#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Neuromuscular blockers in the acute respiratory distress syndrome: A meta-analysis


Authors: Yusi Hua aff001;  Xiaofeng Ou aff002;  Qian Li aff001;  Tao Zhu aff001
Authors place of work: Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China aff001;  Department of Critical Care, West China Hospital of Sichuan University, Chengdu, Sichuan, China aff002
Published in the journal: PLoS ONE 15(1)
Category: Research Article
doi: https://doi.org/10.1371/journal.pone.0227664

Summary

Background

The effects of neuromuscular blocking agents (NMBAs) on adult patients with acute respiratory distress syndrome (ARDS) remain unclear. We performed a meta-analysis of randomized controlled trials (RCTs) to evaluate its effect on mortality.

Methods

We searched the Cochrane (Central) database, Medline, Embase, the Chinese Biomedical Literature Database (SinoMed), WanFang data and ClinicalTrials from inception to June 2019, with language restriction to English and Chinese. We included published RCTs and eligible clinical trials from ClinicalTrials.gov that compared NMBAs with placebo or usual treatment in adults with ARDS. We pooled data using random-effects models. The primary outcome was mortality. The secondary outcomes were the ratio of the partial pressure of arterial oxygen to the fraction of inspired oxygen (PaO2/FIO2), total positive end expiratory pressure (PEEP), plateau pressure (Pplat), days free of ventilator at day 28, barotrauma and ICU-acquired weakness.

Results

We included 6 RCTs (n = 1557). Compared with placebo or usual treatment, NMBAs were associated with lower 21 to 28-day mortality (RR 0.72, 95% CI 0.53–0.97, I2 = 59%). NMBAs significantly improved oxygenation (Pao2:Fio2 ratios) at 48 hours (MD 27.26 mm Hg, 95% CI 1.67, 52.84, I2 = 92%) and reduced the incidence of barotrauma (RR 0.55, 95% CI 0.35, 0.85, I2 = 0). However, NMBAs had no effect on oxygenation (Pao2:Fio2 ratios) (MD 18.41 mm Hg, 95% CI -0.33, 37.14, I2 = 72%) at 24 hours. We also found NMBAs did not affect total PEEP, plateau pressure, days free of ventilation at day 28 and ICU-acquired weakness.

Conclusions

In patients with moderate-to-severe ARDS, the administration of NMBAs could reduce 21 to 28-day mortality and barotrauma, and improve oxygenation at 48 hours, but have no significant effects on 90-day/ICU mortality, days free of ventilation at day 28 and the risk of ICU-acquired weakness. Further large-scale, high-quality RCTs are needed to confirm our findings. Registration: PROSPERO (ID: CRD 42019139656).

Keywords:

Sequence analysis – Death rates – Metaanalysis – Oxygen – Intensive care units – Randomized controlled trials – Acute respiratory distress syndrome – Neuromuscular blockade

Introduction

Acute respiratory distress syndrome (ARDS) is a life-threatening condition characterized by intense lung inflammation, consolidation, and progressive microatelectasis with refractory acute hypoxemia [1, 2]. Despite advances in medical equipments and clinical managements, the incidence and mortality of ARDS are still high [35]. Management of ARDS is a multimodal strategy involves non-pharmacologic interventions and pharmacologic interventions. Non-pharmacologic interventions include protective ventilation strategies, higher positive end-expiratory pressure (PEEP) and prone positioning, and these strategies are accepted because of their beneficial effects on patients with ARDS [2, 5, 6]. While, of the pharmacologic interventions, there are many inconclusive opinions remained.

The use of neuromuscular blocking agents (NMBAs) in ARDS has remained controversial. NMBAs were used in ICU mainly for facilitating lung-protective ventilation, preventing patient–ventilator dyssynchrony. Clinicians commonly consider NMBAs could reduce barotraumas, minimize the work of breathing and improve oxygenation [79]. In the early 2000s, a small, randomized trial conducted in France demonstrated continuous cisatracurium therapy could improve oxygenation of ARDS [8]. After two years, the same group of investigators conducted another randomized controlled trial (RCT) and founded cisatracurium could significantly reduce ARDS patients’ inflammatory biomarkers in both the blood and bronchoalveolar fluid, along with improved oxygenation [10]. In 2010, they reported a large multicenter trial (the ACURASYS trial) of 339 patients that the early administration of cisatracurium in patients with moderate-to-severe ARDS was associated with lower hospital mortality [11]. So, NMBAs have been recommended as clinical practice guideline for the management of severe ARDS and mechanical ventilation patients under certain circumstances [12, 13]. However, despite these encouraging results, the use of NMBAs did not be suggested as a clinical practice guideline for mechanical ventilation in adult patients with ARDS [14], and early neuromuscular blockade was also not widely adopted and strongly recommended in current guidelines [4, 1517]. Resource constraints and limited data about the effects of NMBAs on neuromuscular function and other long-term outcomes may be the main potential concerns.

Some meta-analyses have reported that NMBAs play protective effects on ARDS patients [9, 18, 19]. However, the three included RCTs [8, 10, 11] were conducted in France by the same research group using the same NMBA, cisatracurium. In addition, some limitations including small sample sizes, poor quality trials and the narrative synthesis of data were prone to generate bias and heterogeneity. So, the results of the three meta-analyses should be reassessed. Moreover, recently, the largest multicenter, unblinded, randomized trial of 1006 patients with moderate-to-severe ARDS, the Reevaluation of Systemic Early Neuromuscular Blockade (ROSE) trial, was conducted by The Prevention and Early Treatment of Acute Lung Injury (PETAL) Clinical Trials Network of the National Heart, Lung, and Blood Institute (NHLBI). The investigators of that trial reported that there was no significant difference in mortality at 90 days between patients who received an early and continuous cisatracurium infusion and those who were treated with a usual-care approach with lighter sedation targets [20].

Therefore, based on these controversial findings related to NMBAs administration in adult patients with ARDS, we conducted a meta-analysis of RCTs to identify the benefits and adverse effects of NMBAs in ARDS patients.

Methods

We conducted this study and reported the findings according to the guidelines recommended by the Cochrane Collaboration for Systematic Reviews of Interventions [21] and the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement methodology [22] respectively. The protocol has been registered on PROSPERO (ID: CRD 42019139656).

Literature search

We performed a computerized literature searches included Medline, Embase, CENTRAL (from inception to June 2019), the Chinese Biomedical Literature Database (SinoMed) (from 1978 to June 2019), and WanFang data (from 1990 to 2019), with language restriction to English and Chinese. We also searched ClinicalTrials.gov in June 2019 to identify additional eligible clinical trials for preliminary and unpublished results by contacting with authors if necessary. The exact search strategy is provided in S1 File.

Study selection

After titles screening, we evaluated abstracts for relevance and identified them as included, excluded, or requiring further assessment. We considered randomized controlled studies (RCTs) eligible if they compared the administration of any NMBAs with placebo or usual treatment and included critical adult patients with ARDS, who were undergoing mechanical ventilation through an endotracheal tube and the ratio of the partial pressure of arterial oxygen (PaO2) to the fraction of inspired oxygen (FiO2) was less than 200 with the ventilator set to deliver a positive end-expiratory pressure of 5 cm of water or higher. We included studies only if a full text was available, with the interventions of interest were NMBAs, irrespective of the type, dose or duration. We excluded animal studies, observational studies, preclinical studies and trials of pediatric patients. We also excluded studies published in narrative reviews, commentaries, editorials and case reports.

Data extraction and quality assessment

The primary outcome was mortality (included 21 to 28-day mortality and 90-day/ICU mortality). The secondary outcomes were the ratio of the partial pressure of arterial oxygen to the fraction of inspired oxygen (PaO2/FIO2) at 24 hours and 48 hours, total positive end expiratory pressure (PEEP) at 24 hours and 48 hours, plateau pressure (Pplat) at 24 hours and 48 hours, days free of ventilator at day 28, barotrauma (including pneumothorax, pneumomediastinum, pneumatocele, and subcutaneous emphysema) and ICU-acquired weakness.

Two investigators (Y.H. and X.O.) independently performed an initial screening according to titles or abstracts review, followed by a full-text screening. Detailed study information, study methods, methodologic quality, and outcomes were extracted using a standardized data extraction form. Disagreements were resolved by consensus or by a discussion with a third author (T.Z.). We assessed the risk of bias for each study using the Cochrane collaboration tool to judge the adequacy of random sequence generation, allocation concealment, blinding of participants and personnel, blinding of outcome assessment, incomplete outcome data, selective reporting and other bias [23]. The risk of bias in each of these domains was classified as high risk, low risk, or unclear. The overall risk of bias for an individual trial was categorized as low when the risk of bias was low in all domains; unclear when the risk of bias was unclear in at least one domain, with no high-risk domains; or high when the risk of bias was high in at least one domain [23].

Statistical analysis

We performed all statistical analyses in the present study using Review Manager 5.3 (RevMan, The Cochrane Collaboration, Oxford, United Kingdom) and random-effects models. Risk ratios (RRs) with 95% confidence intervals (CIs) were calculated for binary outcomes, while mean differences (MDs) with 95% CIs were calculated for continuous outcomes. A p value of less than 0.05 was considered to be statistically significant. Data were obtained by direct extraction or by indirect calculation. We converted date reported as median and measure of dispersion to mean and standard deviation assuming a normal distribution by applying two simple formulae [24].

We assessed clinical and methodological heterogeneity according to the study characteristics and statistical heterogeneity by using the Mantel-Haenszel χ2 test and the I2 statistic. We considered heterogeneity to be substantial if the I2 value was 50% or greater or the p value was 0.1 or less [25, 26]. We assessed sensitivity analyses by using alternative effect measures (odds ratios v. risk ratios) and statistical models regarding heterogeneity (random v. fixed effects). For example, to assess the robustness of our primary results (mortality) in sensitivity analyses, we used fixed-effects models and two alternative statistical metrics: odds ratios and risk differences. Because of the different reported durations about mortality, PaO2/FIO2, total PEEP and Pplat, we analyzed mortality as tow subgroups, 21 to 28-day mortality and 90-day/ICU mortality, and we analyzed changes in PaO2/FIO2, total PEEP and Pplat at 24 and 48 hours. To assess the effect of NMBAs on the duration of ventilation, we analyzed days free of ventilator at day 28. We assessed the quality of evidence for clinical outcomes by using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach [27]. Publication bias was not assessed because of the low power associated with the low number of included trails.

Results

Search results and study characteristics

We identified 2049 citations through electronic searches. After screening the titles and abstracts, we reviewed 38 studies in full, and 32 trials were excluded. The details of the 32 excluded trials and reasons for their exclusion are shown in S1 File. Finally, we included 6 RCTs [8, 10, 11, 20, 28, 29] with a total of 1557 patients in the meta-analysis (Fig 1).

Search and selection of randomized controlled trials (RCTs).
Fig. 1. Search and selection of randomized controlled trials (RCTs).

The characteristics of the 6 included trials are shown in Table 1. Four of the trials included 455 patients (29.2%) were conducted in France by the same group of investigators [8, 11, 12, 29], the biggest trial was conducted in 48 hospitals across the United States with 1006 patients [20]. Population sizes of included studies varied greatly, ranging from 24 to 1006, and four trials included less than 100 patients [8, 10, 28, 29]. Most of the studies (5 trials included 1461 patients) were treated with cisatracurium [8, 10, 11, 20, 29], except one study performed in China received vecuronium [28]. All patients met the criteria for moderate to severe ARDS (the moderate to severe ARDS was defined as a baseline PaO2/FIO2≤ 200 mmHg [1]). The primary outcomes of three trials [11, 20, 28] were mortality, consistent with our study. The other three trials considered gas exchange (PaO2/FIO2 ratio) [1], inflammatory response [10] and transpulmonary pressures [29] as their primary outcomes respectively. Five trials reported 21 to 28-day mortality [8, 10, 20, 28], ICU/90-day mortality[8, 10, 20, 29], PaO2/FIO2 ratio at 24 hours[8, 10, 20, 29], PaO2/FIO2 ratio at 48 hours[8, 10, 20, 28, 29], total PEEP at 24 hours[8, 10, 11, 20, 29], Pplat at 24 hours[8, 10, 11, 20, 29] and days free of ventilation at day 28 [8, 10, 11, 20, 29]. Four trials reported total PEEP at 48 hours [8, 10, 20, 29], Pplat at 48 hours [8, 10, 20, 29], barotraumas [8, 10, 11, 20], and ICU-acquired weakness [8, 10, 11, 20].

Tab. 1. Characteristics of included trials.
Characteristics of included trials.

As shown in Table 2 and Fig 2, risks of bias were summarized by the Cochrane Risk of Bias tool; three trials [8, 10, 20] were judged to be at high risk of bias because of limitations in blinding; one trial was categorized as a lower risk of bias [11] with adequate randomized sequences, concealed allocation, blinding and completely reported clinical outcomes. Two trials [28, 29] were assessed to be unclear because insufficient data about whether blinding and concealed allocation were available. According to the GRADE approach, we judged the quality of evidence related to most of clinical outcomes included mortality, PaO2/FIO2 at 24 hours and at 48 hours, total PEEP at 24 hours and at 48 hours, Pplat at 24 hours and 48 hours, days free of ventilator at day 28 and barotrauma as moderate in light of the limitations in blinding and allocation concealment. We judged the quality of evidence related to ICU-acquired weakness as weak because of some imprecision.

Risk of bias summary.
Fig. 2. Risk of bias summary.
Tab. 2. Risk of bias assessment.
Risk of bias assessment.

Mortality

Twenty-one to twenty-eight-day mortality

Five RCTs including 1,533 patients reported data on twenty-one to twenty-eight-day mortality with 32.38% in the NMBA group and 37.58% in the control group. Compared with the control oxygen group, there was a statistically significant reduction of mortality in the NMBA group (RR 0.72, 95% CI 0.53–0.97, I2 = 59%) (Fig 3). Considering four of the included trials were treated with cisatracurium, except the China trial [28] received vecuronium, we omitted the China trial and found that there was no significant difference between the NMBA group and the control group (RR 0.76, 95% CI 0.56–1.04, I2 = 58%) (S1 Fig).

Comparison of mortality between the NMBAs group and Control group: 21 to 28-day mortality and 90-day/ICU mortality.
Fig. 3. Comparison of mortality between the NMBAs group and Control group: 21 to 28-day mortality and 90-day/ICU mortality.

Ninety-day/ICU mortality

In term of 90-day/ICU mortality, there were five trials included in this analysis with 1461 patients, which were treated with cisatracurium. We found no significant difference between the NMBA group and the control group (RR 0.82, 95% CI 0.65–1.05, I2 = 46%) (Fig 3).

Secondary outcomes

The secondary outcomes were summarized in Table 3. Considering the time effect of NMBAs, We analyzed PaO2/FIO2, total PEEP and Pplat at 24 hours and 48 hours respectively.

Tab. 3. Pooled analysis of secondary outcome measures.
Pooled analysis of secondary outcome measures.

Data on PaO2/FIO2 ratios at 24 hours was available from 5 trials (n = 1291). No significant difference was found between the NMBA group and the control group (MD 18.41 mm Hg, 95% CI -0.33, 37.14, I2 = 72%). While at 48 hours, the pooled analysis from 5 trials with 941 patients suggested better PaO2/FIO2 in the NMBA group and the difference was statistically significant compared with the control group (MD 27.26 mm Hg, 95% CI 1.67, 52.84, I2 = 92%).

A total of 5 trials with 1407 patients and 4 trials with 1006 patients were included in the analysis of total PEEP at 24 hours and 48 hours respectively. No significant differences were found between the NMBA group and the control group neither at 24 hours (MD -0.20 cmH2O, 95% CI -0.86, 0.46, I2 = 55%) nor at 48 hours (MD -0.55 cmH2O, 95% CI -1.37, 0.28, I2 = 67%).

A total of 5 trials with 1158 patients and 4 trials with 761 patients were included in the analysis of Pplat at 24 hours and 48 hours respectively. No significant differences were found between the NMBA group and the control group neither at 24 hours (MD 0.05 cm H2O, 95% CI -0.97, 1.06, I2 = 42%) nor at 48 hours (MD -0.08 cm H2O, 95% CI -0.90, 0.74, I2 = 0).

With respect to the effects of NMBAs on days free of ventilator at day 28 (5 trials; n = 1461), we found no significant difference between the NMBA group and the control group (MD 0.72 days, 95% CI -0.49, 1.93, I2 = 12%).

About adverse events, 4 trials (n = 1437) described data on barotrauma. There were 29 (4.01%) patients who developed barotrauma in the NMBA group, 52 (7.29%) in the control group. Pooled analyses of the results showed it was significantly lower in the NMBA group than the control group (RR 0.55, 95% CI 0.35, 0.85, I2 = 0). Four trials (n = 1299) mentioned the incidence of ICU-acquired weakness. There were 50 (7.59%) patients in the NMBA group and 39 (6.09%) in the control group suffered ICU-acquired weakness, the difference was not significant (RR 1.09, 95% CI 0.76, 1.56, I2 = 0). There was no heterogeneity among the included trials about barotrauma and ICU-acquired weakness (I2 = 0, p = 0.70 and I2 = 0, p = 0.90, respectively).

Sensitivity analysis

Sensitivity analyses by using alternative effect measures (odds ratios v. risk ratios) and statistical models regarding heterogeneity (random v. fixed effects) generated statistically similar primary results, with statistically significant reductions in 21 to 28-day mortality and similar effects about 90-day/ICU mortality between the NMBA group and the control group (S2S4 Figs). The ROSE trial [20] was the largest and contributed the greatest weight to the results of our meta-analysis. By omitting the ROSE trial, we found that the effect of NMBA on 90-day/ICU mortality has changed from the same as the control group to statistically significant reduction effect. (RR, 0.72; 95% CI, 0.57, 0.91; P = 0.007; I2 = 0).

Discussion

In this meta-analysis of randomized controlled trials for adult patients with moderate-to-severe ARDS, we found that the treatment of continuous infusion of NMBAs was associated with a lower risk of death at 21 to 28-day, but has no beneficial effects on 90-day/ICU mortality. Moreover, NMBAs treatment can improve PaO2/FIO2 at 48 hours, reduced the risk of barotrauma, and did not affect PaO2/FiO2 at 24 hours, total PEEP, plateau pressure, days free of ventilation at day 28 and ICU-acquired weakness.

Several systematic reviews and meta-analyses evaluated the effects of NMBAs on ARDS patients [9, 18, 19] and they all suggested that NMBAs treatment can improve outcomes in either primary or secondary measures. However, all the previous reviews and meta-analyses were mainly based on three RCTs [8, 10, 11], which were conducted in France by one research group and sued the same NMBA, cisatracurium. Different from previous studies, our meta-analysis had some characteristics. Firstly, this meta-analysis contains comprehensive outcomes: 21 to 28-day mortality, ICU/90-day mortality, PaO2/FIO2 ratio at 24 hours and 48 hours, total PEEP at 24 hours and 48 hours, Pplat at 24 hours and 48 hours, Days free of ventilation at day 28, barotraumas and ICU-acquired weakness. Secondly, based on the current available data, this study was the largest meta-analysis with 6 RCTs and 1557 patients. The sample sizes of previous studies were too small to accurate assessment of these outcomes, and the latest and largest RCT [20] could help to confirm the effects of NMBAs on ARDS. Inadequate number of patients and missing studies may affect the outcomes of NMBAs. Thirdly, in the present meta-analysis, we included the largest RCT [20] conducted in the United States and another RCT [28] using vecuronium, not cisatracurium, which may have substantial effects on the synthetic outcomes of NMBAs.

In terms of the 21 to 28-day mortality reduction associated with NMBAs therapy, there was high heterogeneity (chi2 = 9.69, df = 4, P = 0.05, I2 = 59%) among the included studies, which might because of the heterogeneous population and different NMBAs were used. One of the five included trials [28] conducted in China were treated with vecuronium. By omitting the China trial, we found that there was no significant difference between the NMBA group and the control group (RR 0.76, 95% CI 0.56–1.04, I2 = 58%). So, the decisive conclusion should be made cautiously and further large-scale, multicenter studies are needed to confirm the result. Our meta-analysis showed that there was no significant difference about 90-day/ICU mortality between patients treated with NMBA and patients not treated with NMBA. The use of NMBA usually requires deep sedation, and itself can result in negative outcomes [15, 30, 31]. Unlike other trials, the largest trial included in our study applied lighter sedation strategy in the control group, which may have decreased mortality in that group [20].

The improvements in the PaO2/FIO2 ratio at 48 hours and barotrauma were consistent with the results of some clinical studies and systematic researches [8, 9, 19, 32], although the potential mechanisms have not been entirely characterized. It is suggested that NMBAs block neuromuscular transmission of respiratory muscles, resulting in reducing patient-ventilator dyssynchrony, barotrauma, oxygen consumption, and the accumulation of alveolar fluid [7, 33, 34].

One of the main safety concerns with the administration of neuromuscular blocking agents is muscle weakness, sustained administration of neuromuscular blockade is associated with subsequent neuromuscular weakness [35, 36]. The risk of ICU-acquired weakness associated with NMBAs poses a strong resistance to NMBA usage in the current management of ARDS [32, 37]. Although NMBAs (cisatracurium besylate and vecuronium) have been reported with myopathy[32, 37], our study showed that there were 50 patients (7.59%) occurred ICU-acquired weakness in the NMBA group and 39 patients (6.1%) in the control group, and no significant difference was found between them (RR 1.09, 95% CI 0.76–1.56, I2 = 0%). This result maybe could be explained by the short duration of use of the neuromuscular blockades.

There are some limitations in our meta-analysis. First, our study was based on relatively few trials and 4 of them had small samples (< 100) [8, 10, 28, 29], which may have reduced precision and underestimated heterogeneity. In addition, different sedation strategy was applied in the control group, such as lighter sedation strategy conducted in the ROSE trial [20] may cause potential bias and heterogeneity. Second, different neuromuscular blockades may have different effects. An observational study compared cisatracurium and vecuronium reported that administration with cisatracurium was associated with more days free of ventilation and days not in ICU compared with vecuronium [38]. So, subgroup analyses stratified by different neuromuscular blockades even dosing strategies and specific groups of patients such as trauma, sepsis, pneumonia, and other causes should be planned and performed. Finally, most of the included trials were not double-blinded, because nurses, physiotherapists and other health care professionals were aware of the treatment assignments. The lack of double-blinding may have influenced short-term assessments of neuromuscular function, and the reporting of adverse events.

Conclusion

In patients with moderate-to-severe ARDS, the administration of NMBAs could reduce 21 to 28-day mortality, but have no significant effect on 90-day/ICU mortality.

In addition, continuous infusion of NMBAs could improve PaO2/FIO2 ratios at 48 hours, reduce the incidence of barotrauma, without increasing days free of ventilation at day 28 and the risk of ICU-acquired weakness. The effects of NMBAs on ARDS patients should be re-evaluated.

Supporting information

S1 Checklist [doc]
PRISMA checklist.

S1 Fig [eps]
Comparison of mortality in four included trials treated with cisatracurium.

S2 Fig [eps]
Sensitivity analysis of mortality by using alternative effect measure risk ratio and fixed effects model in randomized controlled trials.

S3 Fig [eps]
Sensitivity analysis of intubation rate by using alternative effect measure odds ratio and random effects model in randomized controlled trials.

S4 Fig [eps]
Sensitivity analysis of intubation rate by using alternative effect measure odds ratio and fixed effects model in randomized controlled trials.

S1 File [docx]
Details of search strategy and excluded studies.


Zdroje

1. Force ADT, Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, et al. Acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012; 307:2526–33. doi: 10.1001/jama.2012.5669 22797452

2. Sweeney RM, McAuley DF. Acute respiratory distress syndrome. Lancet. 2016; 388:2416–30. doi: 10.1016/S0140-6736(16)00578-X 27133972

3. Villar J, Blanco J, Kacmarek RM. Current incidence and outcome of the acute respiratory distress syndrome. Curr Opin Crit Care. 2016; 22:1–6. doi: 10.1097/MCC.0000000000000266 26645551

4. Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, et al. Epidemiology, Patterns of Care, and Mortality for Patients With Acute Respiratory Distress Syndrome in Intensive Care Units in 50 Countries. JAMA. 2016; 315:788–800. doi: 10.1001/jama.2016.0291 26903337

5. Thompson BT, Chambers RC, Liu KD. Acute Respiratory Distress Syndrome. N Engl J Med. 2017; 377:562–72. doi: 10.1056/NEJMra1608077 28792873

6. Fan E, Brodie D, Slutsky AS. Acute Respiratory Distress Syndrome: Advances in Diagnosis and Treatment. JAMA. 2018; 319:698–710. doi: 10.1001/jama.2017.21907 29466596

7. Doorduin J, Nollet JL, Roesthuis LH, van Hees HW, Brochard LJ, Sinderby CA, et al. Partial Neuromuscular Blockade during Partial Ventilatory Support in Sedated Patients with High Tidal Volumes. Am J Respir Crit Care Med. 2017; 195:1033–42. doi: 10.1164/rccm.201605-1016OC 27748627

8. Gainnier M, Roch A, Forel JM, Thirion X, Arnal JM, Donati S, et al. Effect of neuromuscular blocking agents on gas exchange in patients presenting with acute respiratory distress syndrome. Crit Care Med. 2004; 32:113–9. doi: 10.1097/01.CCM.0000104114.72614.BC 14707568

9. Alhazzani W, Alshahrani M, Jaeschke R, Forel JM, Papazian L, Sevransky J, et al. Neuromuscular blocking agents in acute respiratory distress syndrome: a systematic review and meta-analysis of randomized controlled trials. Crit Care. 2013; 17:R43. doi: 10.1186/cc12557 23497608

10. Forel JM, Roch A, Marin V, Michelet P, Demory D, Blache JL, et al. Neuromuscular blocking agents decrease inflammatory response in patients presenting with acute respiratory distress syndrome. Crit Care Med. 2006; 34:2749–57. doi: 10.1097/01.CCM.0000239435.87433.0D 16932229

11. Papazian L, Forel JM, Gacouin A, Penot-Ragon C, Perrin G, Loundou A, et al. Neuromuscular blockers in early acute respiratory distress syndrome. N Engl J Med. 2010; 363:1107–16. doi: 10.1056/NEJMoa1005372 20843245

12. Claesson J, Freundlich M, Gunnarsson I, Laake JH, Moller MH, Vandvik PO, et al. Scandinavian clinical practice guideline on fluid and drug therapy in adults with acute respiratory distress syndrome. Acta Anaesthesiol Scand. 2016; 60:697–709. doi: 10.1111/aas.12713 26988416

13. Hashimoto S, Sanui M, Egi M, Ohshimo S, Shiotsuka J, Seo R, et al. The clinical practice guideline for the management of ARDS in Japan. J Intensive Care. 2017; 5:50. doi: 10.1186/s40560-017-0222-3 28770093

14. Fan E, Del Sorbo L, Goligher EC, Hodgson CL, Munshi L, Walkey AJ, et al. An Official American Thoracic Society/European Society of Intensive Care Medicine/Society of Critical Care Medicine Clinical Practice Guideline: Mechanical Ventilation in Adult Patients with Acute Respiratory Distress Syndrome. Am J Respir Crit Care Med. 2017; 195:1253–63. doi: 10.1164/rccm.201703-0548ST 28459336

15. Barr J, Fraser GL, Puntillo K, Ely EW, Gelinas C, Dasta JF, et al. Clinical practice guidelines for the management of pain, agitation, and delirium in adult patients in the intensive care unit. Crit Care Med. 2013; 41:263–306. doi: 10.1097/CCM.0b013e3182783b72 23269131

16. Devlin JW, Skrobik Y, Gelinas C, Needham DM, Slooter AJC, Pandharipande PP, et al. Executive Summary: Clinical Practice Guidelines for the Prevention and Management of Pain, Agitation/Sedation, Delirium, Immobility, and Sleep Disruption in Adult Patients in the ICU. Crit Care Med. 2018; 46:1532–48. doi: 10.1097/CCM.0000000000003259 30113371

17. Rhodes A, Evans LE, Alhazzani W, Levy MM, Antonelli M, Ferrer R, et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Crit Care Med. 2017; 45:486–552. doi: 10.1097/CCM.0000000000002255 28098591

18. Neto AS, Pereira VG, Esposito DC, Damasceno MC, Schultz MJ. Neuromuscular blocking agents in patients with acute respiratory distress syndrome: a summary of the current evidence from three randomized controlled trials. Ann Intensive Care. 2012; 2:33. doi: 10.1186/2110-5820-2-33 22835162

19. Tao W, Yang LQ, Gao J, Shao J. Neuromuscular blocking agents for adult patients with acute respiratory distress syndrome: A meta-analysis of randomized controlled trials. J Trauma Acute Care Surg. 2018; 85:1102–9. doi: 10.1097/TA.0000000000002057 30462621

20. National Heart L, Blood Institute PCTN, Moss M, Huang DT, Brower RG, Ferguson ND, et al. Early Neuromuscular Blockade in the Acute Respiratory Distress Syndrome. N Engl J Med. 2019; 380:1997–2008. doi: 10.1056/NEJMoa1901686 31112383

21. Higgins JP, Green SA. Cochrane handbook for systematic reviews of interventions. Version 5.1.0 [updated March 2011]. Oxford (UK): The Cochrane Collaboration;. Available: www.handbookcochrane.org (accessed 2016 Dec 6).

22. Moher D, Liberati A, Tetzlaff J, Altman DG, Group P. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. BMJ. 2009; 339:b2535. doi: 10.1136/bmj.b2535 19622551

23. Higgins JP, Altman DG, Gotzsche PC, Juni P, Moher D, Oxman AD, et al. The Cochrane Collaboration's tool for assessing risk of bias in randomised trials. BMJ. 2011; 343:d5928. doi: 10.1136/bmj.d5928 22008217

24. 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. doi: 10.1186/1471-2288-5-13 15840177

25. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ. 2003; 327:557–60. doi: 10.1136/bmj.327.7414.557 12958120

26. Higgins JP, Thompson SG. Quantifying heterogeneity in a meta-analysis. Stat Med. 2002; 21:1539–58. doi: 10.1002/sim.1186 12111919

27. Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ. 2008; 336:924–6. doi: 10.1136/bmj.39489.470347.AD 18436948

28. Lyu G, Wang X, Jiang W, Cai T, Zhang Y. [Clinical study of early use of neuromuscular blocking agents in patients with severe sepsis and acute respiratory distress syndrome]. [Chinese]. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 2014; 26:325–9. doi: 10.3760/cma.j.issn.2095-4352.2014.05.008 24809261

29. Guervilly C, Bisbal M, Forel JM, Mechati M, Lehingue S, Bourenne J, et al. Effects of neuromuscular blockers on transpulmonary pressures in moderate to severe acute respiratory distress syndrome. Intensive Care Medicine. 2017; 43:408–18. doi: 10.1007/s00134-016-4653-4 28013329

30. Shehabi Y, Bellomo R, Reade MC, Bailey M, Bass F, Howe B, et al. Early intensive care sedation predicts long-term mortality in ventilated critically ill patients. Am J Respir Crit Care Med. 2012; 186:724–31. doi: 10.1164/rccm.201203-0522OC 22859526

31. Girard TD, Kress JP, Fuchs BD, Thomason JW, Schweickert WD, Pun BT, et al. Efficacy and safety of a paired sedation and ventilator weaning protocol for mechanically ventilated patients in intensive care (Awakening and Breathing Controlled trial): a randomised controlled trial. Lancet. 2008; 371:126–34. doi: 10.1016/S0140-6736(08)60105-1 18191684

32. Murray MJ, DeBlock H, Erstad B, Gray A, Jacobi J, Jordan C, et al. Clinical Practice Guidelines for Sustained Neuromuscular Blockade in the Adult Critically Ill Patient. Crit Care Med. 2016; 44:2079–103. doi: 10.1097/CCM.0000000000002027 27755068

33. Grawe ES, Bennett S, Hurford WE. Early Paralysis for the Management of ARDS. Respir Care. 2016; 61:830–8. doi: 10.4187/respcare.04734 27094392

34. Matthay MA, McAuley DF, Ware LB. Clinical trials in acute respiratory distress syndrome: challenges and opportunities. Lancet Respir Med. 2017; 5:524–34. doi: 10.1016/S2213-2600(17)30188-1 28664851

35. Price DR, Mikkelsen ME, Umscheid CA, Armstrong EJ. Neuromuscular Blocking Agents and Neuromuscular Dysfunction Acquired in Critical Illness: A Systematic Review and Meta-Analysis. Crit Care Med. 2016; 44:2070–8. doi: 10.1097/CCM.0000000000001839 27513545

36. Puthucheary Z, Rawal J, Ratnayake G, Harridge S, Montgomery H, Hart N. Neuromuscular blockade and skeletal muscle weakness in critically ill patients: time to rethink the evidence? Am J Respir Crit Care Med. 2012; 185:911–7. doi: 10.1164/rccm.201107-1320OE 22550208

37. Griffiths RD, Hall JB. Intensive care unit-acquired weakness. Crit Care Med. 2010; 38:779–87. doi: 10.1097/CCM.0b013e3181cc4b53 20048676

38. Sottile PD, Kiser TH, Burnham EL, Ho PM, Allen RR, Vandivier RW, et al. An Observational Study of the Efficacy of Cisatracurium Compared with Vecuronium in Patients with or at Risk for Acute Respiratory Distress Syndrome. American Journal Of Respiratory And Critical Care Medicine. 2018; 197:897–904. doi: 10.1164/rccm.201706-1132OC 29241014


Článek vyšel v časopise

PLOS One


2020 Číslo 1
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Svět praktické medicíny 1/2024 (znalostní test z časopisu)
nový kurz

Koncepce osteologické péče pro gynekology a praktické lékaře
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Význam metforminu pro „udržitelnou“ terapii diabetu
Autoři: prof. MUDr. Milan Kvapil, CSc., MBA

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#