the underlying mechanism of dexmedetomidine. Mediators Inflamm. 2020; 2020:6136105.
  • 43
    Singh D, Jagannath S, Priye S, et al. The comparison of dexmedetomidine, esmolol, and combination of dexmedetomidine with esmolol for attenuation of sympathomimetic response to laryngoscopy and intubation in patients undergoing coronary artery bypass grafting. Ann Card Anaesth. 2019;22:353–7.
  • 44
    Liu X, Rabin PL, Yuan Y, et al. Effects of anesthetic and sedative agents on sympathetic nerve activity. Heart Rhythm. 2019;16:1875–82. 2019/06/30. DOI: 10.1016.
    » https://doi.org/10.1016
  • 45
    Yang SJ, Fan CN, Wang MJ, et al. Effects of dexmedetomidine on renal microcirculation in ischemia/reperfusion-induced acute kidney injury in rats. Sci Rep. 2021;11:2026.
  • 46
    Honore PM, De Bels D, Preseau T, Spapen HD. Dexmedetomidine: the first new kid on the block for preventing cardiac surgery-associated acute kidney injury? Crit Care. 2018;22:151.
  • 47
    Li S, Fu S, Xiao Y, Xu G. Recent Perioperative Pharmacological Prevention of Acute Kidney Injury after Cardiac Surgery: A Narrative Review [published correction appears in Am J Cardiovasc Drugs. 2018 Feb 26] Am J Cardiovasc Drugs. 2017;17:17–25.
  • 48
    Wu S, Yao H, Cheng N, Guo N, Chen J, Ge M, Cai J. Determining whether dexmedetomidine provides a reno-protective effect in patients receiving laparoscopic radical prostatectomy: a pilot study. Int Urol Nephrol. 2019;51:1553–61.
  • 49
    Wu F, Duan H, Xie Y. Preventive effects of dexmedetomidine on renal dysfunction and hemodynamic stability in malignant obstructive jaundice patients during peri-operative period. Med Sci Monit. 2019;25:6782–7.
  • 50
    Soh S, Shim JK, Song JW, Bae JC, Kwak YL. Effect of dexmedetomidine on acute kidney injury after aortic surgery: a single-centre, placebo-controlled, randomised controlled trial. Br J Anaesth. 2020. https://doi.org/10.1016/j.bja.2019.12.036 Online ahead of print.
    » https://doi.org/10.1016/j.bja.2019.12.036
  • 51
    Bayram A, Ulgey A, Baykan A, Narin N, Narin F, Esmaoglu A, Boyaci A. The effects of dexmedetomidine on early stage renal functions in pediatric patients undergoing cardiac angiography using non-ionic contrast media: a double-blind, randomized clinical trial. Paediatr Anaesth. 2014;24:426–32.
  • 52
    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;10:5285.
  • 53
    Massoth C, Zarbock A, Meersch M. Acute Kidney Injury in Cardiac Surgery. Crit Care Clin. 2021 ;37:267–78.
  • 54
    Küllmar M, Zarbock A, Engelman DT, Chatterjee S, Wagner NM. Prevention of acute kidney injury. Crit Care Clin. 2020;36:691–704.
  • 55
    Zeng X, McMahon GM, Brunelli SM, Bates DW, Waikar SS. Incidence, outcomes, and comparisons across definitions of AKI in hospitalized individuals. Clin J Am Soc Nephrol. 2014;9:12–20.
  • 56
    Koeze J, Keus F, Dieperink W, van der Horst IC, Zijlstra JG, van Meurs M. Incidence, timing and outcome of AKI in critically ill patients varies with the definition used and the addition of urine output criteria. BMC Nephrol. 2017;18:70.

Publication Dates

  • Publication in this collection
    28 June 2024
  • Date of issue
    2024

History

  • Received
    12 Jan 2023
  • Accepted
    03 July 2023
  • Published
    14 July 2023
Sociedade Brasileira de Anestesiologia (SBA) Rua Professor Alfredo Gomes, 36, Botafogo , cep: 22251-080 - Rio de Janeiro - RJ / Brasil , tel: +55 (21) 97977-0024 - Rio de Janeiro - RJ - Brazil
E-mail: editor.bjan@sbahq.org