Skip to main content

Impact of the perioperative inotropic support in grown-up congenital heart patients undergoing cardiac surgery: a propensity score adjusted analysis

Abstract

Background

Grown-up congenital heart (GUCH) patients represent a growing population with a high morbidity risk when undergoing reparative surgery. A main preoperative feature is right ventricular failure, which represents a risk factor for postoperative low cardiac output syndrome. Levosimendan has a potentially beneficial effect. This retrospective study included consecutive GUCH patients with surgeries in a tertiary cardiothoracic centre between 01-01-2013 and 01-10-2017, to test the hypothesis that the postoperative use of levosimendan might be associated with shorter time of mechanical ventilation, when compared with the use of milrinone. To adjust for bias related to the probability of treatment assignment, it uses the inverse propensity score weighting methodology.

Results

Overall 363 patients had GUCH surgeries during the study period, their mean age was 31.39 ± 15.31 years, 87 patients were eligible for analysis in the Levosimendan group and 117 in the Milrinone group. The propensity score used pre- and intraoperative variables and resulted in a good balance between covariates. The Levosimendan group included patients with higher preoperative risk scores, a higher prevalence of left and right ventricular failure, who required more often the addition of epinephrine, renal replacement therapy, prolonged mechanical ventilation and intensive care stay. However, after propensity score weighting, patients in the Levosimendan group had shorter durations of mechanical ventilation (average treatment effect − 37.59 h IQR [− 138.85 to − 19.13], p = 0.01) and intensive care stay (average treatment effect − 3.11 days IQR [− 10.03 to − 1.48], p = 0.009). The number of days of additional epinephrine support was shorter and the vasoactive inotropic scores lower.

Conclusion

We report a beneficial effect in terms of duration of mechanical ventilation and intensive care stay, and on inotropic requirements of the use of levosimendan following GUCH surgeries. The use of levosimendan in this setting requires validation at a larger scale.

Introduction

Grown-up congenital heart (GUCH) patients represent a growing population with complex pathophysiology due to decades of living with abnormal cardiac anatomy, which result in comorbidities affecting all organ systems. Therefore, they are at higher risk of postoperative complications when compared with patients with acquired cardiac diseases [1,2,3,4]. When undergoing surgery in early infancy, about 25% of congenital heart patients experience postoperative low cardiac output syndrome (LCOS) [5]. Management of postoperative LCOS includes inotropic-vasoactive support [6], and sometimes mechanical support. Together with beta-agonists and phosphodiesterase-inhibitors, levosimendan has been used in adults and children with LCOS following cardiac surgery. It has been found beneficial in adults with severely reduced left ventricular (LV) systolic function [7], and safe and beneficial in children with congenital cardiac operations [8,9,10]. The pathophysiology of heart failure in GUCH patients is different from that of acquired heart diseases, and they have a high prevalence of right ventricular failure [11]. Therefore, the pulmonary vasodilatatory effect of levosimendan, as well as the positive inotropic and lusitropic effects is particularly appealing in the context of GUCH surgeries. Based on these considerations, we conducted a retrospective study to test the hypothesis that GUCH patients treated by levosimendan for LCOS following cardiopulmonary bypass (CPB), compared to those having received a conventional inotropic-vasoactive support based on milrinone, might be associated with shorter time of mechanical ventilation.

Materials and methods

This retrospective analysis was conducted in a cohort of consecutive GUCH patients older than 15 years, undergoing surgery between January 1st 2013 and October 3rd 2017 at the University Hospital of Bordeaux, France. The Ethical Committee of the French Anaesthetic Society waived the need for written informed consent of the patients to perform this retrospective analysis, after de-identification of all patient data (reference IRB 00010254 - 2019 - 059). Only patients undergoing surgery with CPB and aortic cross-clamping, and with complete postoperative records were analysed.

Because of the absence of a GUCH-specific risk score, the baseline risk category was assessed by the Euroscore II. Its use has, nevertheless, been shown to result in an underestimation of the perioperative risk [12]. Anaesthesia was induced and maintained using a target controlled infusion of propofol and remifentanil, and with cisatracurium. The arterial and central venous pressure, as well as the venous oxygen saturation and cerebral oxygen saturation were monitored as part of the institutional protocol. Transesophageal ultrasonographic assessment was used in all patients. Normothermic non-pulsatile cardiopulmonary bypass (CPB) was performed using a roller pump and full heparinization (300 U kg−1 heparin and an ACT requirement > 400), the pump flow was set at 3.0–3.5 L min−1 m−2. Warm blood cardioplegia every 20 min and monitoring of the cardioplegic perfusion was performed in all cases, the reinfusion interval was shortened to 12 min in case of ventricular hypertrophy. Monitoring of the pulmonary arterial pressure and of the left atrial pressure was decided at the end of CPB by the attending anaesthesiologist and surgeon according to the underlying pathology and to the preoperative myocardial function.

At the end of CPB, the inotropic-vasoactive strategy was decided by the attending anaesthesiologist, according to the underlying pathology, the preoperative myocardial function, the duration of CPB and cross-clamping and the hemodynamic and ultrasonographic assessment. Patients received either no inotrope, milrinone 0.5 to 1 mcg kg−1 min−1 or levosimendan 0.2 mcg kg−1 min−1 during 24 h, and additional low dose (0.02 to 0.05 mcg kg−1 min−1) epinephrine or norepinephrine, if required. The patients who have not received any inotropic-vasoactive support were not analysed here. Accordingly, the patients were analysed in the Milrinone or the Levosimendan group. Postoperatively, the inotropic-vasoactive support was discontinued according to the daily hemodynamic, ultrasonographic and biological assessment. The vasoactive and inotropic score (VIS) was calculated for the first 4 postoperative days. The Clinical Pulmonary Infection Score (CPIS) [13] was used to diagnose ventilator-associated pneumonia. Renal replacement therapy was available for patients with severe postoperative kidney injury. When required, mechanical ventricular assistance was provided using intra-aortic balloon pump therapy (IABP) or extracorporeal membrane oxygenation (ECMO).

Outcomes

The primary outcome was the duration of mechanical ventilation. This endpoint was chosen because only patients haemodynamically stable without significant organ dysfunction are extubated. Secondary outcomes included epinephrine requirement, VIS, CPIS within 48 h, requirement for renal replacement therapy, duration of ICU and hospital stay.

Statistical analysis

All baseline, intra- and postoperative data were available in the institutional database, in which was stored daily information provided by the attending physicians. Continuous normally distributed variables were expressed as means and standard deviations (SD), otherwise as medians and inter-quartile ranges (IQR) and compared with the Student’s t or Mann–Whitney U tests, as appropriate. Categorical variables were expressed as numbers and percentages and compared with the Chi-square test or Fisher’s exact test, as appropriate. Analysis was conducted in intention to treat.

To avoid bias due to the huge differences observed between characteristics of the control patients and the others, the analysis was restricted to the comparison of the Levosimendan and Milrinone groups. The inverse probability of treatment weighting (IPTW) method was used to control for bias due to selection of patients placed on levosimendan or on milrinone. First, a propensity score model included pre- and intraoperative characteristics which could have influenced the decision of starting levosimendan rather than milrinone at the end of CPB. Then, the contribution of each subject was weighted by 1/propensity score in the Milrinone group, and by 1/(1-propensity score) in the Levosimendan group. These weights assured that, for each combination of the covariates used in the propensity score model, the sum of the contributions of all subjects is equal. Balance on covariates between groups after the IPTW weighting was assessed by computing their standardized differences, and groups were considered balanced if the standardized differences were < 0.25. Short-term outcome variables were compared using IPTW-weighted regression models. Results were expressed as absolute average treatment effect, and 95% confidence intervals were estimated by bootstrapping with 500 re-samples. Statistical significance was set at p < 0.05. Analyses were performed using the basic R software package and the “survey” package (https://www.r-project.org).

Results

Overall, 363 GUCH surgeries were performed during the study period. The patients were on average 31.31 ± 15.63 years old. No patient died within 30 days of admission. As shown in Fig. 1, only 204 patients were analysed here: 117 in the Milrinone group and 87 in the Levosimendan group. As shown in Fig. 2, 49.3% of all patients received one inotropic-vasoactive agent, and 23.8% received two agents. In one patient started on milrinone and epinephrine, levosimendan was introduced on day 2. Overall, 7 patients were placed on IABP (6) or ECMO (1) at the end of CPB, concomitant with starting the inotropic-vasoactive support: 6 in the Levosimendan group and 1 on the Milrinone group. To avoid bias related to the association between this outcome and the postoperative use of inotropes, these patients were not further analysed. Another 2 patients in the Levosimendan group required a second surgery at postoperative day 2 and were placed on ECMO at the end of their second CPB, they were not analysed either, leaving under analysis 87 patients in the Levosimendan group and 117 patients in the Milrinone group. No other patient required mechanical ventricular assistance.

Fig. 1
figure 1

Flowchart of the study cohort

Fig. 2
figure 2

Distribution of the inotropic-vasoactive support among the Milrinone and the Levosimendan groups. Overall, 49.3% of the patients received one inotropic-vasoactive agent, and 23.8% received two agents

The baseline, intra- and post-operative characteristics of the study population are shown in Table 1. The underlying pathologies and surgical interventions are shown in Fig. 3, together with the distribution of the inotropic-vasoactive support. The variables used in the propensity score model are shown in Table 2, together with their standardized differences before and after IPTW. The propensity score model was well calibrated (Hosmer–Lemeshow test P value = 0.59) and had good discrimination (C-index = 0.809). As shown in Table 1, patients in both groups had a high incidence of RV anomaly (failing, hypoplastic, hypertrophic or dilated): 89% in the Milrinone group and 74% in the Levosimendan group. Patients in the Levosimendan group had the highest preoperative risk scores, the highest rates of left and/or right ventricular dysfunction, and underwent redo-surgery more often. As shown in Table 1, they had the longest CPB and cross-clamping durations too. After IPTW weighting, there was a good balance between the pre- and intraoperative variables (Table 2).

Table 1 Baseline, intraoperative and postoperative variables among the treatment groups
Fig. 3
figure 3

Distribution of the inotropic-vasoactive support according to the underlying pathology and procedure. ASD atrial septal defect, VSD: ventricular septal defect

Table 2 Variables used in the propensity score model: balance before and after inverse probability of treatment weighting

The analysis of the outcome variables is shown in Table 3. Before weighting (Table 1), patients in the Levosimendan group required more often additional epinephrine and had longer durations of epinephrine infusion, they required more often renal replacement therapy and mechanical ventricular support, and they had longer durations of mechanical ventilation and longer durations of ICU and hospital stay. However, after IPTW weighting (Table 3), there was a significantly shorter duration of mechanical ventilation and ICU stay in the Levosimendan group. After IPTW weighting, patients in the Milrinone group had significantly higher VIS within the first 4 postoperative days. Although not statistically significant, there was a shorter duration of additional epinephrine support and of hospital stay in patients who received levosimendan.

Table 3 Outcome variables after inverse probability of treatment weighting

Discussion

We report the results of a retrospective study of outcomes following GUCH surgeries, where the patients received either levosimendan or milrinone postoperatively. Although the patients in the Levosimendan group had the highest risk scores, had more often preoperative ventricular dysfunction, and had longer surgeries, after adjustment by a propensity score including baseline and intraoperative variables, they had shorter durations of mechanical ventilation and ICU stay. This result should prompt prospective research to validate the use of levosimendan in the setting of GUCH surgery.

To date, this is the first study which compares levosimendan and milrinone during the perioperative care of GUCH surgeries. A randomized clinical trial of levosimendan vs milrinone in children less than 5 years of age undergoing congenital cardiac surgery has shown equivalence in terms of hemodynamic and biochemical parameters [9]. In neonates undergoing complex cardiac surgeries, a trial of levosimendan vs a more conventional inotropic strategy resulted in lower lactate levels [10]. A meta-analysis of levosimendan trials in paediatric cardiac surgery suggested that the drug was safe and provided a potential clinical benefit when applied to postoperative LCOS [8].

Data published in patients with acquired cardiac disease surgeries are conflicting. Both the LICORN and LEVO-CTS trials [14, 15] showed that a prophylactic 0.1 mcg kg−1 min−1 levosimendan infusion in patients with reduced LV ejection fraction did not result in a significantly lower rate of the short-term composite outcome (defined by a combination of LCOS criteria, requirement for mechanical ventricular assist device, renal replacement therapy, or death) when compared with placebo. The CHEETAH trial [16] showed no reduction in 30-day mortality when a very low 0.07 mcg kg−1 min−1 levosimendan dose was administered to patients with postoperative LCOS. These results are, however, subject of controversy [17]. The low levosimendan regimen in both the LICORN and the CHEETAH trials [14, 16] has been questioned, and details about hemodynamic monitoring are missing. In the CHEETAH trial [16], the patients started on levosimendan were already receiving high doses of epinephrine or dobutamine, which could have reduced the inotropic effect of levosimendan. The medical community agrees that some encouraging evidence of efficacy emerged from LEVO-CTS nevertheless [17]: the lower incidence of LCOS, lesser need for inotropic support by catecholamines, and improvement in CI indicate that levosimendan exhibited efficacy. Moreover, a subgroup analysis of the LEVO-CTS trial demonstrated that levosimendan was associated with lower 90-day mortality and LCOS in patients undergoing isolated coronary artery bypass grafting [18]. A systematic review pooling data from 28 280 patients included in 177 trials of inotropic-vasoactive support used to treat LCOS in vasoplegic syndromes, sepsis and cardiac surgery suggested that levosimendan was the only drug associated with improvement in survival [19]. As a result, a panel of 27 European experts reached a consensus on the recommendations proposed for the prophylactic use of levosimendan in cardiac surgery [20].

GUCH patients have higher risk of postoperative mortality and morbidity when compared with acquired cardiac disease patients. According to the Nationwide Inpatient Sample database, including data from over 2 million patients, the postoperative mortality following GUCH surgeries is 2.6% versus 1.8% following coronary artery bypass grafting (4). When compared with acquired cardiac disease patients, GUCH patients are at higher risk of arrhythmia (51.6 vs 29.8%), sepsis (7.24 vs 4.61%), thromboembolic complications (3.9 vs 1.4%) and neurologic complications (2.6 vs 0.9%)4. Postoperative mortality rates reported in smaller studies vary between 0.7% and 2% [12, 21,22,23], according to whether univentricular heart patients were included or not. The main risk factors of death include a NYHA category ≥ III, an altered RV function, emergency surgery [21], as well as pulmonary hypertension [23]. No patient in the present cohort died within 30 days of surgery, however long-term outcomes were not analysed. Two major risk factors of death, the NYHA category and an altered RV function, were found different between groups and were used to model the propensity score and to adjust for the probability of treatment assignment. Importantly, here we studied a very high risk GUCH population: in the 830 GUCH patients studied by Putman et al. over 17 years (5), less than 40% of the patients had NYHA III or IV category, whereas 60% of the patients in the present cohort were allocated to NYHA III or IV category.

The pathophysiological mechanism of cardiac failure in GUCH patients is complex11, associating: (i) volume overload through residual shunts and valvular regurgitations; (ii) pressure overload through intraventricular or valvular outflow tract obstructions; (iii) pulmonary hypertension; (iv) chronic cyanosis; (v) myocardial injury; (vi) arrhythmia; (vii) and inability of univentricular hearts (especially when following palliation of the hypoplastic left heart syndrome) to cope with the metabolic demand over the long term [24]. RV failure is very common in tetralogy of Fallot patients having undergone trans-annular patch repair, and who suffer of chronic pulmonary regurgitation [25], as well as in patients with chronic pulmonary hypertension subsequent to long-term left-to-right shunting [26, 27]. Additionally, pulmonary vascular resistances may increase postoperatively due to hypoxia, hypercapnia, acidosis, hypothermia, positive pressure ventilation and alpha-receptor stimulation. All together, these may explain the differences reported in the postoperative course of GUCH surgeries, when compared with acquired cardiac diseases. Here, 73.1% of all patients required inotropic-vasoactive support (49.3% of the patients received one inotropic-vasoactive agent, and 23.8% received two), which suggests that there was a high risk of LCOS in the present population (Fig. 2).

In 2016, the Working Group of Grown-Up Congenital Heart Disease and the Heart Failure Association of the European Society of Cardiology published the guidelines for the treatment of heart failure in GUCH patients11. Unfortunately, it did not provide guidelines for the management of acute heart failure, such as seen postoperatively. Since the use of catecholamines or phosphodiesterase-inhibitors in adults with cardiogenic shock is associated with increased mortality [28, 29], the use of newer drugs such as levosimendan to improve systolic function putatively without elevating intracellular calcium and without increasing the myocardial oxygen consumption [30] is appealing.

Milrinone, a conventional heart failure treatment in children and adults, selectively inhibits intracellular cardiac phosphodiesterase type 3, and the positive inotropic effect results through increased intracellular calcium levels. Its positive lusitropic properties, as well as systemic and pulmonary vasodilatation, make milrinone particularly useful when LCOS results from diastolic ventricular dysfunction and RV failure, or in case of pulmonary hypertension, such as often seen in GUCH patients. However, a significant increase in postoperative tachyarrhythmias [31] and mortality risk [32] has been reported.

Levosimendan binds cardiac troponin C and stabilizes calcium-induced conformational changes, which, in turn, promotes the prolonged interaction between actin and myosin filaments during systole. The resulting increase in contractility is unmatched by either milrinone or dobutamine [33, 34]. Levosimendan improves myocardial efficiency without an increase in the myocardial oxygen consumption [34], which reduces the risk of arrhythmia. Its vasodilatory effect is mediated by opening ATP-sensitive potassium channels in systemic, pulmonary, and coronary vascular smooth muscle cells. It has also been suggested that there was a potential for improvement of diastolic function [34].

Levosimendan has been shown to provide hemodynamic support in a wide range of critical illness situations, including cardiogenic or septic shock, weaning from mechanical ventilation, weaning from extracorporeal membrane oxygenation and cardiorenal syndrome [35]. Levosimendan has a positive impact on several specific conditions related to GUCH surgeries. As aforementioned, GUCH patients often present with a failing RV, and a recent meta-analysis reported improved RV function when levosimendan was used to treat RV failure in a variety of heart and lung diseases [36]. Levosimendan induces vasodilatation in the pre-constricted pulmonary circulation [37], which is of particular relevance in the post-CPB setting. By improving the diastolic function, it is of particular interest when LCOS is due to LV or RV diastolic dysfunction, a common finding in patients with residual obstruction of the left or right outflow tract. Experimental work demonstrated an increase in the RV myocardial efficiency when levosimendan was used to treat RV hypertrophy and failure [38], and it has been proposed for the treatment of RV failure in patients with pulmonary hypertension [39].

Vasopressors are unsuitable for GUCH patients, they have a negative impact on the myocardium [40] and on the microcirculation [41], and their use has been linked with poor outcomes after GUCH surgeries [29]. Therefore, the choice made by the authors was to privilege levosimendan, milrinone and volume load, and to associate low dosages of epinephrine and/or norepinephrine in case of systemic hypotension and evidence of inappropriate organ perfusion. Both vasopressors were weaned as soon as possible. Importantly, patients in the Levosimendan group were weaned from epinephrine earlier than patients in the Milrinone group (Table 3), and had lower VIS during the early postoperative days.

The main finding here was a shorter duration of mechanical ventilation and ICU stay in the Levosimendan group. Importantly, the CPIS score was similar in the Levosimendan and in the Milrinone groups, showing that the difference in ventilation durations was not due to the occurrence of postoperative pneumopathy. We could not infer any hypothesis about the pathophysiological pathways leading to this beneficial effect based on this retrospective data analysis. Literature contains several reports linking the use of levosimendan to successful weaning from mechanical ventilation, either due to recovery of myocardial dysfunction [42, 43] or to recovery of diaphragmatic dysfunction [44,45,46]. We hypothesize that the beneficial effect is partly due to its pharmacological properties, since the concentration of the long-lasting active metabolite OR 1896 remains stable up to 8 days after a 24 h infusion, and enables to overcome the increase in the metabolic demand during ventilation weaning through a residual positive inotropic effect.

Limitations

The results of this single-centre and small-sampled retrospective study need to be interpreted with caution, and require validation in prospective trials. Because of the huge initial unbalance between groups, the patients in the Control group were not included in the outcome analysis. Due to missing data inherent to the retrospective design of the study, several hemodynamic and echocardiographic parameters could not be analysed, therefore it was not possible to infer hypothesis about the pathophysiological pathways leading to the beneficial effect of levosimendan. The use of the VIS might have favoured the Levosimendan group here, since the VIS does not account for the vasoactive and inotropic effect of levosimendan. Due to missing data, long-term outcomes were not analysed either.

Conclusion

Critical care management requires an in-depth understanding of underlying pathophysiology of GUCH patients in order to apply contemporary concepts of adult intensive care to this specific population. Our study suggests that when used following surgery in high-risk GUCH patients, levosimendan has a beneficial impact on the duration of mechanical ventilation, of ICU stay, and on inotropic requirements. Further research is needed to validate the use of levosimendan in this setting.

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

References

  1. Giamberti A, Chessa M, Abella R, et al. Morbidity and mortality risk factors in adults with congenital heart disease undergoing cardiac reoperations. Ann Thorac Surg. 2009;88:1284–9.

    Article  PubMed  Google Scholar 

  2. Holst KA, Dearani JA, Burkhart HM, et al. Risk factors and early outcomes of multiple reoperations in adults with congenital heart disease. Ann Thorac Surg. 2011;92:122–8 (discussion 129–130).

    Article  PubMed  Google Scholar 

  3. Kirshbom PM, Myung RJ, Simsic JM, et al. One thousand repeat sternotomies for congenital cardiac surgery: risk factors for reentry injury. Ann Thorac Surg. 2009;88:158–61.

    Article  PubMed  Google Scholar 

  4. Nasr VG, Faraoni D, Valente AM, DiNardo JA. Outcomes and costs of cardiac surgery in adults with congenital heart disease. Pediatr Cardiol. 2017;38:1359–64.

    Article  PubMed  Google Scholar 

  5. Hoffman TM, Wernovsky G, Atz AM, et al. Efficacy and safety of milrinone in preventing low cardiac output syndrome in infants and children after corrective surgery for congenital heart disease. Circulation. 2003;107:996–1002.

    Article  CAS  PubMed  Google Scholar 

  6. Lomivorotov VV, Efremov SM, Kirov MY, Fominskiy EV, Karaskov AM. Low-cardiac-output syndrome after cardiac surgery. J Cardiothorac Vasc Anesth. 2017;31:291–308.

    Article  PubMed  Google Scholar 

  7. Sanfilippo F, Knight JB, Scolletta S, et al. Levosimendan for patients with severely reduced left ventricular systolic function and/or low cardiac output syndrome undergoing cardiac surgery: a systematic review and meta-analysis. Crit Care. 2017;21:252.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Angadi U, Westrope C, Chowdhry MF. Is levosimendan effective in paediatric heart failure and post-cardiac surgeries? Interact Cardiovasc Thorac Surg. 2013;17:710–4.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Momeni M, Rubay J, Matta A, et al. Levosimendan in congenital cardiac surgery: a randomized, double-blind clinical trial. J Cardiothorac Vasc Anesth. 2011;25:419–24.

    Article  CAS  PubMed  Google Scholar 

  10. Ricci Z, Garisto C, Favia I, Vitale V, Di Chiara L, Cogo PE. Levosimendan infusion in newborns after corrective surgery for congenital heart disease: randomized controlled trial. Intensive Care Med. 2012;38:1198–204.

    Article  CAS  PubMed  Google Scholar 

  11. Budts W, Roos-Hesselink J, Radle-Hurst T, et al. Treatment of heart failure in adult congenital heart disease: a position paper of the Working Group of Grown-Up Congenital Heart Disease and the Heart Failure Association of the European Society of Cardiology. Eur Heart J. 2016;37:1419–27.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Putman LM, van Gameren M, Meijboom FJ, et al. Seventeen years of adult congenital heart surgery: a single centre experience. Eur J Cardiothorac Surg. 2009;36:96–104 (Discussion 104).

    Article  PubMed  Google Scholar 

  13. Schurink CAM, Nieuwenhoven CAV, Jacobs JA, et al. Clinical pulmonary infection score for ventilator-associated pneumonia: accuracy and inter-observer variability. Intensive Care Med. 2004;30:217–24.

    Article  PubMed  Google Scholar 

  14. Cholley B, Caruba T, Grosjean S, et al. Effect of levosimendan on low cardiac output syndrome in patients with low ejection fraction undergoing coronary artery bypass grafting with cardiopulmonary bypass: The LICORN Randomized Clinical Trial. JAMA. 2017;318:548–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mehta RH, Leimberger JD, van Diepen S, et al. Levosimendan in patients with left ventricular dysfunction undergoing cardiac surgery. N Engl J Med. 2017;376:2032–42.

    Article  CAS  PubMed  Google Scholar 

  16. Landoni G, Lomivorotov VV, Alvaro G, et al. Levosimendan for hemodynamic support after cardiac surgery. N Engl J Med. 2017;376:2021–31.

    Article  CAS  PubMed  Google Scholar 

  17. Guarracino F, Heringlake M, Cholley B, et al. Use of Levosimendan in cardiac surgery: an update after the LEVO-CTS, CHEETAH, and LICORN trials in the light of clinical practice. J Cardiovasc Pharmacol. 2018;71:1–9.

    Article  CAS  PubMed  Google Scholar 

  18. van Diepen S, Mehta RH, Leimberger JD, et al. Levosimendan in patients with reduced left ventricular function undergoing isolated coronary or valve surgery. J Thorac Cardiovasc Surg. 2019;14(7):1286–95.

    Google Scholar 

  19. Belletti A, Castro ML, Silvetti S, et al. The Effect of inotropes and vasopressors on mortality: a meta-analysis of randomized clinical trials. Br J Anaesth. 2015;115:656–75.

    Article  CAS  PubMed  Google Scholar 

  20. Toller W, Heringlake M, Guarracino F, et al. Preoperative and perioperative use of levosimendan in cardiac surgery: European expert opinion. Int J Cardiol. 2015;184:323–36.

    Article  CAS  PubMed  Google Scholar 

  21. Beurtheret S, Tutarel O, Diller GP, et al. Contemporary cardiac surgery for adults with congenital heart disease. Heart. 2017;103:1194–202.

    Article  PubMed  Google Scholar 

  22. Karangelis D, Mazine A, Narsupalli S, Mendis S, Veldtman G, Nikolaidis N. Morbidity after cardiac surgery in patients with adult congenital heart disease in comparison with acquired disease. Heart Lung Circ. 2018;27:739–44.

    Article  PubMed  Google Scholar 

  23. Vida VL, Berggren H, Brawn WJ, et al. Risk of surgery for congenital heart disease in the adult: a multicentered European study. Ann Thorac Surg. 2007;83:161–8.

    Article  PubMed  Google Scholar 

  24. Roche SL, Redington AN. The failing right ventricle in congenital heart disease. The Canadian journal of cardiology. 2013;29:768–78.

    Article  PubMed  Google Scholar 

  25. Wald RM, Valente AM, Marelli A. Heart failure in adult congenital heart disease: Emerging concepts with a focus on tetralogy of Fallot. Trends Cardiovasc Med. 2015;25:422–32.

    Article  PubMed  Google Scholar 

  26. Kohler D, Arnold R, Loukanov T, Gorenflo M. Right ventricular failure and pathobiology in patients with congenital heart disease—implications for long-term follow-up. Front Pediatr. 2013;1:37.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Pinsky MR. The right ventricle: interaction with the pulmonary circulation. Crit Care. 2016;20:266.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Koponen T, Karttunen J, Musialowicz T, Pietilainen L, Uusaro A, Lahtinen P. Vasoactive-inotropic score and the prediction of morbidity and mortality after cardiac surgery. Br J Anaesth. 2019;122:428–36.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Poterucha JT, Vallabhajosyula S, Egbe AC, et al. Vasopressor magnitude predicts poor outcome in adults with congenital heart disease after cardiac surgery. Congenital Heart Dis. 2019;14:193–200.

    Article  Google Scholar 

  30. Maack C, Eschenhagen T, Hamdani N, et al. Treatments targeting inotropy. Eur Heart J. 2018;40:3626–44.

    Article  CAS  PubMed Central  Google Scholar 

  31. Smith AH, Owen J, Borgman KY, Fish FA, Kannankeril PJ. Relation of milrinone after surgery for congenital heart disease to significant postoperative tachyarrhythmias. Am J Cardiol. 2011;108:1620–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Nielsen DV, Torp-Pedersen C, Skals RK, Gerds TA, Karaliunaite Z, Jakobsen CJ. Intraoperative milrinone versus dobutamine in cardiac surgery patients: a retrospective cohort study on mortality. Crit Care. 2018;22:51.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Banfor PN, Preusser LC, Campbell TJ, et al. Comparative effects of levosimendan, OR-1896, OR-1855, dobutamine, and milrinone on vascular resistance, indexes of cardiac function, and O2 consumption in dogs. Am J Physiol Heart Circ Physiol. 2008;294:H238–48.

    Article  CAS  PubMed  Google Scholar 

  34. Stocker CF, Shekerdemian LS, Norgaard MA, et al. Mechanisms of a reduced cardiac output and the effects of milrinone and levosimendan in a model of infant cardiopulmonary bypass. Crit Care Med. 2007;35:252–9.

    Article  CAS  PubMed  Google Scholar 

  35. Herpain A, Bouchez S, Girardis M, et al. Use of levosimendan in intensive care unit settings: an opinion paper. J Cardiovasc Pharmacol. 2019;73:3–14.

    Article  CAS  PubMed  Google Scholar 

  36. Qiu J, Jia L, Hao Y, et al. Efficacy and safety of levosimendan in patients with acute right heart failure: a meta-analysis. Life Sci. 2017;184:30–6.

    Article  CAS  PubMed  Google Scholar 

  37. De Witt BJ, Ibrahim IN, Bayer E, et al. An analysis of responses to levosimendan in the pulmonary vascular bed of the cat. Anesth Analges. 2002;94:1427–33.

    Article  Google Scholar 

  38. Vildbrad MD, Andersen A, Holmboe S, Ringgaard S, Nielsen JM, Nielsen-Kudsk JE. Acute effects of levosimendan in experimental models of right ventricular hypertrophy and failure. Pulmonary circulation. 2014;4:511–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Hansen MS, Andersen A, Nielsen-Kudsk JE. Levosimendan in pulmonary hypertension and right heart failure. Pulmon Circ. 2018;8:2045894018790905.

    Google Scholar 

  40. Schmittinger CA, Torgersen C, Luckner G, Schroder DC, Lorenz I, Dunser MW. Adverse cardiac events during catecholamine vasopressor therapy: a prospective observational study. Intensive Care Med. 2012;38:950–8.

    Article  CAS  PubMed  Google Scholar 

  41. De Backer D, Foulon P. Minimizing catecholamines and optimizing perfusion. Crit Care. 2019;23:149.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Routsi C, Stanopoulos I, Kokkoris S, Sideris A, Zakynthinos S. Weaning failure of cardiovascular origin: how to suspect, detect and treat-a review of the literature. Ann Intensive Care. 2019;9:6.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Sterba M, Banerjee A, Mudaliar Y. Prospective observational study of levosimendan and weaning of difficult-to-wean ventilator dependent intensive care patients. Crit Care Resuscit J Australas Acad Crit Care Med. 2008;10:182–6.

    Google Scholar 

  44. Doorduin J, Sinderby CA, Beck J, et al. The calcium sensitizer levosimendan improves human diaphragm function. Am J Respir Crit Care Med. 2012;185:90–5.

    Article  CAS  PubMed  Google Scholar 

  45. van Hees HW, Andrade Acuna G, Linkels M, Dekhuijzen PN, Heunks LM. Levosimendan improves calcium sensitivity of diaphragm muscle fibres from a rat model of heart failure. Br J Pharmacol. 2011;162:566–73.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. van Hees HW, Dekhuijzen PN, Heunks LM. Levosimendan enhances force generation of diaphragm muscle from patients with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2009;179:41–7.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

All authors are members of the medical staff in the department and played a key role in the study.

Funding

Support was provided solely by institutional, departmental sources, or both. No external funds were obtained.

Author information

Authors and Affiliations

Authors

Contributions

Study concept and design: PM. Data collection: SS. Data analysis and cleaning: SS, MB, PM. Interpretation of data: PM, SS, MB. Statistical analysis: MB. Drafting the manuscript: PM, SS. Revising the manuscript: PM, MB, AO. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Philippe Mauriat.

Ethics declarations

Ethics approval and consent to participate

The Ethical Committee of the French Anaesthetic Society waived the need for written informed consent of the patients to perform this retrospective analysis, after de-identification of all patient data (reference IRB 00010254 - 2019 - 059). All patients gave written consent to perform the surgery and the perioperative care.

Consent for publication

Not applicable.

Competing interests

Dr. Mauriat, reports receiving honoraria for presentations and consultancy together with financial support from Orion and Medtronic. Drs. Bojan, Soulié, Foulgoc and Tafer declare no conflicts of interest. Professor Ouattara reports receiving honoraria for presentations and consultancy together with financial support from LFB, The Abbvie Pharmaceutical Company, Nordic Pharma BV, Abiomed and Orion.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mauriat, P., Bojan, M., Soulie, S. et al. Impact of the perioperative inotropic support in grown-up congenital heart patients undergoing cardiac surgery: a propensity score adjusted analysis. Ann. Intensive Care 10, 91 (2020). https://doi.org/10.1186/s13613-020-00709-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13613-020-00709-0

Keywords