Skip to main content
Erschienen in: BMC Medicine 1/2013

Open Access 01.12.2013 | Review

Pharmacological treatments in ARDS; a state-of-the-art update

verfasst von: Andrew James Boyle, Rob Mac Sweeney, Daniel Francis McAuley

Erschienen in: BMC Medicine | Ausgabe 1/2013

Abstract

Despite its high incidence and devastating outcomes, acute respiratory distress syndrome (ARDS) has no specific treatment, with effective therapy currently limited to minimizing potentially harmful ventilation and avoiding a positive fluid balance. Many pharmacological therapies have been investigated with limited success to date. In this review article we provide a state-of-the-art update on recent and ongoing trials, as well as reviewing promising future pharmacological therapies in ARDS.
Hinweise

Competing interests

DFM has performed paid consultancy work, and has been a member of advisory boards on acute lung injury for GlaxoSmithKline. In addition the institution for which DFM works has been paid for undertaking bronchoscopy as part of a clinical trial. DFM has also received fees for lecturing for Astrazenica; and has a patent submitted for a novel treatment for ARDS. AJB and RMS have no competing interests to declare.

Authors’ contributions

All authors contributed to the design of the paper. AJB wrote the initial manuscript, RMS and DFM edited it. All authors read and approved the final manuscript.
Abkürzungen
ACE
Angiotensin converting enzyme
ALI
Acute lung injury
ALTA
Albuterol treatment for acute lung injury
ARDS
Acute respiratory distress syndrome
ARENA
The effect of Aspirin on REducing iNflammation in human in vivo model of Acute lung injury
AT1R
Angiotensin 1 receptor
AT2R
Angiotensin 2 receptor
BALTI
β-agonist Lung Injury Trial
CI
Confidence interval
DEXA-ARDS
Efficacy study of dexamethasone to treat the acute respiratory distress syndrome
EVLW
Extravascular lung water
HARP
A randomized clinical trial of hydroxymethylglutaryl–coenzyme a reductase inhibition for acute lung injury
IFN-β
Interferon-β
IV
Intravenous
KGF
Keratinocyte growth factor
LIPS-A
Lung injury prevention study with aspirin
LPS
Lipopolysaccharide
NE
Neutrophil elastase
NMB
Neuromuscular blockade
P/F ratio
PaO2/FiO2 ratio
RAS
Renin-angiotensin system
RR
Relative risk
SAILS
Statins for acutely injured lungs from sepsis
STRIVE
Neutrophil elastase inhibition in acute lung injury
VEGF
Vascular endothelial growth factor
VFD
Ventilator free days.

Introduction

Despite its high incidence and devastating outcomes [1, 2], acute respiratory distress syndrome (ARDS) has no specific treatment, with effective therapy currently limited to minimizing potentially harmful ventilation and avoiding a positive fluid balance. ARDS is characterized by breakdown of the alveolar-capillary barrier, leading to flooding of the alveolar space producing the classical chest radiograph of bilateral pulmonary infiltrates. This non-cardiogenic pulmonary edema is associated with impaired oxygenation, as measured by the PaO2/FiO2 (P/F) ratio, with a lower P/F ratio indicating more severe hypoxia. Acute lung injury (ALI) is defined as a P/F ratio <300 mmHg (40kPa) and ARDS is a sub-group defined on the basis of more severely impaired oxygenation with a P/F ratio <200 mmHg (26.7 kPa).
Since it was first described in 1967 [3], and despite over 40 years of research, few pharmacological therapies have emerged for ARDS. We limited the search strategy for this state-of-the-art update review to recent randomized controlled trials and meta-analyses, as well as a review of promising potential future pharmacological therapies in ARDS in an adult setting.

Neuromuscular blockade

Lung protective ventilation can be achieved in the majority of patients without using neuromuscular blockade (NMB) [4]; however, initial small studies eliminating patient effort via skeletal muscle inhibition with NMB improved patient-ventilator synchrony, as evidenced by reduced airway pressures and improved chest wall compliance. Therefore, in the severely hypoxemic ARDS patient, NMB may permit lower-pressure, lower-tidal volume ventilation with a consequent reduction in ventilator-induced lung injury. These beneficial effects led to a multi-center, randomized, placebo-controlled trial to assess the effect of NMB upon mortality [5] (Table 1). This showed that infusion with cisatracuriumbesylate within 48 hours of mechanical ventilation in patients with moderate ARDS improved 90-day survival. However, no difference was noted between the intervention and placebo groups until Day 20. The biological mechanism by which NMB improves late but not early outcome is unclear. While promising, the protective effect of neuromuscular blockade needs to be confirmed in a further phase 3 trial.
Table 1
Characteristics of trials to date
Study title and abbreviation
Design (all placebo-controlled)
Population of ALI/ARDS A) Timing from ALI onset B) P/F ratio
Number recruited
Intervention
Primary outcome
Result (intervention vs control)
Mortality (intervention vs control)
Neuromuscular Blockade in Early ARDS [5]
Phase 2 RCT
A) 48 hours B) <150
340
Cisatracuriumbesylate: 15 mg initially, then 37.5 mg per hour for 48 hours
90-day survival
31.6% vs 40.7% (P= 0.08)
28-day: 23.7% vs 33.3% (P= 0.05)
The β-Agonist Lung Injury Trial (BALTI) [6]
Phase 1 RCT
A) 48 hours B) <300
40
Intravenous (IV) salbutamol for seven days (15 μg kg-1 h-1)
Extravascular lung water (EVLW) at Day 7
9.2 ± 6 vs 13.2 ±3 ml kg-1 (P= 0.04)
28-day: 58% vs 66% (P= 0.4)
Randomized, Placebo-Controlled Clinical Trial of an Aerosolized β2-Agonist for Treatment of Acute Lung Injury (ALTA) [7]
Phase 2 RCT
A) 48 hours B) < 300
282
Inhaled salbutamol (5 mg) every 4 hours for 10 days/24 hours after extubation
Ventilator-free days (VFD)
Stopped early 14.4 ± 0.9 vs 16.6 ± 0.9 (P= 0.087)
Death before discharge: 24.3 ± 3.5 vs 18.5 ± 3.4 (P= 0.261)
Effect of Intravenous β-2 Agonist Treatment on Clinical Outcomes in Acute Respiratory Distress Syndrome (BALTI-2) [8]
Phase 2 RCT
A) 72 hours B) <200
326
IV salbutamol for seven days (15 μg kg-1 (ideal body weight) h-1)
28-day mortality
Stopped early 34% vs 23% (P= 0.03)
 
Neutrophil Elastase Inhibition in Acute Lung Injury (STRIVE) [9]
Phase 3 RCT
A) 48 hours B) <300
492
Sivelestat infusion
1. 28-day mortality2. VFD
Stopped early 1 26.6% vs 26% (P= 0.847)2. 11.4 ±10.27 vs 11.9 ± 10.1 (P= 0.536)
 
Efficacy and Safety of Corticosteroids for Persistent ARDS (LaSRS) [14]
Phase 2 RCT
A) 7 to 28 days B) P/F <200
180
Moderate-dose IV methylprednisolone, for up to 25 days
60-day mortality
29.2% vs 28.6% (P= 1.0)
 
Methylprednisolone Infusion in Early Severe ARDS [15]
Phase 1 RCT
A) 72 hours B) <300
91
Low-dose IV methylprednisolone, for up to 28 days
Improvement in Lung Injury Score by Day 7
69.8% vs 35.7% (P= 0.002)
Hospital survival 76.2% vs 57.1% (P= 0.07)
A Randomized Clinical Trial of Hydroxymethylglutaryl–Coenzyme A Reductase Inhibition for Acute Lung Injury (HARP) [19]
Phase 2 RCT
A) 48 hours B) <300
60
Simvastatin 80 mg daily, up to 14 days
Reduction in EVLW indexed to actual body weight
13.7 vs 13.4 (P= 0.90) Improvements in secondary outcomes
Hospital survival: 19 vs 19 (P= 1.0)
Nebulized Heparin is Associated with Fewer Days of Mechanical Ventilation in Critically Ill Patients: a Randomized Controlled Trial [21]
Phase 2 RCT
Patients expected to require ventilation for >48 hours, and within 24 hours of ventilation
50
Heparin 25,000 units every 4 to 6 hours, for up to 14 days
Average daily P/F ratio
194.2 ± 62.8 vs 187 ± 38.6 mmHg (P= 0.6) Improvements in secondary outcomes
28-day: 20% vs 16% (P= 0.7)

β-adrenergic agonists

Alveolar edema is a central feature of ARDS, contributing to limitation of gaseous exchange and ventilatory failure. Experimental data suggest β-adrenergic agonists could accelerate alveolar fluid clearance, as well as provide cytoprotection, increased surfactant secretion and decreased endothelial permeability.
Based on the proposed effect upon alveolar fluid clearance, the β-Agonist Lung Injury Trial (BALTI) randomized 60 patients to IV salbutamol or placebo for seven days [6]. In this small, single center study, salbutamol therapy significantly reduced extravascular lung water at Day 7 compared with placebo (Table 1).
Subsequently, two large multi-center, randomized placebo-controlled trials were initiated. The first American study, ALTA (Albuterol Treatment for Acute Lung Injury) [7], enrolled 282 patients with ALI, but failed to demonstrate a difference in ventilator-free days between those receiving inhaled β-agonist therapy and those given placebo, and was stopped early as it breached the futility boundary (Table 1). In the most severely ill, as defined by the presence of shock at randomization, length of stay was significantly increased, suggesting worse outcome in this sub-group.
BALTI-2 was a concurrent UK multi-center study investigating intravenous salbutamol in patients with ARDS, but was terminated early due to excess mortality in the group receiving IV salbutamol [8] (Table 1).
On the basis of these larger trials, β-agonists should be avoided in patients with ALI. It is hypothesized that β-agonists may have a harmful cardiac effect, stimulating tachyarrhythmias and cardiac ischemia, resulting in a poorer outcome.

Neutrophil elastase inhibitors

Neutrophil elastase (NE) is a serine protease found in neutrophil granules and has a range of physiological effects, including anti-microbial actions and modification of tissue repair and inflammation. Excessive NE is capable of degrading endothelial basement membrane, and has been implicated in the pathogenesis of ARDS. Following a positive study in Japan, silvelestat, a neutrophil elastase inhibitor, was investigated in an international randomized, double-blind, placebo-controlled, multi-center phase III trial (STRIVE) [9] (Table 1). The study was stopped prematurely due to an increase in 180-day all-cause mortality.
A more recent meta-analysis of eight clinical trials (including STRIVE) investigating silvelestat has shown it to have no effect on short-term mortality, and a worse outcome for 180-day mortality (Risk Ratio (RR) 1.27, CI 1.00 to 1.62) [10].

Corticosteroids

Given their effective anti-inflammatory properties, there has been extensive interest in the potential role of corticosteroids in both the prevention and treatment of ARDS. Different regimens have been investigated, varying from short courses of high-dose steroids to prolonged courses of lower doses.
High dose corticosteroids do not prevent ARDS in at risk subjects [1113]. Therapeutically, both high-dose and moderate-dose steroids have so far failed to demonstrate efficacy in ARDS. An ARDSnet randomized, double-blind trial in 180 patients with ARDS for more than seven days, showed no effect of prolonged treatment with moderate-dose methylprednisolone compared to placebo [14] (Table 1). Although patients were liberated from mechanical ventilation earlier, patients receiving methylprednisolone were more likely to resume assisted ventilation, which was thought to be secondary to neuromuscular effects. In addition, initiation of treatment after 14 days of ARDS was associated with a harmful effect, with increased mortality at 60 and 180 days.
However, the role of low-dose corticosteroids in established ARDS remains uncertain, with one study of 91 patients demonstrating prolonged low-dose methylprednisolone therapy reduces severity of lung injury by Day 7 of treatment [15] (Table 1).
Despite a systematic review [16] and meta-analysis [17], the role of steroids in ARDS remains unclear, and in light of ongoing uncertainty, further trials are both planned (NCT01731795) and on-going (NCT01284452) (Table 2). It is also worth highlighting that the studies included used what are now considered injurious ventilation strategies. It remains uncertain if steroids provide benefit when combined with lung protective (and, therefore, less inflammatory) ventilator strategies.
Table 2
Ongoing/planned clinical trials in ARDS
Study reference number
Study title and abbreviation
Design
Population A) Timing B) P/F ratio
Anticipated enrollment
Intervention
Primary outcome
Status
NCT01731795
Efficacy Study of Dexamethasone to Treat the Acute Respiratory Distress Syndrome (DEXA-ARDS)
Phase 2/3 RCT
A) 24 hours from ARDS onset B) <200
314
Dexamethasone (20 mg/day for five days, then 10 mg/day for five days)
Ventilator-free days
Not yet recruiting
NCT01284452
Efficacy of hydrocortisone in treatment of severe sepsis/septic shock patients with ALI/ARDS
Phase 2/3 RCT
A) 12 hours from organ dysfunction B) <300
194
Hydrocortisone 50 mg every six hours for seven days
28-day all-cause mortality
Recruiting
ISRCTN88244364
Simvastatin in acute lung injury (HARP-2) [20]
Phase 2/3 RCT
A) 48 hours from ALI onset B) <300
540
Simvastatin 80 mg daily
Ventilator-free days
Recruiting
NCT00979121
Statins for Acutely Injured Lungs from Sepsis (SAILS)
Phase 3 RCT
A) 48 hours from ALI onset B) <300
1000
Rosuvastatin 20 mg daily
Hospital mortality Day 60
Recruiting
ACTRN12612000418875
Nebulized heparin for lung injury
Phase 2 RCT
A) Within 24 hours of mechanical ventilation in at-risk patients B) <300
256
Nebulised Heparin 25,000 international units, every six hours for up to 10 days
Physical function assessed using physical function component of SF-36 health survey
Not yet recruiting
NCT01659307
The effect of Aspirin on REducing iNflammation in human in vivo model of Acute lung injury (ARENA)
Phase 2 RCT
Healthy, non-smoking adults, using an LPS model of ALI
33
Aspirin 75 mg or Aspirin 1,200 mg
Bronchalveolar lavage intraleukin-8 concentration
Not yet recruiting
NCT01504867
LIPS-A: Lung Injury Prevention Study with Aspirin
Phase 2 RCT
Adults admitted to hospital via the emergency department at high-risk of developing ALI
400
Aspirin 325 mg Day 1, then 81 mg daily days 2 to 7
Development of ARDS
Recruiting
ISRCTN95690673
Keratinocyte growth factor in acute lung injury to reduce pulmonary dysfunction (KARE) [28]
Phase 2 RCT
A) 48 hours from ALI onset B) <300
60
Palifermin 60 μg/kg IV daily for up to six days
Oxygenation index at Day 7
Recruiting
ISRCTN27673620
VItamiN D Replacement to Prevent Acute Lung Injury following OesophagectOmy (VINDALOO) [34]
Phase 1/2 RCT
Adults undergoing planned transthoracic esophagectomy
80
Oral Vitamin D (100,000 IU)
EVLW at end of procedure
Recruiting
NCT00789685
Safety, tolerability and preliminary efficacy of FP-1201 in ALI and ARDS
Phase 1/2 Non-randomized
A) 48 hours from ALI onset B) <300
37
Interferon-β, increasing dose over six days
Clinically significant treatment emergent events, and all-cause mortality
Completed

Statins

HMG CoA-reductase inhibitors (statins) have a range of physiological effects beyond their role in cholesterol reduction, including anti-inflammatory actions and endothelial function modulation. Their effect on pulmonary inflammation was confirmed during a randomized, double-blind, placebo-controlled pre-clinical study, where simvastatin demonstrated a variety of anti-inflammatory effects during an inhaled lipopolysaccharide (LPS) model of ARDS in healthy volunteers [18].
A small phase II clinical trial in patients with ARDS (HARP) [19], suggested a potential role for simvastatin in the treatment of ARDS, with benefit in pulmonary and non-pulmonary organ dysfunction with no excess of adverse events in the intervention group (Table 1). Two larger trials are presently recruiting in the UK and Ireland (HARP-2 [20]) and in the USA (SAILS, NCT00979121), investigating simvastatin and rosuvastatin, respectively. A phase two trial in Oklahoma was recently terminated due to poor enrollment (NCT01195428).

Heparin

During the inflammatory process of ARDS fibrin is deposited throughout the alveolus, both intra- and extra-vascularly, impairing oxygenation. Experimental data show that, among other effects, heparin can reduce fibrin deposition. This led to a small study investigating the efficacy of nebulized heparin in patients at risk for ARDS [21]. Although there was no significant effect on the P/F ratio, this study suggested heparin may increase the number of ventilator-free days (VFD) (Table 1). The results of this trial have prompted further studies investigating the long-term impact of nebulized heparin in patients at risk of ARDS (ACTRN12612000418875) (Table 2).

Aspirin

During ARDS, platelets become activated and play an important role in disease progression by sequestering within the lung, forming micro-thrombi and attracting inflammatory cells to injured tissue. The potent anti-platelet effect of aspirin may offer a therapeutic approach to this pathological process. Observational data associated pre-hospital anti-platelet use with a reduction in subsequent ARDS incidence [22]. This finding was repeated in a separate study, although when propensity to receive aspirin was included in the analysis, the effect was lost [23].
Clinical trials are planned to investigate the effect of aspirin on reducing inflammation in a human model of ARDS (ARENA, NCT01659307), while others are ongoing to assess the impact of aspirin in the prevention of ARDS (LIPS-A, NCT01504867) (Table 2).

Angiotensin converting enzyme inhibitors/angiotensin receptor blockers

The renin-angiotensin system (RAS) plays an important role in the pathogenesis of ARDS, with angiotensin converting enzyme 1 (ACE1) directing a RAS signal to the angiotensin 1 receptor (AT1R), mediating alveolar vasoconstriction, permeability and fibrosis. A variant of ACE1, ACE2, diverts a RAS signal to AT2R, which promotes alveolar vasodilation, decreased permeability and apoptosis, thus opposing the alternative potentially injurious signaling mechanism [24]. Angiotensin receptor blockers attenuate ventilator-induced lung injury in animal models [25], and ACE-inhibitor or angiotensin receptor blocker therapy on discharge were associated with reduced mortality in acute respiratory failure patients [26]. Collectively, these data provide encouragement for future clinical trials in this area.

Stem cell therapy

Regenerative medicine is an emerging field, using stem cells or growth factors to aid the repair of damaged tissue and organs. Stem cells exhibit anti-inflammatory, immunomodulatory and reparative effects, largely mediated through secreted growth factors, although cell to cell contact between stem cells and alveoli also mediates important effects [27].
This prompts questions regarding the optimal delivery of stem cell therapy, as animal models of ARDS have shown survival to increase when treatment was delivered directly to the bronchial tree [28]. In addition, recent evidence in ex vivo human lung models of ALI support the investigation of delivering stem cells directly to the lung [29]. Clinical trials are awaited in this promising area.

Growth factors

Keratinocyte growth factor (KGF), an epithelial growth factor secreted by fibroblasts, has an important role in lung injury repair [30]. It increases alveolar cellular proliferation in ARDS, particularly of type II alveolar cells, enhancing repair. KGF may also have a role during the injurious process, reducing endothelial permeability and alveolar edema [30], and improving alveolar fluid clearance [31]. Following the completion of a small pre-clinical trial testing KGF in an LPS-model of ARDS (ISRCTN98813895), for which results are awaited, a phase II trial has commenced investigating the efficacy and safety of intravenous KGF (palifermin) in ARDS [32] (ISRCTN95690673) (Table 2).

Other potential therapies

Following data showing an immunomodulatory effect of vitamin D, animal models of ALI demonstrate that intra-tracheal administration of vitamin D can reduce neutrophil recruitment to the lung [33], which has obvious implications for future therapy in ALI. Vitamin D is currently being tested in patients at risk of developing ALI following esophagectomy [34] (Table 2).
Used in the management of multiple sclerosis, interferon-β (IFN-β) has been shown in vitro and in animal models of ALI to reduce vascular leakage and improve capillary endothelial barrier function [35]. IFN-β therapy has been studied in a phase I/II study in the UK, for which results are awaited (NCT00789685) (Table 2).
Finally, vascular endothelial growth factor (VEGF), an important molecule in the control of vascular permeability, has been found to be elevated in patients with ARDS [36]. The presence of VEGF inhibitors may prompt future randomized-controlled trials.
Other therapies, including the use of nitric oxide [37], prostacyclin [38] and surfactant [39], have been investigated and found to be ineffective. These additional therapies, plus others, are beyond the scope of this review, but have been covered in recent review articles [40, 41]

Conclusion

Despite many interventions being studied, to date there has been little success in developing effective pharmacological therapies for the management of ARDS. However, given the high associated morbidity and mortality, pressure remains to continue efforts to improve outcomes. Increasing numbers of pharmacological therapies are being investigated, and with encouraging pre-clinical and early clinical results, it is expected that over the coming years some will develop into useful agents for the prevention and treatment of ARDS.

Authors’ information

AJB is an Academic Foundation Year Two trainee. RMS is a specialist registrar in anesthesia and intensive care medicine. DFM is a professor and consultant in intensive care medicine. DFM has received funding from the Northern Ireland Public Health Agency Research and Development Division Translational Research Group for Critical Care. AJB and RMS are employed by the Belfast Health and Social Care Trust, while DFM has a joint appointment between Belfast Health and Social Care Trust and Queen’s University Belfast.

Acknowledgements

AJB, RMS and DFM received no funding for writing this article.
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

DFM has performed paid consultancy work, and has been a member of advisory boards on acute lung injury for GlaxoSmithKline. In addition the institution for which DFM works has been paid for undertaking bronchoscopy as part of a clinical trial. DFM has also received fees for lecturing for Astrazenica; and has a patent submitted for a novel treatment for ARDS. AJB and RMS have no competing interests to declare.

Authors’ contributions

All authors contributed to the design of the paper. AJB wrote the initial manuscript, RMS and DFM edited it. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Rubenfeld GD, Caldwell E, Peabody E, Weaver J, Martin DP, Neff M, Stern EJ, Hudson LD: Incidence and outcomes of acute lung injury. N Engl J Med. 2005, 353: 1685-1693. 10.1056/NEJMoa050333.CrossRefPubMed Rubenfeld GD, Caldwell E, Peabody E, Weaver J, Martin DP, Neff M, Stern EJ, Hudson LD: Incidence and outcomes of acute lung injury. N Engl J Med. 2005, 353: 1685-1693. 10.1056/NEJMoa050333.CrossRefPubMed
2.
Zurück zum Zitat Herridge MS, Cheung AM, Tansey CM, Matte-Martyn A, Diaz-Granados N, Al-Saidi F, Cooper AB, Guest CB, Mazer CD, Mehta S, Stewart TE, Barr A, Cook D, Slutsky AS: One-year outcomes in survivors of the acute respiratory distress syndrome. N Engl J Med. 2003, 348: 683-693. 10.1056/NEJMoa022450.CrossRefPubMed Herridge MS, Cheung AM, Tansey CM, Matte-Martyn A, Diaz-Granados N, Al-Saidi F, Cooper AB, Guest CB, Mazer CD, Mehta S, Stewart TE, Barr A, Cook D, Slutsky AS: One-year outcomes in survivors of the acute respiratory distress syndrome. N Engl J Med. 2003, 348: 683-693. 10.1056/NEJMoa022450.CrossRefPubMed
3.
Zurück zum Zitat Ashbaugh D, Boyd Bigelow D, Petty T, Levine B: Acute respiratory distress in adults. Lancet. 1967, 290: 319-323. 10.1016/S0140-6736(67)90168-7.CrossRef Ashbaugh D, Boyd Bigelow D, Petty T, Levine B: Acute respiratory distress in adults. Lancet. 1967, 290: 319-323. 10.1016/S0140-6736(67)90168-7.CrossRef
4.
Zurück zum Zitat The Acute Respiratory Distress Syndrome Network: Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med. 2000, 342: 1301-1308.CrossRef The Acute Respiratory Distress Syndrome Network: Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med. 2000, 342: 1301-1308.CrossRef
5.
Zurück zum Zitat Papazian L, Forel J-M, Gacouin A, Penot-Ragon C, Perrin G, Loundou A, Jaber S, Arnal J-M, Perez D, Seghboyan J-M, Constantin J-M, Courant P, Lefrant J-Y, Guérin C, Prat G, Morange S, Roch A: Neuromuscular blockers in early acute respiratory distress syndrome. N Engl J Med. 2010, 363: 1107-1116. 10.1056/NEJMoa1005372.CrossRefPubMed Papazian L, Forel J-M, Gacouin A, Penot-Ragon C, Perrin G, Loundou A, Jaber S, Arnal J-M, Perez D, Seghboyan J-M, Constantin J-M, Courant P, Lefrant J-Y, Guérin C, Prat G, Morange S, Roch A: Neuromuscular blockers in early acute respiratory distress syndrome. N Engl J Med. 2010, 363: 1107-1116. 10.1056/NEJMoa1005372.CrossRefPubMed
6.
Zurück zum Zitat Perkins GD, McAuley DF, Thickett DR, Gao F: The β-Agonist Lung Injury Trial (BALTI) a randomized placebo-controlled clinical trial. Am J Respir Crit Care Med. 2006, 173: 281-287. 10.1164/rccm.200508-1302OC.CrossRefPubMed Perkins GD, McAuley DF, Thickett DR, Gao F: The β-Agonist Lung Injury Trial (BALTI) a randomized placebo-controlled clinical trial. Am J Respir Crit Care Med. 2006, 173: 281-287. 10.1164/rccm.200508-1302OC.CrossRefPubMed
7.
Zurück zum Zitat The National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network: Randomized, placebo-controlled clinical trial of an aerosolized β2-agonist for treatment of acute lung injury. Am J Respir Crit Care Med. 2011, 184: 561-568.CrossRef The National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network: Randomized, placebo-controlled clinical trial of an aerosolized β2-agonist for treatment of acute lung injury. Am J Respir Crit Care Med. 2011, 184: 561-568.CrossRef
8.
Zurück zum Zitat Smith FG, Perkins GD, Gates S, Young D, McAuley DF, Tunnicliffe W, Khan Z, Lamb SE: Effect of intravenous β-2 agonist treatment on clinical outcomes in acute respiratory distress syndrome (BALTI-2): a multicentre, randomised controlled trial. Lancet. 2012, 379: 229-235. 10.1016/S0140-6736(11)61623-1.CrossRefPubMedCentral Smith FG, Perkins GD, Gates S, Young D, McAuley DF, Tunnicliffe W, Khan Z, Lamb SE: Effect of intravenous β-2 agonist treatment on clinical outcomes in acute respiratory distress syndrome (BALTI-2): a multicentre, randomised controlled trial. Lancet. 2012, 379: 229-235. 10.1016/S0140-6736(11)61623-1.CrossRefPubMedCentral
9.
Zurück zum Zitat Zeiher BG, Artigas A, Vincent J-L, Dmitrienko A, Jackson K, Thompson BT, Bernard G: Neutrophil elastase inhibition in acute lung injury: results of the STRIVE study. Crit Care Med. 2004, 32: 1695-1702. 10.1097/01.CCM.0000133332.48386.85.CrossRefPubMed Zeiher BG, Artigas A, Vincent J-L, Dmitrienko A, Jackson K, Thompson BT, Bernard G: Neutrophil elastase inhibition in acute lung injury: results of the STRIVE study. Crit Care Med. 2004, 32: 1695-1702. 10.1097/01.CCM.0000133332.48386.85.CrossRefPubMed
10.
Zurück zum Zitat Iwata K, Doi A, Ohji G, Oka H, Oba Y, Takimoto K, Igarashi W, Gremillion DH, Shimada T: Effect of neutrophil elastase inhibitor (sivelestat sodium) in the treatment of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS): a systematic review and meta-analysis. Intern Med. 2010, 49: 2423-2432. 10.2169/internalmedicine.49.4010.CrossRefPubMed Iwata K, Doi A, Ohji G, Oka H, Oba Y, Takimoto K, Igarashi W, Gremillion DH, Shimada T: Effect of neutrophil elastase inhibitor (sivelestat sodium) in the treatment of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS): a systematic review and meta-analysis. Intern Med. 2010, 49: 2423-2432. 10.2169/internalmedicine.49.4010.CrossRefPubMed
11.
Zurück zum Zitat Weigelt JA, Norcross JF, Borman KR, Snyder WH: Early steroid therapy for respiratory failure. Arch Surg. 1985, 120: 536-540. 10.1001/archsurg.1985.01390290018003.CrossRefPubMed Weigelt JA, Norcross JF, Borman KR, Snyder WH: Early steroid therapy for respiratory failure. Arch Surg. 1985, 120: 536-540. 10.1001/archsurg.1985.01390290018003.CrossRefPubMed
12.
Zurück zum Zitat Sprung CL, Caralis PV, Marcial EH, Pierce M, Gelbard MA, Long WM, Duncan RC, Tendler MD, Karpf M: The effects of high-dose corticosteroids in patients with septic shock. N Engl J Med. 1984, 311: 1137-1143. 10.1056/NEJM198411013111801.CrossRefPubMed Sprung CL, Caralis PV, Marcial EH, Pierce M, Gelbard MA, Long WM, Duncan RC, Tendler MD, Karpf M: The effects of high-dose corticosteroids in patients with septic shock. N Engl J Med. 1984, 311: 1137-1143. 10.1056/NEJM198411013111801.CrossRefPubMed
13.
Zurück zum Zitat Luce JM, Montgomery AB, Marks JD, Turner J, Metz CA, Murray JF: Ineffectiveness of high-dose methylprednisolone in preventing parenchymal lung injury and improving mortality in patients with septic shock. Am Rev Respir Dis. 1988, 138: 62-68. 10.1164/ajrccm/138.1.62.CrossRefPubMed Luce JM, Montgomery AB, Marks JD, Turner J, Metz CA, Murray JF: Ineffectiveness of high-dose methylprednisolone in preventing parenchymal lung injury and improving mortality in patients with septic shock. Am Rev Respir Dis. 1988, 138: 62-68. 10.1164/ajrccm/138.1.62.CrossRefPubMed
14.
Zurück zum Zitat The National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network: Efficacy and safety of corticosteroids for persistent acute respiratory distress syndrome. N Engl J Med. 2006, 354: 1671-1684.CrossRef The National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network: Efficacy and safety of corticosteroids for persistent acute respiratory distress syndrome. N Engl J Med. 2006, 354: 1671-1684.CrossRef
15.
Zurück zum Zitat Meduri GU, Golden E, Freire AX, Taylor E, Zaman M, Carson SJ, Gibson M, Umberger R: Methylprednisolone infusion in early severe ARDS: results of a randomized controlled trial. Chest. 2007, 131: 954-963. 10.1378/chest.06-2100.CrossRefPubMed Meduri GU, Golden E, Freire AX, Taylor E, Zaman M, Carson SJ, Gibson M, Umberger R: Methylprednisolone infusion in early severe ARDS: results of a randomized controlled trial. Chest. 2007, 131: 954-963. 10.1378/chest.06-2100.CrossRefPubMed
16.
Zurück zum Zitat Deal EN, Hollands JM, Schramm GE, Micek ST: Role of corticosteroids in the management of acute respiratory distress syndrome. Clin Ther. 2008, 30: 787-799. 10.1016/j.clinthera.2008.05.012.CrossRefPubMed Deal EN, Hollands JM, Schramm GE, Micek ST: Role of corticosteroids in the management of acute respiratory distress syndrome. Clin Ther. 2008, 30: 787-799. 10.1016/j.clinthera.2008.05.012.CrossRefPubMed
17.
Zurück zum Zitat Peter JV, John P, Graham PL, Moran JL, George IA, Bersten A: Corticosteroids in the prevention and treatment of acute respiratory distress syndrome (ARDS) in adults: meta-analysis. BMJ. 2008, 336: 1006-1009. 10.1136/bmj.39537.939039.BE.CrossRefPubMedPubMedCentral Peter JV, John P, Graham PL, Moran JL, George IA, Bersten A: Corticosteroids in the prevention and treatment of acute respiratory distress syndrome (ARDS) in adults: meta-analysis. BMJ. 2008, 336: 1006-1009. 10.1136/bmj.39537.939039.BE.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Shyamsundar M, McKeown STW, O’Kane CM, Craig TR, Brown V, Thickett DR, Matthay MA, Taggart CC, Backman JT, Elborn JS, McAuley DF: Simvastatin decreases lipopolysaccharide-induced pulmonary inflammation in healthy volunteers. Am J Respir Crit Care Med. 2009, 179: 1107-1114. 10.1164/rccm.200810-1584OC.CrossRefPubMedPubMedCentral Shyamsundar M, McKeown STW, O’Kane CM, Craig TR, Brown V, Thickett DR, Matthay MA, Taggart CC, Backman JT, Elborn JS, McAuley DF: Simvastatin decreases lipopolysaccharide-induced pulmonary inflammation in healthy volunteers. Am J Respir Crit Care Med. 2009, 179: 1107-1114. 10.1164/rccm.200810-1584OC.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Craig TR, Duffy MJ, Shyamsundar M, McDowell C, O’Kane CM, Elborn JS, McAuley DF: A randomized clinical trial of hydroxymethylglutaryl– coenzyme a reductase inhibition for acute lung injury (the HARP study). Am J Respir Crit Care Med. 2011, 183: 620-626. 10.1164/rccm.201003-0423OC.CrossRefPubMed Craig TR, Duffy MJ, Shyamsundar M, McDowell C, O’Kane CM, Elborn JS, McAuley DF: A randomized clinical trial of hydroxymethylglutaryl– coenzyme a reductase inhibition for acute lung injury (the HARP study). Am J Respir Crit Care Med. 2011, 183: 620-626. 10.1164/rccm.201003-0423OC.CrossRefPubMed
20.
Zurück zum Zitat McAuley DF, Laffey JG, O’Kane CM, Cross M, Perkins GD, Murphy L, McNally C, Crealey G, Stevenson M, the HARP-2 investigators on behalf of the Irish Critical Care Trials Group: Hydroxymethylglutaryl-CoA reductase inhibition with simvastatin in Acute lung injury to Reduce Pulmonary dysfunction (HARP-2) trial: study protocol for a randomized controlled trial. Trials. 2012, 13: 170-10.1186/1745-6215-13-170.CrossRefPubMedPubMedCentral McAuley DF, Laffey JG, O’Kane CM, Cross M, Perkins GD, Murphy L, McNally C, Crealey G, Stevenson M, the HARP-2 investigators on behalf of the Irish Critical Care Trials Group: Hydroxymethylglutaryl-CoA reductase inhibition with simvastatin in Acute lung injury to Reduce Pulmonary dysfunction (HARP-2) trial: study protocol for a randomized controlled trial. Trials. 2012, 13: 170-10.1186/1745-6215-13-170.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Dixon B, Schultz MJ, Smith R, Fink JB, Santamaria JD, Campbell DJ: Nebulized heparin is associated with fewer days of mechanical ventilation in critically ill patients: a randomized controlled trial. Crit Care. 2010, 14: R180-10.1186/cc9286.CrossRefPubMedPubMedCentral Dixon B, Schultz MJ, Smith R, Fink JB, Santamaria JD, Campbell DJ: Nebulized heparin is associated with fewer days of mechanical ventilation in critically ill patients: a randomized controlled trial. Crit Care. 2010, 14: R180-10.1186/cc9286.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Erlich JM, Talmor DS, Cartin-Ceba R, Gajic O, Kor DJ: Prehospitalization antiplatelet therapy is associated with a reduced incidence of acute lung injury: a population-based cohort study. Chest. 2011, 139: 289-295. 10.1378/chest.10-0891.CrossRefPubMed Erlich JM, Talmor DS, Cartin-Ceba R, Gajic O, Kor DJ: Prehospitalization antiplatelet therapy is associated with a reduced incidence of acute lung injury: a population-based cohort study. Chest. 2011, 139: 289-295. 10.1378/chest.10-0891.CrossRefPubMed
23.
Zurück zum Zitat Kor DJ, Erlich J, Gong MN, Malinchoc M, Carter RE, Gajic O, Talmor DS: Association of prehospitalization aspirin therapy and acute lung injury: results of a multicenter international observational study of at-risk patients. Crit Care Med. 2011, 39: 2393-2400. 10.1097/CCM.0b013e318225757f.CrossRefPubMedPubMedCentral Kor DJ, Erlich J, Gong MN, Malinchoc M, Carter RE, Gajic O, Talmor DS: Association of prehospitalization aspirin therapy and acute lung injury: results of a multicenter international observational study of at-risk patients. Crit Care Med. 2011, 39: 2393-2400. 10.1097/CCM.0b013e318225757f.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, Yang P, Sarao R, Wada T, Leong-Poi H, Crackower MA, Fukamizu A, Hui C-C, Hein L, Uhlig S, Slutsky AS, Jiang C, Penninger JM: Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 2005, 436: 112-116. 10.1038/nature03712.CrossRefPubMed Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, Yang P, Sarao R, Wada T, Leong-Poi H, Crackower MA, Fukamizu A, Hui C-C, Hein L, Uhlig S, Slutsky AS, Jiang C, Penninger JM: Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 2005, 436: 112-116. 10.1038/nature03712.CrossRefPubMed
25.
Zurück zum Zitat Yao S, Feng D, Wu Q, Li K, Wang L: Losartan attenuates ventilator-induced lung injury. J Surg Res. 2008, 145: 25-32. 10.1016/j.jss.2007.03.075.CrossRefPubMed Yao S, Feng D, Wu Q, Li K, Wang L: Losartan attenuates ventilator-induced lung injury. J Surg Res. 2008, 145: 25-32. 10.1016/j.jss.2007.03.075.CrossRefPubMed
26.
Zurück zum Zitat Noveanu M, Breidthardt T, Reichlin T, Gayat E, Potocki M, Pargger H, Heise A, Meissner J, Twerenbold R, Muravitskaya N, Mebazaa A, Mueller C: Effect of oral β-blocker on short and long-term mortality in patients with acute respiratory failure: results from the BASEL-II-ICU study. Crit Care. 2010, 14: R198-10.1186/cc9317.CrossRefPubMedPubMedCentral Noveanu M, Breidthardt T, Reichlin T, Gayat E, Potocki M, Pargger H, Heise A, Meissner J, Twerenbold R, Muravitskaya N, Mebazaa A, Mueller C: Effect of oral β-blocker on short and long-term mortality in patients with acute respiratory failure: results from the BASEL-II-ICU study. Crit Care. 2010, 14: R198-10.1186/cc9317.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K, Rowlands DJ, Quadri SK, Bhattacharya S, Bhattacharya J: Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012, 18: 759-765. 10.1038/nm.2736.CrossRefPubMedPubMedCentral Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K, Rowlands DJ, Quadri SK, Bhattacharya S, Bhattacharya J: Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat Med. 2012, 18: 759-765. 10.1038/nm.2736.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Gupta N, Su X, Popov B, Lee JW, Serikov V, Matthay MA: Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J Immunol. 2007, 179: 1855-1863.CrossRefPubMed Gupta N, Su X, Popov B, Lee JW, Serikov V, Matthay MA: Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J Immunol. 2007, 179: 1855-1863.CrossRefPubMed
29.
Zurück zum Zitat Lee JW, Krasnodembskaya A, McKenna DH, Song Y, Abbott J, Matthay MA: Therapeutic effects of human mesenchymal stem cells in ex vivo human lungs injured with live bacteria. Am J Respir Crit Care Med. 2013, 187: 751-760. 10.1164/rccm.201206-0990OC.CrossRefPubMedPubMedCentral Lee JW, Krasnodembskaya A, McKenna DH, Song Y, Abbott J, Matthay MA: Therapeutic effects of human mesenchymal stem cells in ex vivo human lungs injured with live bacteria. Am J Respir Crit Care Med. 2013, 187: 751-760. 10.1164/rccm.201206-0990OC.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Ware LB, Matthay MA: Keratinocyte and hepatocyte growth factors in the lung: roles in lung development, inflammation, and repair. Am J Physiol Lung Cell Mol Physiol. 2002, 282: L924-L940.CrossRefPubMed Ware LB, Matthay MA: Keratinocyte and hepatocyte growth factors in the lung: roles in lung development, inflammation, and repair. Am J Physiol Lung Cell Mol Physiol. 2002, 282: L924-L940.CrossRefPubMed
31.
Zurück zum Zitat Matthay MA, Goolaerts A, Howard JP, Lee JW: Mesenchymal stem cells for acute lung injury: preclinical evidence. Crit Care Med. 2010, 38: S569-S573.CrossRefPubMedPubMedCentral Matthay MA, Goolaerts A, Howard JP, Lee JW: Mesenchymal stem cells for acute lung injury: preclinical evidence. Crit Care Med. 2010, 38: S569-S573.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Cross LJ, O’Kane CM, McDowell C, Elborn JJ, Matthay MA, McAuley DF: Keratinocyte growth factor in acute lung injury to reduce pulmonary dysfunction – a randomised placebo-controlled trial (KARE): study protocol. Trials. 2013, 14: 51-10.1186/1745-6215-14-51.CrossRefPubMedPubMedCentral Cross LJ, O’Kane CM, McDowell C, Elborn JJ, Matthay MA, McAuley DF: Keratinocyte growth factor in acute lung injury to reduce pulmonary dysfunction – a randomised placebo-controlled trial (KARE): study protocol. Trials. 2013, 14: 51-10.1186/1745-6215-14-51.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Takano Y, Mitsuhashi H, Ueno K: 1α,25-dihydroxyvitamin D3 inhibits neutrophil recruitment in hamster model of acute lung injury. Steroids. 2011, 76: 1305-1309. 10.1016/j.steroids.2011.06.009.CrossRefPubMed Takano Y, Mitsuhashi H, Ueno K: 1α,25-dihydroxyvitamin D3 inhibits neutrophil recruitment in hamster model of acute lung injury. Steroids. 2011, 76: 1305-1309. 10.1016/j.steroids.2011.06.009.CrossRefPubMed
34.
Zurück zum Zitat Parekh D, Dancer RC, Lax S, Cooper MS, Martineau AR, Fraser WD, Tucker O, Alderson D, Perkins GD, Gao-Smith F, Thickett DR: Vitamin D to prevent acute lung injury following oesophagectomy (VINDALOO): study protocol for a randomised placebo controlled trial. Trials. 2013, 14: 100-10.1186/1745-6215-14-100.CrossRefPubMedPubMedCentral Parekh D, Dancer RC, Lax S, Cooper MS, Martineau AR, Fraser WD, Tucker O, Alderson D, Perkins GD, Gao-Smith F, Thickett DR: Vitamin D to prevent acute lung injury following oesophagectomy (VINDALOO): study protocol for a randomised placebo controlled trial. Trials. 2013, 14: 100-10.1186/1745-6215-14-100.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Kiss J, Yegutkin GG, Koskinen K, Savunen T, Jalkanen S, Salmi M: IFN-β protects from vascular leakage via up-regulation of CD73. Eur J Immunol. 2007, 37: 3334-3338. 10.1002/eji.200737793.CrossRefPubMed Kiss J, Yegutkin GG, Koskinen K, Savunen T, Jalkanen S, Salmi M: IFN-β protects from vascular leakage via up-regulation of CD73. Eur J Immunol. 2007, 37: 3334-3338. 10.1002/eji.200737793.CrossRefPubMed
36.
Zurück zum Zitat Ware LB, Kaner RJ, Crystal RG, Schane R, Trivedi NN, McAuley D, Matthay MA: VEGF levels in the alveolar compartment do not distinguish between ARDS and hydrostatic pulmonary oedema. Eur Respir J. 2005, 26: 101-105. 10.1183/09031936.05.00106604.CrossRefPubMedPubMedCentral Ware LB, Kaner RJ, Crystal RG, Schane R, Trivedi NN, McAuley D, Matthay MA: VEGF levels in the alveolar compartment do not distinguish between ARDS and hydrostatic pulmonary oedema. Eur Respir J. 2005, 26: 101-105. 10.1183/09031936.05.00106604.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Adhikari NKJ, Burns KEA, Friedrich JO, Granton JT, Cook DJ, Meade MO: Effect of nitric oxide on oxygenation and mortality in acute lung injury: systematic review and meta-analysis. BMJ. 2007, 334: 779-779. 10.1136/bmj.39139.716794.55.CrossRefPubMedPubMedCentral Adhikari NKJ, Burns KEA, Friedrich JO, Granton JT, Cook DJ, Meade MO: Effect of nitric oxide on oxygenation and mortality in acute lung injury: systematic review and meta-analysis. BMJ. 2007, 334: 779-779. 10.1136/bmj.39139.716794.55.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Afshari A, Brok J, Møller AM, Wetterslev J: Aerosolized prostacyclin for acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Cochrane Database Syst Rev. 2010, CD007733. Afshari A, Brok J, Møller AM, Wetterslev J: Aerosolized prostacyclin for acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Cochrane Database Syst Rev. 2010, CD007733.
39.
Zurück zum Zitat Spragg RG, Taut FJH, Lewis JF, Schenk P, Ruppert C, Dean N, Krell K, Karabinis A, Günther A: Recombinant Surfactant Protein C–based Surfactant for Patients with Severe Direct Lung Injury. Am J Respir Crit Care Med. 2011, 183: 1055-1061. 10.1164/rccm.201009-1424OC.CrossRefPubMed Spragg RG, Taut FJH, Lewis JF, Schenk P, Ruppert C, Dean N, Krell K, Karabinis A, Günther A: Recombinant Surfactant Protein C–based Surfactant for Patients with Severe Direct Lung Injury. Am J Respir Crit Care Med. 2011, 183: 1055-1061. 10.1164/rccm.201009-1424OC.CrossRefPubMed
40.
Zurück zum Zitat Mac Sweeney R, McAuley DF: Pharmacological therapy for acute lung injury. Open Crit Care Med J. 2010, 3: 7-19. Mac Sweeney R, McAuley DF: Pharmacological therapy for acute lung injury. Open Crit Care Med J. 2010, 3: 7-19.
41.
Zurück zum Zitat Raghavendran K, Pryhuber GS, Chess PR, Davidson BA, Knight PR, Notter RH: Pharmacotherapy of acute lung injury and acute respiratory distress syndrome. Curr Med Chem. 2008, 15: 1911-1924. 10.2174/092986708785132942.CrossRefPubMedPubMedCentral Raghavendran K, Pryhuber GS, Chess PR, Davidson BA, Knight PR, Notter RH: Pharmacotherapy of acute lung injury and acute respiratory distress syndrome. Curr Med Chem. 2008, 15: 1911-1924. 10.2174/092986708785132942.CrossRefPubMedPubMedCentral
Metadaten
Titel
Pharmacological treatments in ARDS; a state-of-the-art update
verfasst von
Andrew James Boyle
Rob Mac Sweeney
Daniel Francis McAuley
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
BMC Medicine / Ausgabe 1/2013
Elektronische ISSN: 1741-7015
DOI
https://doi.org/10.1186/1741-7015-11-166

Weitere Artikel der Ausgabe 1/2013

BMC Medicine 1/2013 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 24 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Update Allgemeinmedizin

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