Skip to main content

The clinical effectiveness of sivelestat in treating sepsis patients with both acute respiratory distress syndrome and septic cardiomyopathy

Abstract

Background

We aimed to assess the efficacy of the neutrophil elastase inhibitor, sivelestat, in the treatment of sepsis-induced acute respiratory distress syndrome (ARDS) and septic cardiomyopathy (SCM).

Methods

Between January 2019 and December 2021, we conducted a randomized trial on patients who had been diagnosed with sepsis-induced acute respiratory distress syndrome (ARDS) and septic cardiomyopathy (SCM) at Wuhan Union Hospital. The patients were divided into two groups by random envelop method, the Sivelestat group and the Control group. We measured the serum concentrations of Interleukin (IL)-6, IL-8, Tumor necrosis factor-α (TNF-α), and High-mobility group box 1 (HMGB1) at five time points, which were the baseline, 12 h, 24 h, 48 h, and 72 h after admission to the ICU. We evaluated the cardiac function by sonography and the heart rate variability (HRV) with 24-hour Holter recording between the time of admission to the intensive care unit (ICU) and 72 h after Sivelestat treatment.

Results

From January 2019 to December 2021, a total of 70 patients were included in this study. The levels of IL-6, IL-8, and TNF-α were significantly lower in the Sivelestat group at different time points (12 h, 24 h, 48 h, and 72 h). HMGB1 levels were significantly lower at 72 h after Sivelestat treatment (19.46 ± 2.63pg/mL vs. 21.20 ± 2.03pg/mL, P = 0.003). The stroke volume (SV), tricuspid annular plane systolic excursion (TAPSE), early to late diastolic transmitral flow velocity (E/A), early (e’) and late (a’) diastoles were significantly low in the Control group compared with the Sivelestat group. Tei index was high in the Control group compared with the Sivelestat group (0.60 ± 0.08 vs. 0.56 ± 0.07, P = 0.029). The result of HRV showed significant differences in standard deviation of normal-to-normal intervals (SDNN), low frequency (LF), and LF/HF (high frequency) between the two groups.

Conclusions

Sivelestat can significantly reduce the levels of serum inflammatory factors, improve cardiac function, and reduce heart rate variability in patients with Sepsis-induced ARDS and SCM.

Peer Review reports

Introduction

Sepsis is a severe medical condition that can lead to high rates of morbidity and mortality [1]. As sepsis becomes more severe, the likelihood of complications such as acute respiratory distress syndrome (ARDS)/ acute lung injury (ALI) and septic cardiomyopathy (SCM) increases [2, 3]. ARDS/ALI has been linked to sepsis for almost a century [4], with more than half of Intensive Care Unit (ICU) admissions for sepsis resulting in ARDS development [5]. SCM is a life-threatening condition that results in myocardial dysfunction and multiple organ failure, occurring in 40–60% of septic shock patients [6,7,8]. Despite significant progress in ARDS/ALI treatment [9], treatment options for SCM remain limited, resulting in a high mortality rate [10].

Sivelestat, a selective neutrophil elastase inhibitor, has shown promise in treating various conditions, including ALI, refractory Kawasaki disease, and acute pancreatitis [11]. Its protease-inhibiting and anti-inflammatory effects have been demonstrated as effective in shortening ICU duration and reducing ventilator use in ARDS/ALI patients [12]. Additionally, studies suggest that Sivelestat can reduce and prevent tissue ischemia and reperfusion injury (IRI) in multiple organs [13]. Earlier studies in Japan have confirmed that the effects of sivelestat in reducing the permeability of pulmonary blood vessels [14], inhibiting mucus secretion in the epithelial layer [15], and decreasing the production of inflammatory cytokines such as IL-1β, IL-6, and TNF-α at clinically available concentrations [16] and protecting against postperfusion-induced lung injury. Although the specific mechanisms of Sivelestat’s role in sepsis are not yet fully understood, previous studies have pointed to its anti-inflammatory, anti-oxidative stress, and anti-apoptotic properties [17]. The present study aims to determine the therapeutic effect of Sivelestat in treating sepsis-induced ALI and SCM, with the goal of improving patient outcomes and reducing mortality rates.

Materials and methods

Design, patients, and grouping

A prospective randomized controlled study was conducted on patients with Sepsis-induced ARDS/ALI and SCM, who were admitted to the ICU from January 2019 to December 2021. Patients under the age of 18, those with severe liver cirrhosis, previous liver transplantation, severe central nervous system disease, uncontrolled malignancy, or clotting disorders were excluded from the study [18]. The Sivelestat group and the Control group were classified based on whether patients received the Sivelestat administration or not and it was determined by the random envelop method. This study was approved by the ethics committee of Wuhan Union Hospital (No. 2018-0598-08) and was registered on the China Clinical Trial Registry (ChiCTR-OPC-17,013,126). It was performed in accordance with the 1964 declaration of Helsinki and later amendments. All methods were carried out in accordance with relevant guidelines and regulations. Informed consent was obtained from all subjects and/or their legal guardians.

Diagnosis of sepsis-induced ARDS/ALI and SCM

According to the criteria established by the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis3), we defined sepsis as a condition where an infection triggers a life-threatening host response leading to organ dysfunction [19, 20]. We measured organ dysfunction by monitoring changes in Sequential Organ Failure Assessment (SOFA) scores that an increase in score of 2 points or more was considered indicative of dysfunction [21]. To diagnose ARDS, we followed specific criteria: 1) severe hypoxemia (PaO2/FiO2 ≤ 300 mmHg), 2) acute symptoms that developed within a week, 3) bilateral radiographic abnormalities (not attributable to atelectasis), and 4) The disorder was not caused by heart failure [22]. Meanwhile, we diagnosed SCM based on three criteria as follows: First, the patient had been diagnosed with sepsis; Second, the patient had one of the following three ultrasound abnormalities: A) left ventricular systolic mitral annulus velocity (LV-Sm) < 8 cm/s or Left Ventricular Ejection Fractions (LVEF) < 50%; B) Right ventricular systolic mitral annulus velocity (RV-Sm) < 12 cm/s; or C) peak early diastolic transmitral flow velocity/peak early diastolic mitral annular velocity (E/e’) > 15 or peak early diastolic mitral annular velocity (e’) < 8 cm/s; Third, the patient had no history of chronic heart disease, including coronary heart disease, chronic heart failure, regional ventricular wall motion abnormality, dilated cardiomyopathy, hypertrophic obstructive cardiomyopathy, congenital heart disease, or heart valve disease [23].

Interventions and treatment

Each ICU physician made antibiotic selections that were appropriate for the patients and a low dose of steroids was given to manage septic shock. Patients diagnosed with ARDS and SCM received intravenous injections of Sivelestat (Shanghai Huilun Pharmaceutical Co., LTD, China) at a rate of 0.2 mg/kg/hr on the day of diagnosis [24] and the treatment was continued until either the patient was discharged from the ICU or a period of 14 days. Accordingly, patients in the Control group were under the same treatment regiments such as cardiac function and respiratory function maintenance, which were consistent with those in the Sivelestat group, except that patients did not receive the Sivelestat.

Outcomes

The Clinical Effectiveness of Sivelestat was evaluated based on plasma inflammatory factor levels, cardiac function, and heart rate variability (HRV), among which the cardiac function evaluated by sonography was the primary outcome. The patients’ test results indicating abnormal liver and kidney function were considered as adverse events.

At baseline and 4 additional time points (12 h, 24 h, 48 h, and 72 h after Sivelestat administration), clinical data were collected. The data included infection sites, assessment system values (such as the SOFA score, Acute Physiology and Chronic Health Evaluation II [APACHE II] score, and Systemic Inflammatory Response Syndrome [SIRS] score), and previous medical history. Blood samples were drawn, centrifuged, and stored at -80 °C [25]. Lab data, including levels of Serum Tumor Necrosis Factor-alpha (TNF-α), Interleukin-8 (IL-8), Interleukin-6 (IL-6), and High Mobility Group Box 1 (HMGB1), were measured at different time points via enzyme-linked immunosorbent assay (ELISA) by Wuhan ADANTI Biotechnology Co., Ltd.

Cardiac function evaluated by sonography

At the time of ICU admission and 72 h after the Sivelestat treatment, a GE system equipped with a multiplane 5-MHz transesophageal echocardiographic transducer was used for Cardiac Function Evaluated. LV systolic function was evaluated using visual gestalt method, and Simpson’s method, fractional shortening. The visual gestalt method qualitatively assessed LV function based on LV size, contractility, and thickening of myocardial segments, and was classified into hyperdynamic, normal, mild, moderate, and severe LV dysfunction. Using Simpson’s method, readings were taken in apical 4-chamber view to calculate Left Ventricular End Diastolic Volume (LVEDV), Left Ventricular End Systolic Volume (LVESV), and Left Ventricular Ejection Fraction (LVEF) [26]. Tissue Doppler Imaging was used to assess mitral annular flow velocity, and lateral mitral annular flow velocities, and annular velocities during early (e’) and late (a’) diastoles were evaluated. Early to late diastolic transmitral flow velocity (E/A) was calculated to assess diastolic function, and E to early diastolic mitral annular tissue velocity (E/e’) was calculated to estimate LV filling pressures [27]. Additionally, in patients with Regional Wall Motion Abnormalities (RWMA), both lateral and septal mitral leaflet annular velocities were measured and averaged. Tricuspid Annular Plane Systolic Excursion (TAPSE) was used to evaluate right ventricular systolic function. Moreover, the Tei index was calculated by the pulsed Doppler method as (a-b)/b. These ultrasound measurements were conducted by an experienced ultrasound specialist to avoid inter-observer bias, and to avoid intra-observer bias, an average of three consecutive recordings were taken.

To eliminate any inter-/intra- observer bias, an average of three consecutive recordings were taken and the average results were measured by an independent attending sonographer. The results and readings would be reviewed by another independent attending sonographer, and if there was a significant discrepancy, the head of the Ultrasound Department would make a final decision.

Electrocardiogram assessment

At the time of admission to the ICU (Baseline) and 72 h after the administration of Sivelestat, all patients underwent a 24-hour Holter recording using 3-channel real-time tape recorders. HRV was evaluated in both time and frequency domains [28]. During the analysis, only normal heartbeats were recorded, and all artifacts were removed. In the time domain, HRV parameters including the standard deviation of all R-R intervals (SDNN), the mean of the standard deviations of all R-R intervals for all 5-minute segments (SDANN), the square root of the mean squared differences of successive R-R intervals (RMSSD), and the percentage of adjacent normal-to-normal intervals differing by over 50 milliseconds (PNN50) were calculated [29]. In the frequency domain, heart rate power in the low-frequency range (0.04–0.15 Hz) (LF) and in the high-frequency range (0.15–0.40 Hz) (HF) were estimated. Additionally, the ratio of LF/HF was also calculated [30].

To eliminate any inter-/intra- observer bias, an average of three consecutive recordings were taken and the results were calculated automatically. Two independent researchers verify the final calculations and ultimately take the average of the three measurements.

Sample size calculation and statistical analysis

With the cardiac function index as the primary outcome, the 15.0 version of PASS software was used to calculate the sample size according to the pre-trial results. When there were 35 people in each group, the sample size could meet the statistical power of α = 0.05, β = 0.8.

All statistical analyses were performed using SPSS 24.0. Continuous variables are presented as mean ± standard deviation (SD). Analysis of serum levels, which included HMGB1, IL-6, IL-8, and TNF-a, was conducted using repeated measures analysis of variance (ANOVA) to compare within-/between- group differences. Nonparametric continuous variables were analyzed by the Mann-Whitney U-test. Categorical variables were examined using the χ2 test. Statistical significance was defined as P values < 0.05.

Results

From January 2019 to December 2021, A total of 97 patients were diagnosed with acute respiratory distress syndrome (ARDS) and septic cardiomyopathy (SCM). According to the inclusion and exclusion criteria, 70 people were eventually included with 35 in each group. (Fig. 1.)

Fig. 1
figure 1

The enrollment flow chart

Demography

No differences in age, body mass index (BMI), APACHE II score, SIRS score, SOFA score, and infection sites were observed between the two groups, P > 0.05. (Table 1).

Table 1 Characteristics of patients on baseline

ELISA results

The Sivelestat group exhibited significantly lower levels of IL-6 at various time points (12 h, 24 h, 48 h, and 72 h) compared to the Control group. The maximum difference was observed at 72 h after ICU admission (26.00 ± 5.39 pg/mL vs. 32.26 ± 6.13 pg/mL, P < 0.001; Fig. 2-A).

Fig. 2
figure 2

Changes of IL-6 (A), IL-8 (B), TNF-a (C), and HMGB-1 (D) before and after the treatment of Sivelestat IL-6: Interleukin-6; IL-8: Interleukin-8; TNF-a: Tumor necrosis factor-a; HMGB1: High-mobility group box 1 0 is the time of admission to the ICU (Baseline); 12 h, 24 h, 48 h, and 72 h are respective hours after taking the Sivelestat treatment. Data are presented as Mean ± Standard Deviation

Similarly, IL-8 levels were significantly lower in the Sivelestat group at different time points (12 h, 24 h, 48 h, and 72 h), and the Sivelestat group exhibited significantly lower IL-8 levels at 48 h after ICU admission (32.83 ± 4.27 pg/mL vs. 41.26 ± 6.13 pg/mL, P < 0.001; Fig. 2-B).

The changes in TNF-a levels also showed a consistent trend with that of IL-6 and IL-8, and the levels were significantly lower at 72 h after ICU admission (20.40 ± 2.57 pg/mL vs. 25.60 ± 4.94 pg/mL, P < 0.001; Fig. 2-C).

No significant difference was observed in HMGB1 levels between the two groups at baseline, 12 h, 24 h, and 48 h. However, the Sivelestat group showed significantly lower levels of HMGB1 at 72 h after ICU admission (19.46 ± 2.63pg/mL vs. 21.20 ± 2.03pg/mL, P = 0.003; Fig. 2-D).

Evaluation of cardiac function

The echocardiographic features of two groups have been presented in Table 2. The comparisons of reduced peak early diastolic flow velocity (peak E), maximum inferior vena cava diameter (IVCmax), and minimum inferior vena cava diameter (IVCmin) showed no significant difference between the groups. However, the stroke volume (SV) was observed to be significantly lower in the Control group in comparison to the Sivelestat group (71.62 ± 12.32 ml vs. 78.68 ± 15.24ml, P = 0.037). Furthermore, the Tei index was significantly higher in the Control group in comparison to the Sivelestat group (0.60 ± 0.08 vs. 0.56 ± 0.07, P = 0.029), while the TAPSE value was significantly lower in the Control group in comparison to the Sivelestat group (16.63 ± 3.27 vs. 18.32 ± 3.62, P = 0.044). The Control group also exhibited significantly lower E/A, e’, and a’ values in comparison to the Sivelestat group, P < 0.05. The echocardiographic images in Fig. 3 demonstrate the efficacy of Sivelestat in treating SMC.

Table 2 Echocardiographic assessment between groups
Fig. 3
figure 3

Echocardiographic imaging from Parasternal left ventricular short axis view. (A) Echocardiographic imaging at ICU admission. (B) Echocardiographic imaging at 72 h after the Sivelestat treatment

Comparisons about the electrocardiogram

The HRV indexes of the Sivelestat group and the Control group were presented in Table 3. There were no significant differences observed between the two groups, except for SDNN, LF, and LF/HF. The Control group exhibited a significant decrease in these three indices when compared to the Sivelestat group, P < 0.05.

Table 3 Comparisons of heart rate variability between the two groups

Discussion

Sepsis is a condition where the body’s immune system responds abnormally to microbial infection resulting in systemic and dysregulated inflammation and the condition is associated with high mortality rates and can progress to sepsis syndrome and septic shock, which can be potentially lethal [31, 32]. The pathophysiology of sepsis and septic shock involves the cardiovascular system, although the underlying mechanisms of sepsis-associated myocardial depression remain unclear [33]. Recent research has elucidated several mediators that participate in the development of sepsis, including TNF-α, interleukins (IL), platelet activating factor (PAF), leukotrienes, thromboxane A2, and activators of the complement cascade [34, 35]. Toll-like receptor 4 (TLR 4) has also been implicated in myocardial depression caused by sepsis, particularly in recognizing bacterial endotoxin E, Gram-negative bacterial lipopolysaccharide (LPS), or lipooligosaccharide [36]. Myocardial depressant factors recognized to date include cytokines, the complement system, nitric oxide (NO), lipopolysaccharides (LPS), and high mobility protein box 1 (HMGB1), which include tumor necrosis TNF-α, interleukin-1β (IL-1β), interleukin-6 (IL-6), and HMGB1.

According to reports, Sivelestat, a specific inhibitor of neutrophil elastase (NE), has been found to not only inhibit NE itself but also cytokine production by monocytes [37]. Additionally, it has been found to inhibit nuclear factor kappa B (NF-κB), thereby decreasing the release of cytokines including HMGB1, regulating the production of IL-8 and MCP-1 in AEC-II [38, 39]. It appears that Sivelestat is capable of binding endotoxin or affecting the action of endotoxin-binding protein, thereby blocking endotoxin-triggered TNF-alpha production by macrophages both in vitro and in vivo, and inhibiting the binding of endotoxin to Toll-like receptors [40]. Sivelestat has also shown a protective effect on ischemia-reperfusion injury by inhibiting nitric oxide release during the ischemia and reperfusion phase [41]. This effect of Sivelestat resulted in the inhibition of nitric oxide-mediated vasodilation and the subsequent suppression of increased organ blood flow during reperfusion [42]. Moreover, Sivelestat has been found to inhibit the production of cytokines in culture systems even in the absence of neutrophils. Numerous studies have highlighted the beneficial effects of Sivelestat treatment in attenuating inflammatory and edematous responses in the lungs, mitigating acute lung injuries caused by endotoxins, mitigating mesenteric ischemia-reperfusion injuries, and reducing myocardial injuries following cardioplegic arrest [43]. Results of animal-based experiments have revealed that Sivelestat appears to have a cardioprotective effect against myocardial depression seen following global ischemia. The mechanism underlying the outcome could be associated with decreased formation of reactive oxygen species (ROS) and conservation of nitric oxide [44]. In another study, Toyama et al. reported an improvement not only in respiratory index but also the fractional area of the left ventricle during pediatric cardiovascular surgery with cardiopulmonary bypass, after administering Sivelestat [45]. Nonetheless, its effectiveness in preserving cardiac function during sepsis remains enigmatic in clinical practice. Although the promise of sivelestat is exciting, a number of questions remain. The timing and duration of sivelestat intervention may be crucial to its ultimate success. However, in the STRIVE study, some adverse events occurred, such as hypersensitivity, hepatobiliary disorders, blood and lymphatic system disorders, renal and urinary disorders. Although prespecified stopping guidelines were not met, a negative trend in long-term mortality prompted the DSMB to recommend suspension of enrollment and discontinuation of study drug [14]. To date, available clinical study data, including for the STRIVE study and the related postmarketing study, indicate no particular concerns regarding adverse events. The occurrence of adverse events must be confirmed in larger prospective RCTS and should be assessed over a longer period of follow-up.

Our study has demonstrated that administering Sivelestat immediately after SCM diagnosis resulted in a significant reduction in inflammatory factor levels such as IL-6, IL-8 and TNF-a, thereby improving SCM condition. These findings suggest that early Sivelestat treatment could be effective in managing SCM. Since the severity of organ dysfunction in SCM is associated with outcomes, attenuating SCM with Sivelestat may lead to a significant improvement in morbidity and mortality. Therefore, in our future studies, we plan to investigate the effect of Sivelestat on long-term prognosis and make a comparison to evaluate its effectiveness.

There are still some limitations in our study. First, statistical data related to prognosis are absent and no comparisons of mortality rate are made. Second, comparisons of treatment costs between groups are not studied. Third, the follow-up period is relatively short. In addition, the sample size of this study is relatively small due to the research topic, research funding and time constraints. We will increase the multi-center study and expand the sample size in the follow-up study to eliminate systematic bias and make the research results more rigorous and accurate.

Conclusions

Sivelestat can significantly reduce the level of plasma inflammatory factors, improve cardiac function, and reduce heart rate variability in patients with sepsis-induced ARDS and SCM.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Hayakawa M, Katabami K, Wada T, et al. Sivelestat (selective neutrophil elastase inhibitor) improves the mortality rate of sepsis associated with both acute respiratory distress syndrome and disseminated intravascular coagulation patients. Shock Augusta Ga. 2010;33(1):14–8.

    Article  CAS  PubMed  Google Scholar 

  2. Park I, Kim M, Choe K, et al. Neutrophils disturb pulmonary microcirculation in sepsis-induced acute lung injury. Eur Respir J. 2019;53(3):1800786.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. L’Heureux M, Sternberg M, Brath L, Turlington J, Kashiouris MG. Sepsis-Induced Cardiomyopathy: a Comprehensive Review. Curr Cardiol Rep. 2020;22(5):35.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Fein AM, Calalang-Colucci MG. Acute lung injury and acute respiratory distress syndrome in sepsis and septic shock. Crit Care Clin. 2000;16(2):289–317.

    Article  CAS  PubMed  Google Scholar 

  5. Sevransky JE, Martin GS, Shanholtz C, et al. Mortality in sepsis versus non-sepsis induced acute lung injury. Crit Care Lond Engl. 2009;13(5):R150.

    Article  Google Scholar 

  6. Mikolka P, Kosutova P, Balentova S, et al. Early cardiac injury in acute respiratory distress syndrome: comparison of two experimental models. Physiol Res. 2020;69(Suppl 3):S421–32.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Chen H, Liu Q, Liu X, Jin J. Berberine attenuates septic cardiomyopathy by inhibiting TLR4/NF-κB signalling in rats. Pharm Biol. 2021;59(1):121–8.

    Article  CAS  PubMed  Google Scholar 

  8. Zhang J, Wang L, Xie W, et al. Melatonin attenuates ER stress and mitochondrial damage in septic cardiomyopathy: a new mechanism involving BAP31 upregulation and MAPK-ERK pathway. J Cell Physiol. 2020;235(3):2847–56.

    Article  CAS  PubMed  Google Scholar 

  9. Guérin C, Thompson T, Brower R. The ten diseases that look like ARDS. Intensive Care Med. 2015;41(6):1099–102.

    Article  PubMed  Google Scholar 

  10. Vincelj J. [SEPTIC CARDIOMYOPATHY]. Acta Med Croat Cas Hravatske Akad Med Znan. 2015;69(3):177–82.

    CAS  Google Scholar 

  11. Liu M, Zhang J, Dong L, et al. Detection of sivelestat and its metabolite in small volumes of plasma from Chinese ALI/ARDS patients with SIRS via high-throughput UPLC-MS/MS: a pharmacokinetic study. J Pharm Biomed Anal. 2021;195:113876.

    Article  CAS  PubMed  Google Scholar 

  12. Gao X, Zhang R, Lei Z, et al. Efficacy, safety, and pharmacoeconomics of sivelestat sodium in the treatment of septic acute respiratory distress syndrome: a retrospective cohort study. Ann Palliat Med. 2021;10(11):11910–7.

    Article  PubMed  Google Scholar 

  13. Kambe M, Bessho R, Fujii M, Ochi M, Shimizu K. Sivelestat reduces myocardial ischemia and reperfusion injury in rat hearts even when administered after onset of myocardial ischemia. Interact Cardiovasc Thorac Surg. 2009;8(6):629–34.

    Article  PubMed  Google Scholar 

  14. Takeda Y, Tamaoki J, Yamawaki I, et al. Role of neutrophil elastase in allergen-induced airway microvascular leakage in sensitized guinea pigs. Arerugi. 1997;46(6):496–501.

    CAS  PubMed  Google Scholar 

  15. Effect of human neutrophil elastase on. Tracheal mucociliary transport in anesthetized quails. Jpn J Pharmacol. 1997;75(4):439–42.

    Article  Google Scholar 

  16. Haga Y, Ogawa M. Neutrophil elastase inhibitor (ONO-5046Na) decreases production of proinflammatory cytokines by lipopolysaccharide-stimulated human monocytes. Res Commun Mol Pathol Pharmacol. 1997;98(3):243–8.

    CAS  PubMed  Google Scholar 

  17. Nie H, Xiong Q, Lan G, et al. Sivelestat alleviates atherosclerosis by improving intestinal barrier function and reducing Endotoxemia. Front Pharmacol. 2022;13:838688.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bréchot N, Hajage D, Kimmoun A, et al. Venoarterial extracorporeal membrane oxygenation to rescue sepsis-induced cardiogenic shock: a retrospective, multicentre, international cohort study. Lancet Lond Engl. 2020;396(10250):545–52.

    Article  Google Scholar 

  19. Sartelli M, Kluger Y, Ansaloni L, et al. Raising concerns about the Sepsis-3 definitions. World J Emerg Surg WJES. 2018;13:6.

    Article  PubMed  Google Scholar 

  20. Li Y, Lu B, Yu M, Zhai J, Yao Y, Chai Y. Diagnostic value and significance of serum miR-132 combined with miR-223 for sepsis-induced cardiomyopathy. Exp Ther Med. 2021;22(6):1396.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Song J, Moon S, Park DW, et al. Biomarker combination and SOFA score for the prediction of mortality in sepsis and septic shock: a prospective observational study according to the Sepsis-3 definitions. Med (Baltim). 2020;99(22):e20495.

    Article  Google Scholar 

  22. Hussain M, Xu C, Ahmad M, et al. Acute respiratory distress syndrome: bench-to-Bedside approaches to improve Drug Development. Clin Pharmacol Ther. 2018;104(3):484–94.

    Article  PubMed  Google Scholar 

  23. Lu NF, Jiang L, Zhu B, et al. Elevated plasma histone H4 levels are an important risk factor in the development of septic cardiomyopathy. Balk Med J. 2020;37(2):72–8.

    CAS  Google Scholar 

  24. Hoshi K, Kurosawa S, Kato M, Andoh K, Satoh D, Kaise A. Sivelestat, a neutrophil elastase inhibitor, reduces mortality rate of critically ill patients. Tohoku J Exp Med. 2005;207(2):143–8.

    Article  CAS  PubMed  Google Scholar 

  25. Hennø LT, Storjord E, Christiansen D, et al. Effect of the anticoagulant, storage time and temperature of blood samples on the concentrations of 27 multiplex assayed cytokines - consequences for defining reference values in healthy humans. Cytokine. 2017;97:86–95.

    Article  PubMed  Google Scholar 

  26. Tamborini G, Piazzese C, Lang RM, et al. Feasibility and accuracy of Automated Software for Transthoracic three-Dimensional Left Ventricular volume and function analysis: comparisons with two-Dimensional Echocardiography, three-Dimensional Transthoracic Manual Method, and Cardiac magnetic resonance imaging. J Am Soc Echocardiogr off Publ Am Soc Echocardiogr. 2017;30(11):1049–58.

    Article  Google Scholar 

  27. Mitter SS, Shah SJ, Thomas JD. A test in Context: E/A and E/e’ to assess diastolic dysfunction and LV filling pressure. J Am Coll Cardiol. 2017;69(11):1451–64.

    Article  PubMed  Google Scholar 

  28. Lees T, Shad-Kaneez F, Simpson AM, Nassif NT, Lin Y, Lal S. Heart Rate Variability as a Biomarker for Predicting Stroke, Post-stroke complications and Functionality. Biomark Insights. 2018;13:1177271918786931.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Alcantara JMA, Plaza-Florido A, Amaro-Gahete FJ, et al. Impact of using different levels of threshold-based artefact correction on the quantification of Heart Rate Variability in three independent human cohorts. J Clin Med. 2020;9(2):325.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Franco OS, Júnior AOS, Signori LU, Prietsch SOM, Zhang L. Cardiac autonomic modulation assessed by heart rate variability in children with asthma. Pediatr Pulmonol. 2020;55(6):1334–9.

    Article  PubMed  Google Scholar 

  31. de Souza DC, Machado FR. Epidemiology of Pediatric septic shock. J Pediatr Intensive Care. 2019;8(1):3–10.

    Article  PubMed  Google Scholar 

  32. Naime ACA, Ganaes JOF, Lopes-Pires ME, Sepsis. The involvement of platelets and the current treatments. Curr Mol Pharmacol. 2018;11(4):261–9.

    Article  PubMed  Google Scholar 

  33. Hai PD, Phuong LL, Dung NM, et al. Subclinical left ventricular systolic dysfunction in patients with septic shock based on Sepsis-3 definition: a speckle-tracking Echocardiography Study. Crit Care Res Pract. 2020;2020:6098654.

    PubMed  PubMed Central  Google Scholar 

  34. Zhang C, Wang K, Yang L, et al. Lipid metabolism in inflammation-related diseases. Analyst. 2018;143(19):4526–36.

    Article  CAS  PubMed  Google Scholar 

  35. Ravikumar N, Sayed MA, Poonsuph CJ, Sehgal R, Shirke MM, Harky A. Septic cardiomyopathy: from basics to management choices. Curr Probl Cardiol. 2021;46(4):100767.

    Article  PubMed  Google Scholar 

  36. Wang Y, Zhang L, Zhao X, Yang W, Zhang R. An experimental study of the protective effect of simvastatin on sepsis-induced myocardial depression in rats. Biomed Pharmacother Biomedecine Pharmacother. 2017;94:705–11.

    Article  CAS  Google Scholar 

  37. Akamoto S, Okano K, Sano T, et al. Neutrophil elastase inhibitor (sivelestat) preserves antitumor immunity and reduces the inflammatory mediators associated with major surgery. Surg Today. 2007;37(5):359–65.

    Article  CAS  PubMed  Google Scholar 

  38. Ogawa R, Mori R, Iida K, et al. Effects of the early administration of sivelestat sodium on bronchopulmonary dysplasia in infants: a retrospective cohort study. Early Hum Dev. 2017;115:71–6.

    Article  CAS  PubMed  Google Scholar 

  39. Sahebnasagh A, Saghafi F, Safdari M, et al. Neutrophil elastase inhibitor (sivelestat) may be a promising therapeutic option for management of acute lung injury/acute respiratory distress syndrome or disseminated intravascular coagulation in COVID-19. J Clin Pharm Ther. 2020;45(6):1515–9.

    Article  CAS  PubMed  Google Scholar 

  40. Misumi T, Tanaka T, Mikawa K, Nishina K, Morikawa O, Obara H. Effects of sivelestat, a new elastase inhibitor, on IL-8 and MCP-1 production from stimulated human alveolar epithelial type II cells. J Anesth. 2006;20(3):159–65.

    Article  PubMed  Google Scholar 

  41. Nomura N, Asano M, Saito T, Nakayama T, Mishima A. Sivelestat attenuates lung injury in surgery for congenital heart disease with pulmonary hypertension. Ann Thorac Surg. 2013;96(6):2184–91.

    Article  PubMed  Google Scholar 

  42. Horinokita I, Hayashi H, Yoshizawa R, et al. Possible involvement of progranulin in the protective effect of elastase inhibitor on cerebral ischemic injuries of neuronal and glial cells. Mol Cell Neurosci. 2021;113:103625.

    Article  CAS  PubMed  Google Scholar 

  43. Fujii M, Bessho R. Neutrophil elastase inhibitor Sivelestat attenuates myocardial Injury after Cardioplegic arrest in Rat hearts. Ann Thorac Cardiovasc Surg off J Assoc Thorac Cardiovasc Surg Asia. 2020;26(5):263–9.

    Google Scholar 

  44. Akiyama D, Hara T, Yoshitomi O, Maekawa T, Cho S, Sumikawa K. Postischemic infusion of sivelestat sodium hydrate, a selective neutrophil elastase inhibitor, protects against myocardial stunning in swine. J Anesth. 2010;24(4):575–81.

    Article  PubMed  Google Scholar 

  45. Toyama S, Hatori F, Shimizu A, Takagi T. A neutrophil elastase inhibitor, sivelestat, improved respiratory and cardiac function in pediatric cardiovascular surgery with cardiopulmonary bypass. J Anesth. 2008;22(4):341–6.

    Article  PubMed  Google Scholar 

Download references

Funding

This study was supported by the National Natural Science Foundation of China (81700339), and the national key research and development program of China (No.2016YFA0101100,2016YFA0100900).

Author information

Authors and Affiliations

Authors

Contributions

Hui Lv and Xiaoke Shang contributed to the conception and design of the study; Langjing Huang and Xiuhong Yang performed the experimentsï¼›Changdong Zhang and Yao Yu collected and analyzed data; Hui Lv and Xiaoke Shang wrote the manuscript; All authors reviewed and approved the final version of the manuscript.

Corresponding author

Correspondence to Xiaoke Shang.

Ethics declarations

Ethics approval and consent to participate

The study approved by the ethics committee of Wuhan Union Hospital (No. 2018-0598-08). We certify that the study was performed in accordance with the 1964 declaration of HELSINKI and later amendments. All methods were carried out in accordance with relevant guidelines and regulations. Informed consents were obtained from all subjects and/or their legal guardians.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lv, H., Huang, L., Yang, X. et al. The clinical effectiveness of sivelestat in treating sepsis patients with both acute respiratory distress syndrome and septic cardiomyopathy. J Cardiothorac Surg 19, 399 (2024). https://doi.org/10.1186/s13019-024-02835-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13019-024-02835-3

Keywords