Acute respiratory distress syndrome

Michael A Matthay, Rachel L Zemans, Guy A Zimmerman, Yaseen M Arabi, Jeremy R Beitler, Alain Mercat, Margaret Herridge, Adrienne G Randolph, Carolyn S Calfee, Michael A Matthay, Rachel L Zemans, Guy A Zimmerman, Yaseen M Arabi, Jeremy R Beitler, Alain Mercat, Margaret Herridge, Adrienne G Randolph, Carolyn S Calfee

Abstract

The acute respiratory distress syndrome (ARDS) is a common cause of respiratory failure in critically ill patients and is defined by the acute onset of noncardiogenic pulmonary oedema, hypoxaemia and the need for mechanical ventilation. ARDS occurs most often in the setting of pneumonia, sepsis, aspiration of gastric contents or severe trauma and is present in ~10% of all patients in intensive care units worldwide. Despite some improvements, mortality remains high at 30-40% in most studies. Pathological specimens from patients with ARDS frequently reveal diffuse alveolar damage, and laboratory studies have demonstrated both alveolar epithelial and lung endothelial injury, resulting in accumulation of protein-rich inflammatory oedematous fluid in the alveolar space. Diagnosis is based on consensus syndromic criteria, with modifications for under-resourced settings and in paediatric patients. Treatment focuses on lung-protective ventilation; no specific pharmacotherapies have been identified. Long-term outcomes of patients with ARDS are increasingly recognized as important research targets, as many patients survive ARDS only to have ongoing functional and/or psychological sequelae. Future directions include efforts to facilitate earlier recognition of ARDS, identifying responsive subsets of patients and ongoing efforts to understand fundamental mechanisms of lung injury to design specific treatments.

Conflict of interest statement

M.A.M. declares grant support from Bayer (current), GlaxoSmithKline (prior) and Amgen (prior); has served as Data Safety and Monitoring Board chair for Roche-Genentech and has served as a consultant for GlaxoSmithKline, Bayer, Boehringer, CSL Berhring, Navigen, Quark and Cerus. G.A.Z. has served as a consultant for Navigen. Y.M.A. has served as a consultant for Gilead Sciences (past), Regeneron (past) and SAB Therapeutics (current). A.M. received fees for serving on a steering committee for Faron Pharmaceuticals, consulting fees from Air Liquide Medical Systems, grant support for research and lecture fees from Fisher & Paykel and Covidien, and lecture fees from Drager, Pfizer and ResMed. A.G.R. declares grant support from Roche-Genentech (current) and has served as a consultant for La Jolla Pharma and Bristol Meyer Squibb. C.S.C. declares grant support from Bayer (current) and GlaxoSmithKline (prior) and has served as a consultant for GlaxoSmithKline, Bayer, Boehringer, Prometic, Roche-Genentech, CSL Behring and Quark. The remaining authors declare no competing interests.

Figures

Fig. 1. The normal alveolus.
Fig. 1. The normal alveolus.
The alveolar epithelium is a continuous monolayer of alveolar type I (ATI) cells (very thin cells that permit gas exchange) and alveolar type II (ATII) cells (which produce surfactant to enable lung expansion with a low surface tension); both cells transport ions and fluid from the alveolus to maintain dry airspaces. The intact alveolar epithelium is linked by intercellular tight junctions. Tight junctions are responsible for barrier function and regulating the movement of fluid and ions across the epithelium and are composed of transmembrane claudins and occludins and cytoplasmic zonula occludens (ZO) proteins that anchor tight junctions to the actin cytoskeleton. Alveolar epithelial cells express plasma membrane E-cadherin and β-catenin. β-Catenin also functions as a transcriptional cofactor and has a role in cell turnover in the subset of ATII cells that function as stem cells during homeostasis. Endothelial cells serve to regulate the influx of fluid and inflammatory cells into the interstitial space and are connected by intercellular junctions comprising tight junctions and adherens junctions. Adherens junctions contain vascular endothelial cadherin (VE-cadherin), which mediates cell–cell contact through its extracellular domain and has a key role in barrier function. p120–catentin binds to and stabilizes VE-cadherin, which is linked to the actin cytoskeleton via α-catenin (α in the figure) and has multiple additional functional interactions. TIE2 acts in concert with vascular endothelial-protein tyrosine phosphatase (VE-PTP), which dephosphorylates VE-cadherin, stabilizing it. Normally, transvascular flux of fluid out of the capillary moves water and low-molecular-weight solutes into the interstitial space and then into the lymphatics; in health, this fluid does not cross the epithelial barrier. Resident alveolar macrophages populate the airspaces, providing host defence. Large numbers of polymorphonuclear leukocytes (PMNs) reside in the alveolar capillaries and can be rapidly mobilized to the airspaces in the setting of infection. β, β-catenin; BASC, bronchioalveolar stem cell; ENaC, epithelial sodium channel; JAM, junctional adhesion molecule; RBC, red blood cell.
Fig. 2. Microscopic findings in lung tissue…
Fig. 2. Microscopic findings in lung tissue in patients with ARDS.
In acute respiratory distress syndrome (ARDS), features of diffuse alveolar damage (DAD), such as in the acute ‘exudative’ phase (~7 days) (panel a), are typically followed by alveolar type II (ATII) cell hyperplasia and interstitial fibrosis in a ‘proliferative’ phase. Eosinophilic depositions termed hyaline membranes are defining features of DAD (pink structure lining the central alveolus, indicated by the arrowhead in panel b) are defining features of DAD. Leukocytes are embedded in the hyaline membranes (arrows in panel b). Electron microscopic analyses (panel c) demonstrate that alterations in endothelial and epithelial cells are critical features of acute alveolar injury in ARDS,. Focal epithelial destruction of alveolar type I (ATI) cells and denudation of the alveolar basement membrane occur early in ARDS, whereas endothelial continuity is preserved with modest alterations in most cases. The pattern shown in panel c was identified in the lungs of a patient with indirect acute lung injury resulting from sepsis,. A, alveolar space; BM, basement membrane; C, capillary; EC, erythrocyte; EN, endothelial cell; HM, hyaline membrane; LC, leukocyte. Reprinted with permission of the American Thoracic Society. Copyright © 2019 American Thoracic Society. Matthay, M. A. & Zimmerman, G. A. (2005) Acute lung injury and the acute respiratory distress syndrome: four decades of inquiry into pathogenesis and rational management. Am. J. Respir. Cell Mol. Biol. 33, 319–327. The American Journal of Respiratory and Critical Care Medicine is an official journal of the American Thoracic Society.
Fig. 3. The injured alveolus.
Fig. 3. The injured alveolus.
A variety of insults (such as acid, viruses, ventilator-associated lung injury, hyperoxia or bacteria) can injure the epithelium, either directly or by inducing inflammation, which in turn injures the epithelium. Direct injury is inevitably exacerbated by a secondary wave of inflammatory injury. Activation of Toll-like receptors (not shown) on alveolar type II (ATII) cells and resident macrophages induces the secretion of chemokines, which recruit circulating immune cells into the airspaces. As neutrophils migrate across the epithelium, they release toxic mediators, including proteases, reactive oxygen species (ROS) and neutrophil extracellular traps (NETs), which have an important role in host defence but cause endothelial and epithelial injury. Monocytes also migrate into the lung and can cause injury, including epithelial cell apoptosis via IFNβ-dependent release of tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), which activates death receptors. Activated platelets form aggregates with polymorphonuclear (PMN) leukocytes, which are involved in NET formation, and monocyte–platelet aggregates. Red blood cells (RBCs) release cell-free haemoglobin, which exacerbates injury via oxidant-dependent mechanisms. Angiopoietin 2 inhibits TIE2-stabilization of vascular endothelial cadherin (VE-cadherin); vascular endothelial growth factor and other permeability-promoting agonists also destabilize VE-cadherin via dissociation from p120-catenin, resulting in its internalization and enhanced paracellular permeability. Additionally, loss of cell–cell adhesion in the setting of actomyosin contraction results in the formation of occasional gaps between endothelial cells. Epithelial injury also includes wounding of the plasma membrane, which can be induced by bacterial pore-forming toxins or mechanical stretch, and mitochondrial dysfunction. Together, these effects result in endothelial and epithelial permeability, which further facilitate the transmigration of leukocytes and lead to the influx of oedematous fluid and RBCs. Airspace filling with oedematous fluid causes hypoxaemia, resulting in the need for mechanical ventilation. The vascular injury and alveolar oedema contribute to the decreased ability to excrete CO2 (hypercapnia), accounting for the elevated pulmonary dead space in acute respiratory distress syndrome. In turn, hypoxaemia and hypercapnia impair vectorial sodium transport, reducing alveolar oedema clearance. ATI, alveolar type I cell; BASC, bronchioalveolar stem cell; ENaC, epithelial sodium channel.
Fig. 4. Epithelial cell regeneration in ARDS.
Fig. 4. Epithelial cell regeneration in ARDS.
Mice in which the alveolar type II (ATII) epithelial cells and all their progeny express green fluorescent protein (GFP) (SftpcCreERT2;mTmG mice) were treated with intratracheal lipopolysaccharide to induce lung injury. Mice were euthanized 27 days later and lung sections were stained for GFP (green), the alveolar type I (ATI) cell marker T1α (purple) and 4′,6-diamidino-2-phenylindole (DAPI; for nuclear staining (blue)). Some ATII cell-derived cells (GFP-staining cells in panels a (×40) and c (×40)) expressed ATI markers (T1α-staining cells in panels b (×40) and c), as shown by dual GFP-staining and T1α-staining cells (panel c) — indicating transdifferentiation during repair after lung injury. Arrowheads indicate nascent ATI cells that transdifferentiated from ATII cells during repair after injury (dual GFP-staining and T1α-staining cells). Arrows indicate ATI cells that withstood the initial injury (GFP-negative but T1a-staining cells). These experimental data support the notion that ATI cells are damaged during acute lung injury and are then replaced by ATII cells that transdifferentiate into ATI cells. Reprinted with permission of the American Thoracic Society. Copyright © 2019 American Thoracic Society. Jansing, N. L. et al. (2017) Unbiased quantitation of alveolar type II to alveolar type I cell transdifferentiation during repair after lung injury in mice. Am. J. Respir. Cell Mol. Biol. 57, 519–526. The American Journal of Respiratory and Critical Care Medicine is an official journal of the American Thoracic Society.
Fig. 5. The repaired alveolus.
Fig. 5. The repaired alveolus.
Several mechanisms promote endothelial cell junctional reassembly. Slit binds to its receptor, ROBO4, stabilizing the adherens junctions by promoting the association between p120–catenin and vascular endothelial cadherin (VE-cadherin) (not shown). Activated platelets release the lipid mediator sphingosine 1-phosphate (S1P), which activates Rho/Rac signalling to induce cytoskeletal reorganization that promotes endothelial barrier integrity (not shown). The receptor tyrosine kinase TIE2 is bound by its activating ligand, angiopoietin 1, which results in actin cytoskeletal reorganization and stability of VE-cadherin at the adherens junctions (not shown). To repair the damaged epithelium, surviving alveolar type II (ATII) cells replace lost epithelial cells via proliferation and differentiation into alveolar type I (ATI) cells. Many growth factors promote ATII proliferation, including keratinocyte growth factor (KGF), epidermal growth factor (EGF), hepatocyte growth factor (HGF) and granulocyte–macrophage colony-stimulating factor (GM-CSF); similarly, transcriptional pathways also promote ATII proliferation, including the WNT–β-catenin pathway,, and the forkhead box protein M1 (FOXM1) pathway. Toll-like receptor 4 and hyaluronan signalling also contribute. In severe injury, alternate progenitors (keratin 5-expressing epithelial progenitors (KRT5+) and club cells) are mobilized to proliferate and differentiate into ATII cells. Withdrawal of β-catenin signalling induces ATII cells to transdifferentiate into ATI cells. Fibroblasts and endothelial cells (and epithelial cells) secrete epithelial growth factors,; for example, platelet-derived stromal cell-derived factor 1 (SDF1) stimulates endothelial cells to secrete matrix metalloproteinase 14 (MMP14), which cleaves heparin-bound EGF (HB-EGF), enabling it to ligate the EGF receptor and stimulate ATII cell proliferation. Membrane pores can be patched and damaged mitochondria can be removed by mitophagy. Once the alveolar epithelium is regenerated, pro-resolving macrophages clear dead cells and debris and ATII and ATI epithelial cells reabsorb oedematous fluid. BASC, bronchioalveolar stem cell; ENaC, epithelial sodium channel; PMN, polymorphonuclear; RBC, red blood cell; Treg cell, regulatory T cell.
Fig. 6. Distinguishing ARDS on radiography.
Fig. 6. Distinguishing ARDS on radiography.
Similarities in the chest radiographs from a patient with acute respiratory distress syndrome (ARDS) from influenza pneumonia (panel a) and a patient with pulmonary oedema due to cardiac failure (panel b) reflect the difficulty in identifying ARDS. In both cases, diffuse bilateral parenchymal opacities are consistent with alveolar filling. The cardiac silhouette (panel b) is slightly more globular, consistent with heart failure; however, this feature is not reliable for distinguishing ARDS from cardiogenic pulmonary oedema.
Fig. 7. Common respiratory pathogens in ARDS…
Fig. 7. Common respiratory pathogens in ARDS and associated demographic features and comorbidities.
Common community-acquired and hospital-acquired pathogens that cause pneumonia should always be considered in patients with suspected acute respiratory distress syndrome (ARDS). Some organisms such as Streptococcus pneumoniae are more common as community-acquired infections whereas Pseudomonas aeruginosa is more common as a hospital-acquired infection in ARDS. Enterobacteriaceae include Klebsiella pneumoniae, Escherichia coli and Enterobacter species. The group ‘other respiratory viruses’ includes parainfluenza virus, human metapneumovirus virus, respiratory syncytial virus, rhinovirus, coronaviruses and adenovirus. A detailed, expanded version of this figure can be found in Supplementary Fig. 1. COPD, chronic obstructive pulmonary disease.
Fig. 8. Identifying patients with early acute…
Fig. 8. Identifying patients with early acute lung injury before progression to ARDS by the Berlin criteria.
Anterior–posterior chest radiographs in a critically ill 48-year-old man who presented to the emergency department with worsening dyspnoea, hypoxaemia (oxygen saturation of 70% on room air) and a 3-day history of fever, chills and a productive cough. He also had acute kidney failure with severe oliguria and a serum creatinine of 6.2 mg per dl. His systemic blood pressure was low, at 105/50 mmHg. He was diagnosed with acute pneumonia, acute renal failure and sepsis. a | Chest radiograph showing right lower lobe consolidation consistent with pneumonia. At this time, the patient was breathing spontaneously with 6 litres nasal oxygen that increased his oxygen saturation to 91%. b | Chest radiograph taken 24 hours later showing an endotracheal tube in place (arrows) for positive-pressure ventilation with bilateral opacities, consistent with the Berlin radiographic criteria. At this time, the patient had a partial pressure of arterial oxygen (PaO2) to fraction of inspired oxygen (FiO2) ratio of 125 mmHg on positive-pressure ventilation with a tidal volume of 6 ml per kg predicted body weight and a positive end-expiratory airway pressure of 15 cmH2O. The patient also had a central line (arrowhead) inserted for administration of fluids and vasopressors as he progressed to developing septic shock. Time elapsed between the images demonstrates the potential window for early acute respiratory distress syndrome (ARDS) detection and early administration of therapies designed to prevent progression.

References

    1. Ashbaugh DG, Bigelow DB, Petty TL, Levine BE. Acute respiratory distress in adults. Lancet. 1967;2:319–323. doi: 10.1016/S0140-6736(67)90168-7.
    1. Bernard GR, et al. The American-European Consensus Conference on ARDS. Definitions, mechanisms, relevant outcomes, and clinical trial coordination. Am. J. Respir. Crit. Care Med. 1994;149:818–824. doi: 10.1164/ajrccm.149.3.7509706.
    1. Ranieri VM, et al. Acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012;307:2526–2533.
    1. Rubenfeld GD, et al. Incidence and outcomes of acute lung injury. N. Engl. J. Med. 2005;353:1685–1693. doi: 10.1056/NEJMoa050333.
    1. Bellani G, 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.
    1. Pham T, Rubenfeld GD. Fifty years of research in ARDS. The epidemiology of acute respiratory distress syndrome. A 50th birthday review. Am. J. Respir. Crit. Care Med. 2017;195:860–870. doi: 10.1164/rccm.201609-1773CP.
    1. Cortegiani A, et al. Immunocompromised patients with acute respiratory distress syndrome: secondary analysis of the LUNG SAFE database. Crit. Care. 2018;22:157. doi: 10.1186/s13054-018-2079-9.
    1. Moss M, Bucher B, Moore FA, Moore EE, Parsons PE. The role of chronic alcohol abuse in the development of acute respiratory distress syndrome in adults. JAMA. 1996;275:50–54. doi: 10.1001/jama.1996.03530250054027.
    1. Calfee CS, et al. Active and passive cigarette smoking and acute lung injury after severe blunt trauma. Am. J. Respir. Crit. Care Med. 2011;183:1660–1665. doi: 10.1164/rccm.201011-1802OC.
    1. Calfee CS, et al. Cigarette smoke exposure and the acute respiratory distress syndrome. Crit. Care Med. 2015;43:1790–1797. doi: 10.1097/CCM.0000000000001089.
    1. Ware LB, et al. Long-term ozone exposure increases the risk of developing the acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 2016;193:1143–1150. doi: 10.1164/rccm.201507-1418OC.
    1. Reilly JP, et al. Low to moderate air pollutant exposure and acute respiratory distress syndrome after severe trauma. Am. J. Respir. Crit. Care Med. 2018;199:62–70. doi: 10.1164/rccm.201803-0435OC.
    1. Mangialardi RJ, et al. Hypoproteinemia predicts acute respiratory distress syndrome development, weight gain, and death in patients with sepsis. Ibuprofen Sepsis Study Group. Crit. Care Med. 2000;28:3137–3145. doi: 10.1097/00003246-200009000-00001.
    1. Moss M, et al. Diabetic patients have a decreased incidence of acute respiratory distress syndrome. Crit. Care Med. 2000;28:2187–2192. doi: 10.1097/00003246-200007000-00001.
    1. Honiden S, Gong MN. Diabetes, insulin, and development of acute lung injury. Crit. Care Med. 2009;37:2455–2464. doi: 10.1097/CCM.0b013e3181a0fea5.
    1. Boyle AJ, et al. Identifying associations between diabetes and acute respiratory distress syndrome in patients with acute hypoxemic respiratory failure: an analysis of the LUNG SAFE database. Crit. Care. 2018;22:268. doi: 10.1186/s13054-018-2158-y.
    1. Toy P, et al. Transfusion-related acute lung injury: incidence and risk factors. Blood. 2012;119:1757–1767. doi: 10.1182/blood-2011-08-370932.
    1. Robinson BR, et al. Application of the Berlin definition in PROMMTT patients: the impact of resuscitation on the incidence of hypoxemia. J. Trauma Acute Care Surg. 2013;75:S61–S67. doi: 10.1097/TA.0b013e31828fa408.
    1. Howard BM, et al. Differences in degree, differences in kind: characterizing lung injury in trauma. J. Trauma Acute Care Surg. 2015;78:735–741. doi: 10.1097/TA.0000000000000583.
    1. Erickson SE, et al. Racial and ethnic disparities in mortality from acute lung injury. Crit. Care Med. 2009;37:1–6. doi: 10.1097/CCM.0b013e31819292ea.
    1. Ryb GE, Cooper C. Race/ethnicity and acute respiratory distress syndrome: a National Trauma Data Bank study. J. Natl Med. Assoc. 2010;102:865–869. doi: 10.1016/S0027-9684(15)30700-8.
    1. Cochi SE, Kempker JA, Annangi S, Kramer MR, Martin GS. Mortality trends of acute respiratory distress syndrome in the United States from 1999 to 2013. Ann. Am. Thorac Soc. 2016;13:1742–1751.
    1. Moss M, Mannino DM. Race and gender differences in acute respiratory distress syndrome deaths in the United States: an analysis of multiple-cause mortality data (1979–1996) Crit. Care Med. 2002;30:1679–1685. doi: 10.1097/00003246-200208000-00001.
    1. Reilly JP, Christie JD, Meyer NJ. Fifty years of research in ARDS. Genomic contributions and opportunities. Am. J. Respir. Crit. Care Med. 2017;196:1113–1121. doi: 10.1164/rccm.201702-0405CP.
    1. Meyer NJ, Calfee CS. Novel translational approaches to the search for precision therapies for acute respiratory distress syndrome. Lancet Respir. Med. 2017;5:512–523. doi: 10.1016/S2213-2600(17)30187-X.
    1. Reilly JP, et al. Plasma angiopoietin-2 as a potential causal marker in sepsis-associated ARDS development: evidence from Mendelian randomization and mediation analysis. Intensive Care Med. 2018;44:1849–1858. doi: 10.1007/s00134-018-5328-0.
    1. Schouten LR, et al. Incidence and mortality of acute respiratory distress syndrome in children: a systematic review and meta-analysis. Crit. Care Med. 2016;44:819–829. doi: 10.1097/CCM.0000000000002008.
    1. Khemani RG, et al. Paediatric acute respiratory distress syndrome incidence and epidemiology (PARDIE): an international, observational study. Lancet Respir. Med. 2018;7:115–128. doi: 10.1016/S2213-2600(18)30344-8.
    1. Bindl L, et al. Gender-based differences in children with sepsis and ARDS: the ESPNIC ARDS Database Group. Intensive Care Med. 2003;29:1770–1773. doi: 10.1007/s00134-003-1948-z.
    1. Nye S, Whitley RJ, Kong M. Viral infection in the development and progression of pediatric acute respiratory distress syndrome. Front. Pediatr. 2016;4:128. doi: 10.3389/fped.2016.00128.
    1. de Roulet A, et al. Pediatric trauma-associated acute respiratory distress syndrome: incidence, risk factors, and outcomes. J. Pediatr. Surg. 2018 doi: 10.1016/j.jpedsurg.2018.07.005.
    1. Randolph AG. Management of acute lung injury and acute respiratory distress syndrome in children. Crit. Care Med. 2009;37:2448–2454. doi: 10.1097/CCM.0b013e3181aee5dd.
    1. Spicer AC, et al. A simple and robust bedside model for mortality risk in pediatric patients with acute respiratory distress syndrome. Pediatr. Crit. Care Med. 2016;17:907–916. doi: 10.1097/PCC.0000000000000865.
    1. Bhattacharya J, Matthay MA. Regulation and repair of the alveolar-capillary barrier in acute lung injury. Annu. Rev. Physiol. 2013;75:593–615. doi: 10.1146/annurev-physiol-030212-183756.
    1. Matthay MA. Resolution of pulmonary edema. Thirty years of progress. Am. J. Respir. Crit. Care Med. 2014;189:1301–1308. doi: 10.1164/rccm.201403-0535OE.
    1. Matthay MA, Ware LB, Zimmerman GA. The acute respiratory distress syndrome. J. Clin. Invest. 2012;122:2731–2740. doi: 10.1172/JCI60331.
    1. Bachofen M, Weibel ER. Structural alterations of lung parenchyma in the adult respiratory distress syndrome. Clin. Chest Med. 1982;3:35–56.
    1. Fein A, et al. The value of edema fluid protein measurement in patients with pulmonary edema. Am. J. Med. 1979;67:32–38. doi: 10.1016/0002-9343(79)90066-4.
    1. Nuckton TJ, et al. Pulmonary dead-space fraction as a risk factor for death in the acute respiratory distress syndrome. N. Engl. J. Med. 2002;346:1281–1286. doi: 10.1056/NEJMoa012835.
    1. Katzenstein AL, Bloor CM, Leibow AA. Diffuse alveolar damage—the role of oxygen, shock, and related factors. A review. Am. J. Pathol. 1976;85:209–228.
    1. Mendez JL, Hubmayr RD. New insights into the pathology of acute respiratory failure. Curr. Opin. Crit. Care. 2005;11:29–36. doi: 10.1097/00075198-200502000-00005.
    1. Cardinal-Fernandez P, Lorente JA, Ballen-Barragan A, Matute-Bello G. Acute respiratory distress syndrome and diffuse alveolar damage. New insights on a complex relationship. Ann. Am. Thorac Soc. 2017;14:844–850. doi: 10.1513/AnnalsATS.201609-728PS.
    1. Thille AW, et al. Chronology of histological lesions in acute respiratory distress syndrome with diffuse alveolar damage: a prospective cohort study of clinical autopsies. Lancet Respir. Med. 2013;1:395–401. doi: 10.1016/S2213-2600(13)70053-5.
    1. Thille AW, et al. Comparison of the Berlin definition for acute respiratory distress syndrome with autopsy. Am. J. Respir. Crit. Care Med. 2013;187:761–767. doi: 10.1164/rccm.201211-1981OC.
    1. Cardinal-Fernandez P, et al. The presence of diffuse alveolar damage on open lung biopsy is associated with mortality in patients with acute respiratory distress syndrome: a systematic review and meta-analysis. Chest. 2016;149:1155–1164. doi: 10.1016/j.chest.2016.02.635.
    1. Bachofen M, Weibel ER. Alterations of the gas exchange apparatus in adult respiratory insufficiency associated with septicemia. Am. Rev. Respir. Dis. 1977;116:589–615. doi: 10.1164/arrd.1977.116.4.589.
    1. Tomashefski JF., Jr Pulmonary pathology of acute respiratory distress syndrome. Clin. Chest Med. 2000;21:435–466. doi: 10.1016/S0272-5231(05)70158-1.
    1. Albertine KH, et al. Fas and fas ligand are up-regulated in pulmonary edema fluid and lung tissue of patients with acute lung injury and the acute respiratory distress syndrome. Am. J. Pathol. 2002;161:1783–1796. doi: 10.1016/S0002-9440(10)64455-0.
    1. Wang L, et al. Novel role of the human alveolar epithelium in regulating intra-alveolar coagulation. Am. J. Respir. Cell Mol. Biol. 2007;36:497–503. doi: 10.1165/rcmb.2005-0425OC.
    1. Bastarache JA, Fremont RD, Kropski JA, Bossert FR, Ware LB. Procoagulant alveolar microparticles in the lungs of patients with acute respiratory distress syndrome. Am. J. Physiol. Lung Cell. Mol. Physiol. 2009;297:L1035–L1041. doi: 10.1152/ajplung.00214.2009.
    1. Cheng KT, et al. Caspase-11-mediated endothelial pyroptosis underlies endotoxemia-induced lung injury. J. Clin. Invest. 2017;127:4124–4135. doi: 10.1172/JCI94495.
    1. Brigham KL, Woolverton WC, Blake LH, Staub NC. Increased sheep lung vascular permeability caused by pseudomonas bacteremia. J. Clin. Invest. 1974;54:792–804. doi: 10.1172/JCI107819.
    1. Wiener-Kronish JP, Albertine KH, Matthay MA. Differential responses of the endothelial and epithelial barriers of the lung in sheep to Escherichia coli endotoxin. J. Clin. Invest. 1991;88:864–875. doi: 10.1172/JCI115388.
    1. Gotts JE, Abbott J, Matthay MA. Influenza causes prolonged disruption of the alveolar-capillary barrier in mice unresponsive to mesenchymal stem cell therapy. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014;307:L395–L406. doi: 10.1152/ajplung.00110.2014.
    1. Frye M, et al. Interfering with VE-PTP stabilizes endothelial junctions in vivo via Tie-2 in the absence of VE-cadherin. J. Exp. Med. 2015;212:2267–2287. doi: 10.1084/jem.20150718.
    1. Giannotta M, Trani M, Dejana E. VE-cadherin and endothelial adherens junctions: active guardians of vascular integrity. Dev. Cell. 2013;26:441–454. doi: 10.1016/j.devcel.2013.08.020.
    1. Broermann A, et al. Dissociation of VE-PTP from VE-cadherin is required for leukocyte extravasation and for VEGF-induced vascular permeability in vivo. J. Exp. Med. 2011;208:2393–2401. doi: 10.1084/jem.20110525.
    1. Corada M, et al. Vascular endothelial-cadherin is an important determinant of microvascular integrity in vivo. Proc. Natl Acad. Sci. USA. 1999;96:9815–9820. doi: 10.1073/pnas.96.17.9815.
    1. Schulte D, et al. Stabilizing the VE-cadherin-catenin complex blocks leukocyte extravasation and vascular permeability. EMBO J. 2011;30:4157–4170. doi: 10.1038/emboj.2011.304.
    1. Williams AE, et al. Evidence for chemokine synergy during neutrophil migration in ARDS. Thorax. 2017;72:66–73. doi: 10.1136/thoraxjnl-2016-208597.
    1. Lefrancais E, Mallavia B, Zhuo H, Calfee CS, Looney MR. Maladaptive role of neutrophil extracellular traps in pathogen-induced lung injury. JCI Insight. 2018;3:98178. doi: 10.1172/jci.insight.98178.
    1. Abdulnour RE, et al. Early intravascular events are associated with development of acute respiratory distress syndrome. A substudy of the LIPS-A clinical trial. Am. J. Respir. Crit. Care Med. 2018;197:1575–1585. doi: 10.1164/rccm.201712-2530OC.
    1. Short KR, et al. Influenza virus damages the alveolar barrier by disrupting epithelial cell tight junctions. Eur. Respir. J. 2016;47:954–966. doi: 10.1183/13993003.01282-2015.
    1. Schlingmann B, et al. Regulation of claudin/zonula occludens-1 complexes by hetero-claudin interactions. Nat. Commun. 2016;7:12276. doi: 10.1038/ncomms12276.
    1. Shaver CM, et al. Cell-free hemoglobin promotes primary graft dysfunction through oxidative lung endothelial injury. JCI Insight. 2018;3:98546. doi: 10.1172/jci.insight.98546.
    1. Budinger GR, et al. Epithelial cell death is an important contributor to oxidant-mediated acute lung injury. Am. J. Respir. Crit. Care Med. 2011;183:1043–1054. doi: 10.1164/rccm.201002-0181OC.
    1. Hogner K, et al. Macrophage-expressed IFN-beta contributes to apoptotic alveolar epithelial cell injury in severe influenza virus pneumonia. PLOS Pathog. 2013;9:e1003188. doi: 10.1371/journal.ppat.1003188.
    1. Vaughan AE, et al. Lineage-negative progenitors mobilize to regenerate lung epithelium after major injury. Nature. 2015;517:621–625. doi: 10.1038/nature14112.
    1. Imai Y, et al. Injurious mechanical ventilation and end-organ epithelial cell apoptosis and organ dysfunction in an experimental model of acute respiratory distress syndrome. JAMA. 2003;289:2104–2112. doi: 10.1001/jama.289.16.2104.
    1. Herrero R, et al. The biological activity of FasL in human and mouse lungs is determined by the structure of its stalk region. J. Clin. Invest. 2011;121:1174–1190. doi: 10.1172/JCI43004.
    1. Saffarzadeh M, et al. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLOS ONE. 2012;7:e32366. doi: 10.1371/journal.pone.0032366.
    1. Brauer R, et al. Syndecan-1 attenuates lung injury during influenza infection by potentiating c-Met signaling to suppress epithelial apoptosis. Am. J. Respir. Crit. Care Med. 2016;194:333–344. doi: 10.1164/rccm.201509-1878OC.
    1. Hook JL, et al. Disruption of staphylococcal aggregation protects against lethal lung injury. J. Clin. Invest. 2018;128:1074–1086. doi: 10.1172/JCI95823.
    1. Vohwinkel CU, et al. Elevated CO(2) levels cause mitochondrial dysfunction and impair cell proliferation. J. Biol. Chem. 2011;286:37067. doi: 10.1074/jbc.M111.290056.
    1. Ware LB, Matthay MA. Alveolar fluid clearance is impaired in the majority of patients with acute lung injury and the acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 2001;163:1376–1383. doi: 10.1164/ajrccm.163.6.2004035.
    1. Hogan BL, et al. Repair and regeneration of the respiratory system: complexity, plasticity, and mechanisms of lung stem cell function. Cell Stem Cell. 2014;15:123–138. doi: 10.1016/j.stem.2014.07.012.
    1. Ray S, et al. Rare SOX2(+) airway progenitor cells generate KRT5(+) cells that repopulate damaged alveolar parenchyma following influenza virus infection. Stem Cell Rep. 2016;7:817–825. doi: 10.1016/j.stemcr.2016.09.010.
    1. Quantius J, et al. Influenza virus infects epithelial stem/progenitor cells of the distal lung: impact on Fgfr2b-driven epithelial repair. PLOS Pathog. 2016;12:e1005544. doi: 10.1371/journal.ppat.1005544.
    1. Xi Y, et al. Local lung hypoxia determines epithelial fate decisions during alveolar regeneration. Nat. Cell Biol. 2017;19:904–914. doi: 10.1038/ncb3580.
    1. Nabhan AN, Brownfield DG, Harbury PB, Krasnow MA, Desai TJ. Single-cell Wnt signaling niches maintain stemness of alveolar type 2 cells. Science. 2018;359:1118–1123. doi: 10.1126/science.aam6603.
    1. Dial CF, Tune MK, Doerschuk CM, Mock JR. Foxp3+regulatory T cell expression of keratinocyte growth factor enhances lung epithelial proliferation. Am. J. Respir. Cell Mol. Biol. 2017;57:162–173. doi: 10.1165/rcmb.2017-0019OC.
    1. Zemans RL, et al. Neutrophil transmigration triggers repair of the lung epithelium via beta-catenin signaling. Proc. Natl Acad. Sci. USA. 2011;108:15990–15995. doi: 10.1073/pnas.1110144108.
    1. Cong X, Hubmayr RD, Li C, Zhao X. Plasma membrane wounding and repair in pulmonary diseases. Am. J. Physiol. Lung Cell. Mol. Physiol. 2017;312:L371–L391. doi: 10.1152/ajplung.00486.2016.
    1. Schumacker PT, et al. Mitochondria in lung biology and pathology: more than just a powerhouse. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014;306:L962–L974. doi: 10.1152/ajplung.00073.2014.
    1. Fang X, Neyrinck AP, Matthay MA, Lee JW. Allogeneic human mesenchymal stem cells restore epithelial protein permeability in cultured human alveolar type II cells by secretion of angiopoietin-1. J. Biol. Chem. 2010;285:26211–26222. doi: 10.1074/jbc.M110.119917.
    1. Koval M, et al. Extracellular matrix influences alveolar epithelial claudin expression and barrier function. Am. J. Respir. Cell Mol. Biol. 2010;42:172–180. doi: 10.1165/rcmb.2008-0270OC.
    1. Gwozdzinska P, et al. Hypercapnia impairs ENaC cell surface stability by promoting phosphorylation, polyubiquitination and endocytosis of beta-ENaC in a human alveolar epithelial cell line. Front. Immunol. 2017;8:591. doi: 10.3389/fimmu.2017.00591.
    1. Vadasz I, Sznajder JI. Gas exchange disturbances regulate alveolar fluid clearance during acute lung injury. Front. Immunol. 2017;8:757. doi: 10.3389/fimmu.2017.00757.
    1. Nikolaidis NM, et al. Mitogenic stimulation accelerates influenza-induced mortality by increasing susceptibility of alveolar type II cells to infection. Proc. Natl Acad. Sci. USA. 2017;114:E6613–E6622. doi: 10.1073/pnas.1621172114.
    1. Albert RK. The role of ventilation-induced surfactant dysfunction and atelectasis in causing acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 2012;185:702–708. doi: 10.1164/rccm.201109-1667PP.
    1. Webb HH, Tierney DF. Experimental pulmonary edema due to intermittent positive pressure ventilation with high inflation pressures. Protection by positive end-expiratory pressure. Am. Rev. Respir. Dis. 1974;110:556–565.
    1. Parker JC, Townsley MI, Rippe B, Taylor AE, Thigpen J. Increased microvascular permeability in dog lungs due to high peak airway pressures. J. Appl. Physiol. Respir. Environ. Exerc. Physiol. 1984;57:1809–1816.
    1. Dreyfuss D, Soler P, Basset G, Saumon G. High inflation pressure pulmonary edema. Respective effects of high airway pressure, high tidal volume, and positive end-expiratory pressure. Am. Rev. Respir. Dis. 1988;137:1159–1164. doi: 10.1164/ajrccm/137.5.1159.
    1. Brower RG, et al. 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. doi: 10.1056/NEJM200005043421801.
    1. Tremblay L, Valenza F, Ribeiro SP, Li J, Slutsky AS. Injurious ventilatory strategies increase cytokines and c-fos m-RNA expression in an isolated rat lung model. J. Clin. Invest. 1997;99:944–952. doi: 10.1172/JCI119259.
    1. Frank JA, et al. Low tidal volume reduces epithelial and endothelial injury in acid-injured rat lungs. Am. J. Respir. Crit. Care Med. 2002;165:242–249. doi: 10.1164/ajrccm.165.2.2108087.
    1. Thompson BT, Chambers RC, Liu KD. Acute respiratory distress syndrome. N. Engl. J. Med. 2017;377:562–572. doi: 10.1056/NEJMra1608077.
    1. Sprung CL, Rackow EC, Fein IA, Jacob AI, Isikoff SK. The spectrum of pulmonary edema: differentiation of cardiogenic, intermediate, and noncardiogenic forms of pulmonary edema. Am. Rev. Respir. Dis. 1981;124:718–722.
    1. Idell S, et al. Angiotensin converting enzyme in bronchoalveolar lavage in ARDS. Chest. 1987;91:52–56. doi: 10.1378/chest.91.1.52.
    1. Rubin DB, et al. Elevated von Willebrand factor antigen is an early plasma predictor of acute lung injury in nonpulmonary sepsis syndrome. J. Clin. Invest. 1990;86:474–480. doi: 10.1172/JCI114733.
    1. Donnelly SC, et al. Role of selectins in development of adult respiratory distress syndrome. Lancet. 1994;344:215–219. doi: 10.1016/S0140-6736(94)92995-5.
    1. Parsons PE, Matthay MA, Ware LB, Eisner MD. Elevated plasma levels of soluble TNF receptors are associated with morbidity and mortality in patients with acute lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 2005;288:L426–L431. doi: 10.1152/ajplung.00302.2004.
    1. Meduri GU, et al. Inflammatory cytokines in the BAL of patients with ARDS. Persistent elevation over time predicts poor outcome. Chest. 1995;108:1303–1314. doi: 10.1378/chest.108.5.1303.
    1. Agrawal A, et al. Plasma angiopoietin-2 predicts the onset of acute lung injury in critically ill patients. Am. J. Respir. Crit. Care Med. 2013;187:736–742. doi: 10.1164/rccm.201208-1460OC.
    1. Eisner MD, et al. Plasma surfactant protein levels and clinical outcomes in patients with acute lung injury. Thorax. 2003;58:983–988. doi: 10.1136/thorax.58.11.983.
    1. Uchida T, et al. Receptor for advanced glycation end-products is a marker of type I cell injury in acute lung injury. Am. J. Respir. Crit. Care Med. 2006;173:1008–1015. doi: 10.1164/rccm.200509-1477OC.
    1. Ware LB, et al. Prognostic and pathogenetic value of combining clinical and biochemical indices in patients with acute lung injury. Chest. 2010;137:288–296. doi: 10.1378/chest.09-1484.
    1. Parsons PE, et al. Lower tidal volume ventilation and plasma cytokine markers of inflammation in patients with acute lung injury. Crit. Care Med. 2005;33:1–6. doi: 10.1097/01.CCM.0000149854.61192.DC.
    1. Calfee CS, et al. Plasma receptor for advanced glycation end products and clinical outcomes in acute lung injury. Thorax. 2008;63:1083–1089. doi: 10.1136/thx.2008.095588.
    1. Ranieri VM, et al. Effect of mechanical ventilation on inflammatory mediators in patients with acute respiratory distress syndrome: a randomized controlled trial. JAMA. 1999;282:54–61. doi: 10.1001/jama.282.1.54.
    1. Calfee CS, et al. Distinct molecular phenotypes of direct versus indirect ARDS in single-center and multicenter studies. Chest. 2015;147:1539–1548. doi: 10.1378/chest.14-2454.
    1. Calfee CS, et al. Subphenotypes in acute respiratory distress syndrome: latent class analysis of data from two randomised controlled trials. Lancet Respir. Med. 2014;2:611–620. doi: 10.1016/S2213-2600(14)70097-9.
    1. Famous KR, et al. Acute respiratory distress syndrome subphenotypes respond differently to randomized fluid management strategy. Am. J. Respir. Crit. Care Med. 2017;195:331–338. doi: 10.1164/rccm.201603-0645OC.
    1. Calfee CS, et al. Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised controlled trial. Lancet Respir. Med. 2018;6:691–698. doi: 10.1016/S2213-2600(18)30177-2.
    1. Sinha P, et al. Latent class analysis of ARDS subphenotypes: a secondary analysis of the statins for acutely injured lungs from sepsis (SAILS) study. Intensive Care Med. 2018;44:1859–1869. doi: 10.1007/s00134-018-5378-3.
    1. McAuley DF, et al. Simvastatin in the acute respiratory distress syndrome. N. Engl. J. Med. 2014;371:1695–1703. doi: 10.1056/NEJMoa1403285.
    1. Bos LD, et al. Understanding heterogeneity in biological phenotypes of ARDS by leukocyte expression profiles. Am. J. Respir. Crit. Care Med. 2019 doi: 10.1164/rccm.201809-1808OC.
    1. Morrell ED, et al. Peripheral and alveolar cell transcriptional programs are distinct in acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 2018;197:528–532. doi: 10.1164/rccm.201703-0614LE.
    1. Kangelaris KN, et al. Increased expression of neutrophil-related genes in patients with early sepsis-induced ARDS. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015;308:L1102–L1113. doi: 10.1152/ajplung.00380.2014.
    1. Rubenfeld GD, Caldwell E, Granton J, Hudson LD, Matthay MA. Interobserver variability in applying a radiographic definition for ARDS. Chest. 1999;116:1347–1353. doi: 10.1378/chest.116.5.1347.
    1. Wheeler AP, et al. Pulmonary-artery versus central venous catheter to guide treatment of acute lung injury. N. Engl. J. Med. 2006;354:2213–2224. doi: 10.1056/NEJMoa061895.
    1. Ware LB, Fremont RD, Bastarache JA, Calfee CS, Matthay MA. Determining the aetiology of pulmonary oedema by the oedema fluid-to-plasma protein ratio. Eur. Respir. J. 2010;35:331–337. doi: 10.1183/09031936.00098709.
    1. Rice TW, et al. Vascular pedicle width in acute lung injury: correlation with intravascular pressures and ability to discriminate fluid status. Crit. Care. 2011;15:R86. doi: 10.1186/cc10084.
    1. Riviello ED, et al. Hospital incidence and outcomes of the acute respiratory distress syndrome using the Kigali Modification of the Berlin Definition. Am. J. Respir. Crit. Care Med. 2016;193:52–59. doi: 10.1164/rccm.201503-0584OC.
    1. Palakshappa JA, Meyer NJ. Which patients with ARDS benefit from lung biopsy? Chest. 2015;148:1073–1082. doi: 10.1378/chest.15-0076.
    1. Khemani RG, Smith LS, Zimmerman JJ, Erickson S. Pediatric acute respiratory distress syndrome: definition, incidence, and epidemiology: proceedings from the Pediatric Acute Lung Injury Consensus Conference. Pediatr. Crit. Care Med. 2015;16:S23–S40. doi: 10.1097/PCC.0000000000000432.
    1. De Luca D, et al. The Montreux definition of neonatal ARDS: biological and clinical background behind the description of a new entity. Lancet Respir. Med. 2017;5:657–666. doi: 10.1016/S2213-2600(17)30214-X.
    1. Kao KC, et al. Coinfection and mortality in pneumonia-related acute respiratory distress syndrome patients with bronchoalveolar lavage: a prospective observational study. Shock. 2017;47:615–620. doi: 10.1097/SHK.0000000000000802.
    1. Hong DK, et al. Liquid biopsy for infectious diseases: sequencing of cell-free plasma to detect pathogen DNA in patients with invasive fungal disease. Diagn. Microbiol. Infect. Dis. 2018;92:210–213. doi: 10.1016/j.diagmicrobio.2018.06.009.
    1. Fischer N, et al. Rapid metagenomic diagnostics for suspected outbreak of severe pneumonia. Emerg. Infect. Dis. 2014;20:1072–1075. doi: 10.3201/eid2006.131526.
    1. Hasvold J, Sjoding M, Pohl K, Cooke C, Hyzy RC. The role of human metapneumovirus in the critically ill adult patient. J. Crit. Care. 2016;31:233–237. doi: 10.1016/j.jcrc.2015.09.035.
    1. Robert D, et al. A series of five adult cases of respiratory syncytial virus-related acute respiratory distress syndrome. Anaesth. Intensive Care. 2008;36:230–234.
    1. Ferguson ND, et al. Clinical risk conditions for acute lung injury in the intensive care unit and hospital ward: a prospective observational study. Crit. Care. 2007;11:R96. doi: 10.1186/cc6113.
    1. Matthay MA. Challenges in predicting which patients will develop ARDS. Lancet Respir. Med. 2016;4:847–848. doi: 10.1016/S2213-2600(16)30306-X.
    1. Levitt JE, Calfee CS, Goldstein BA, Vojnik R, Matthay MA. Early acute lung injury: criteria for identifying lung injury prior to the need for positive pressure ventilation*. Crit. Care Med. 2013;41:1929–1937. doi: 10.1097/CCM.0b013e31828a3d99.
    1. Gajic O, et al. Early identification of patients at risk of acute lung injury: evaluation of lung injury prediction score in a multicenter cohort study. Am. J. Respir. Crit. Care Med. 2011;183:462–470. doi: 10.1164/rccm.201004-0549OC.
    1. Kor DJ, et al. Effect of aspirin on development of ARDS in at-risk patients presenting to the emergency department: the LIPS-A randomized clinical trial. JAMA. 2016;315:2406–2414. doi: 10.1001/jama.2016.6330.
    1. Levitt JE, Bedi H, Calfee CS, Gould MK, Matthay MA. Identification of early acute lung injury at initial evaluation in an acute care setting prior to the onset of respiratory failure. Chest. 2009;135:936–943. doi: 10.1378/chest.08-2346.
    1. Levitt JE, Matthay MA. Clinical review: early treatment of acute lung injury—paradigm shift toward prevention and treatment prior to respiratory failure. Crit. Care. 2012;16:223. doi: 10.1186/cc11144.
    1. Afshar M, et al. Injury characteristics and von Willebrand Factor for the prediction of acute respiratory distress syndrome in patients with burn injury: development and internal validation. Ann. Surg. 2018 doi: 10.1097/SLA.0000000000002795.
    1. Liu X, et al. Plasma sRAGE enables prediction of acute lung injury after cardiac surgery in children. Crit. Care. 2012;16:R91. doi: 10.1186/cc11354.
    1. Jabaudon M, et al. Receptor for advanced glycation end-products and ARDS prediction: a multicentre observational study. Sci. Rep. 2018;8:2603. doi: 10.1038/s41598-018-20994-x.
    1. Luce JM, et al. 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. doi: 10.1164/ajrccm/138.1.62.
    1. Weigelt JA, Norcross JF, Borman KR, Snyder WH., 3rd Early steroid therapy for respiratory failure. Arch. Surg. 1985;120:536–540. doi: 10.1001/archsurg.1985.01390290018003.
    1. Festic E, et al. Randomized clinical trial of a combination of an inhaled corticosteroid and beta agonist in patients at risk of developing the acute respiratory distress syndrome. Crit. Care Med. 2017;45:798–805. doi: 10.1097/CCM.0000000000002284.
    1. Determann RM, et al. Ventilation with lower tidal volumes as compared with conventional tidal volumes for patients without acute lung injury: a preventive randomized controlled trial. Crit. Care. 2010;14:R1. doi: 10.1186/cc8230.
    1. Neto AS, Jaber S. What’s new in mechanical ventilation in patients without ARDS: lessons from the ARDS literature. Intensive Care Med. 2016;42:787–789. doi: 10.1007/s00134-016-4309-4.
    1. Writing Group for the, P. I. et al. Effect of a low versus intermediate tidal volume strategy on ventilator-free days in intensive care unit patients without ARDS: a randomized clinical trial. JAMA. 2018;320:1872–1880. doi: 10.1001/jama.2018.14280.
    1. Li G, et al. Eight-year trend of acute respiratory distress syndrome: a population-based study in Olmsted County, Minnesota. Am. J. Respir. Crit. Care Med. 2011;183:59–66. doi: 10.1164/rccm.201003-0436OC.
    1. Ahmed AH, et al. The role of potentially preventable hospital exposures in the development of acute respiratory distress syndrome: a population-based study. Crit. Care Med. 2014;42:31–39. doi: 10.1097/CCM.0b013e318298a6db.
    1. Frat JP, et al. High-flow oxygen through nasal cannula in acute hypoxemic respiratory failure. N. Engl. J. Med. 2015;372:2185–2196. doi: 10.1056/NEJMoa1503326.
    1. Azoulay E, et al. Effect of high-flow nasal oxygen versus standard oxygen on 28-day mortality in immunocompromised patients with acute respiratory failure: the HIGH randomized clinical trial. JAMA. 2018;320:2099–2107. doi: 10.1001/jama.2018.14282.
    1. Drusano GL. What are the properties that make an antibiotic acceptable for therapy of community-acquired pneumonia? J. Antimicrob. Chemother. 2011;66(Suppl. 3):61–67.
    1. Roberts JA, et al. Continuous versus intermittent beta-lactam infusion in severe sepsis. A meta-analysis of individual patient data from randomized trials. Am. J. Respir. Crit. Care Med. 2016;194:681–691. doi: 10.1164/rccm.201601-0024OC.
    1. Ross JT, Matthay MA, Harris HW. Secondary peritonitis: principles of diagnosis and intervention. BMJ. 2018;361:k1407. doi: 10.1136/bmj.k1407.
    1. Gattinoni L, et al. Lung recruitment in patients with the acute respiratory distress syndrome. N. Engl. J. Med. 2006;354:1775–1786. doi: 10.1056/NEJMoa052052.
    1. Gattinoni L, Pesenti A. The concept of “baby lung”. Intensive Care Med. 2005;31:776–784. doi: 10.1007/s00134-005-2627-z.
    1. Beitler JR, et al. Volume delivered during recruitment maneuver predicts lung stress in acute respiratory distress syndrome. Crit. Care Med. 2016;44:91–99. doi: 10.1097/CCM.0000000000001355.
    1. Amato MB, et al. Driving pressure and survival in the acute respiratory distress syndrome. N. Engl. J. Med. 2015;372:747–755. doi: 10.1056/NEJMsa1410639.
    1. Talmor D, et al. Mechanical ventilation guided by esophageal pressure in acute lung injury. N. Engl. J. Med. 2008;359:2095–2104. doi: 10.1056/NEJMoa0708638.
    1. Beitler, J. R. et al. EPVent-2 Study Group. Effect of titrating positive end-expiratory pressure (PEEP) with an esophageal pressure-guided strategy versus an empiric high PEEP-FiO2 strategy on death and days free from mechanical ventilation among patients with acute respiratory distress syndrome: a randomized clinical trial. JAMA (in the press).
    1. Brower RG, et al. Higher versus lower positive end-expiratory pressures in patients with the acute respiratory distress syndrome. N. Engl. J. Med. 2004;351:327–336. doi: 10.1056/NEJMoa032193.
    1. Meade MO, et al. Ventilation strategy using low tidal volumes, recruitment maneuvers, and high positive end-expiratory pressure for acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. JAMA. 2008;299:637–645. doi: 10.1001/jama.299.6.637.
    1. Mercat A, et al. Positive end-expiratory pressure setting in adults with acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. JAMA. 2008;299:646–655. doi: 10.1001/jama.299.6.646.
    1. Briel M, et al. Higher versus lower positive end-expiratory pressure in patients with acute lung injury and acute respiratory distress syndrome: systematic review and meta-analysis. JAMA. 2010;303:865–873. doi: 10.1001/jama.2010.218.
    1. Cavalcanti AB, et al. Effect of lung recruitment and titrated positive end-expiratory pressure (PEEP) vs low PEEP on mortality in patients with acute respiratory distress syndrome: a randomized clinical trial. JAMA. 2017;318:1335–1345. doi: 10.1001/jama.2017.14171.
    1. Sahetya SK, Brower RG. Lung recruitment and titrated PEEP in moderate to severe ARDS: is the door closing on the open lung? JAMA. 2017;318:1327–1329. doi: 10.1001/jama.2017.13695.
    1. Goligher EC, et al. Lung recruitment maneuvers for adult patients with acute respiratory distress syndrome. A systematic review and meta-analysis. Ann. Am. Thorac Soc. 2017;14:S304–S311. doi: 10.1513/AnnalsATS.201704-340OT.
    1. Gattinoni L, Taccone P, Carlesso E, Marini JJ. Prone position in acute respiratory distress syndrome. Rationale, indications, and limits. Am. J. Respir. Crit. Care Med. 2013;188:1286–1293. doi: 10.1164/rccm.201308-1532CI.
    1. Guerin C, et al. Prone positioning in severe acute respiratory distress syndrome. N. Engl. J. Med. 2013;368:2159–2168. doi: 10.1056/NEJMoa1214103.
    1. Beitler JR, et al. Prone positioning reduces mortality from acute respiratory distress syndrome in the low tidal volume era: a meta-analysis. Intensive Care Med. 2014;40:332–341. doi: 10.1007/s00134-013-3194-3.
    1. Fan E, 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;195:1253–1263. doi: 10.1164/rccm.201703-0548ST.
    1. Brochard L, Slutsky A, Pesenti A. Mechanical ventilation to minimize progression of lung injury in acute respiratory failure. Am. J. Respir. Crit. Care Med. 2017;195:438–442. doi: 10.1164/rccm.201605-1081CP.
    1. Beitler JR, et al. Quantifying unintended exposure to high tidal volumes from breath stacking dyssynchrony in ARDS: the BREATHE criteria. Intensive Care Med. 2016;42:1427–1436. doi: 10.1007/s00134-016-4423-3.
    1. Papazian L, et al. Neuromuscular blockers in early acute respiratory distress syndrome. N. Engl. J. Med. 2010;363:1107–1116. doi: 10.1056/NEJMoa1005372.
    1. Huang DT, et al. Design and rationale of the reevaluation of systemic early neuromuscular blockade trial for acute respiratory distress syndrome. Ann. Am. Thorac Soc. 2017;14:124–133. doi: 10.1513/AnnalsATS.201608-629OT.
    1. Ely EW, et al. Delirium as a predictor of mortality in mechanically ventilated patients in the intensive care unit. JAMA. 2004;291:1753–1762. doi: 10.1001/jama.291.14.1753.
    1. Schweickert WD, et al. Early physical and occupational therapy in mechanically ventilated, critically ill patients: a randomised controlled trial. Lancet. 2009;373:1874–1882. doi: 10.1016/S0140-6736(09)60658-9.
    1. Antonelli M, et al. A comparison of noninvasive positive-pressure ventilation and conventional mechanical ventilation in patients with acute respiratory failure. N. Engl. J. Med. 1998;339:429–435. doi: 10.1056/NEJM199808133390703.
    1. Bellani G, et al. Noninvasive ventilation of patients with acute respiratory distress syndrome. Insights from the LUNG SAFE Study. Am. J. Respir. Crit. Care Med. 2017;195:67–77. doi: 10.1164/rccm.201606-1306OC.
    1. Patel BK, Wolfe KS, Pohlman AS, Hall JB, Kress JP. Effect of noninvasive ventilation delivered by helmet versus face mask on the rate of endotracheal intubation in patients with acute respiratory distress syndrome: a randomized clinical trial. JAMA. 2016;315:2435–2441. doi: 10.1001/jama.2016.6338.
    1. Beitler JR, Owens RL, Malhotra A. Unmasking a role for noninvasive ventilation in early acute respiratory distress syndrome. JAMA. 2016;315:2401–2403. doi: 10.1001/jama.2016.5987.
    1. Matthay MA. Saving lives with high-flow nasal oxygen. N. Engl. J. Med. 2015;372:2225–2226. doi: 10.1056/NEJMe1504852.
    1. Calfee CS, Matthay MA. Nonventilatory treatments for acute lung injury and ARDS. Chest. 2007;131:913–920. doi: 10.1378/chest.06-1743.
    1. Wiedemann HP, et al. Comparison of two fluid-management strategies in acute lung injury. N. Engl. J. Med. 2006;354:2564–2575. doi: 10.1056/NEJMoa062200.
    1. Mikkelsen ME, et al. The adult respiratory distress syndrome cognitive outcomes study: long-term neuropsychological function in survivors of acute lung injury. Am. J. Respir. Crit. Care Med. 2012;185:1307–1315. doi: 10.1164/rccm.201111-2025OC.
    1. Zinter MS, et al. Positive cumulative fluid balance is associated with mortality in pediatric acute respiratory distress syndrome in the setting of acute kidney injury. Pediatr. Crit. Care Med. 2019 doi: 10.1097/PCC.0000000000001845.
    1. Fielding-Singh V, Matthay MA, Calfee CS. Beyond low tidal volume ventilation: treatment adjuncts for severe respiratory failure in acute respiratory distress syndrome. Crit. Care Med. 2018;46:1820–1831. doi: 10.1097/CCM.0000000000003406.
    1. Combes A, et al. Position paper for the organization of extracorporeal membrane oxygenation programs for acute respiratory failure in adult patients. Am. J. Respir. Crit. Care Med. 2014;190:488–496. doi: 10.1164/rccm.201404-0630CP.
    1. Davies A, et al. Extracorporeal membrane oxygenation for 2009 influenza A(H1N1) acute respiratory distress syndrome. JAMA. 2009;302:1888–1895. doi: 10.1001/jama.2009.1535.
    1. Combes A, et al. Extracorporeal membrane oxygenation for severe acute respiratory distress syndrome. N. Engl. J. Med. 2018;378:1965–1975. doi: 10.1056/NEJMoa1800385.
    1. Mi MY, Matthay MA, Morris AH. Extracorporeal membrane oxygenation for severe acute respiratory distress syndrome. N. Engl. J. Med. 2018;379:884–887. doi: 10.1056/NEJMclde1804601.
    1. Fanelli V, et al. Feasibility and safety of low-flow extracorporeal carbon dioxide removal to facilitate ultra-protective ventilation in patients with moderate acute respiratory distress sindrome. Crit. Care. 2016;20:36. doi: 10.1186/s13054-016-1211-y.
    1. Terragni PP, et al. Tidal volume lower than 6ml/kg enhances lung protection: role of extracorporeal carbon dioxide removal. Anesthesiology. 2009;111:826–835. doi: 10.1097/ALN.0b013e3181b764d2.
    1. Bein T, et al. Lower tidal volume strategy (approximately 3ml/kg) combined with extracorporeal CO2 removal versus ‘conventional’ protective ventilation (6ml/kg) in severe ARDS: the prospective randomized Xtravent-study. Intensive Care Med. 2013;39:847–856. doi: 10.1007/s00134-012-2787-6.
    1. Meduri GU, et al. Prolonged glucocorticoid treatment is associated with improved ARDS outcomes: analysis of individual patients’ data from four randomized trials and trial-level meta-analysis of the updated literature. Intensive Care Med. 2016;42:829–840. doi: 10.1007/s00134-015-4095-4.
    1. Steinberg KP, et al. Efficacy and safety of corticosteroids for persistent acute respiratory distress syndrome. N. Engl. J. Med. 2006;354:1671–1684. doi: 10.1056/NEJMoa051693.
    1. Ewald H, et al. Adjunctive corticosteroids for Pneumocystis jiroveci pneumonia in patients with HIV infection. Cochrane Database Syst. Rev. 2015;4:CD006150.
    1. Griffiths MJ, Evans TW. Inhaled nitric oxide therapy in adults. N. Engl. J. Med. 2005;353:2683–2695. doi: 10.1056/NEJMra051884.
    1. Taylor RW, et al. Low-dose inhaled nitric oxide in patients with acute lung injury: a randomized controlled trial. JAMA. 2004;291:1603–1609. doi: 10.1001/jama.291.13.1603.
    1. Repesse X, Charron C, Vieillard-Baron A. Acute cor pulmonale in ARDS: rationale for protecting the right ventricle. Chest. 2015;147:259–265. doi: 10.1378/chest.14-0877.
    1. Young D, et al. High-frequency oscillation for acute respiratory distress syndrome. N. Engl. J. Med. 2013;368:806–813. doi: 10.1056/NEJMoa1215716.
    1. Ferguson ND, et al. High-frequency oscillation in early acute respiratory distress syndrome. N. Engl. J. Med. 2013;368:795–805. doi: 10.1056/NEJMoa1215554.
    1. Bateman ST, et al. Early high-frequency oscillatory ventilation in pediatric acute respiratory failure. A propensity score analysis. Am. J. Respir. Crit. Care Med. 2016;193:495–503. doi: 10.1164/rccm.201507-1381OC.
    1. Putensen C, et al. Long-term effects of spontaneous breathing during ventilatory support in patients with acute lung injury. Am. J. Respir. Crit. Care Med. 2001;164:43–49. doi: 10.1164/ajrccm.164.1.2001078.
    1. Putensen C, Mutz NJ, Putensen-Himmer G, Zinserling J. Spontaneous breathing during ventilatory support improves ventilation-perfusion distributions in patients with acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 1999;159:1241–1248. doi: 10.1164/ajrccm.159.4.9806077.
    1. Lalgudi Ganesan S, Jayashree M, Singhi SC, Bansal A. Airway pressure release ventilation in pediatric acute respiratory distress syndrome: a randomized controlled trial. Am. J. Respir. Crit. Care Med. 2018;198:1199–1207. doi: 10.1164/rccm.201705-0989OC.
    1. Matthay MA, McAuley DF, Ware LB. Clinical trials in acute respiratory distress syndrome: challenges and opportunities. Lancet Respir. Med. 2017;5:524–534. doi: 10.1016/S2213-2600(17)30188-1.
    1. Downs JB, Olsen GN. Pulmonary function following adult respiratory distress syndrome. Chest. 1974;65:92–93. doi: 10.1378/chest.65.1.92.
    1. Lakshminarayan S, Stanford RE, Petty TL. Prognosis after recovery from adult respiratory distress syndrome. Am. Rev. Respir. Dis. 1976;113:7–16.
    1. Klein JJ, van Haeringen JR, Sluiter HJ, Holloway R, Peset R. Pulmonary function after recovery from the adult respiratory distress syndrome. Chest. 1976;69:350–355. doi: 10.1378/chest.69.3.350.
    1. Weinert CR, Gross CR, Kangas JR, Bury CL, Marinelli WA. Health-related quality of life after acute lung injury. Am. J. Respir. Crit. Care Med. 1997;156:1120–1128. doi: 10.1164/ajrccm.156.4.9611047.
    1. Schelling G, et al. Health-related quality of life and posttraumatic stress disorder in survivors of the acute respiratory distress syndrome. Crit. Care Med. 1998;26:651–659. doi: 10.1097/00003246-199804000-00011.
    1. Davidson TA, Caldwell ES, Curtis JR, Hudson LD, Steinberg KP. Reduced quality of life in survivors of acute respiratory distress syndrome compared with critically ill control patients. JAMA. 1999;281:354–360. doi: 10.1001/jama.281.4.354.
    1. McHugh LG, et al. Recovery of function in survivors of the acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 1994;150:90–94. doi: 10.1164/ajrccm.150.1.8025779.
    1. Angus DC, et al. Quality-adjusted survival in the first year after the acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 2001;163:1389–1394. doi: 10.1164/ajrccm.163.6.2005123.
    1. Herridge MS, et al. One-year outcomes in survivors of the acute respiratory distress syndrome. N. Engl. J. Med. 2003;348:683–693. doi: 10.1056/NEJMoa022450.
    1. Herridge MS, et al. Functional disability 5 years after acute respiratory distress syndrome. N. Engl. J. Med. 2011;364:1293–1304. doi: 10.1056/NEJMoa1011802.
    1. Fan E, et al. Physical complications in acute lung injury survivors: a two-year longitudinal prospective study. Crit. Care Med. 2014;42:849–859. doi: 10.1097/CCM.0000000000000040.
    1. Needham DM, et al. Risk factors for physical impairment after acute lung injury in a national, multicenter study. Am. J. Respir. Crit. Care Med. 2014;189:1214–1224. doi: 10.1164/rccm.201401-0158OC.
    1. Pfoh ER, et al. Physical declines occurring after hospital discharge in ARDS survivors: a 5-year longitudinal study. Intensive Care Med. 2016;42:1557–1566. doi: 10.1007/s00134-016-4530-1.
    1. Hopkins RO, et al. Neuropsychological sequelae and impaired health status in survivors of severe acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 1999;160:50–56. doi: 10.1164/ajrccm.160.1.9708059.
    1. Hopkins RO, et al. Two-year cognitive, emotional, and quality-of-life outcomes in acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 2005;171:340–347. doi: 10.1164/rccm.200406-763OC.
    1. Dowdy DW, et al. Intensive care unit hypoglycemia predicts depression during early recovery from acute lung injury. Crit. Care Med. 2008;36:2726–2733. doi: 10.1097/CCM.0b013e31818781f5.
    1. Pandharipande PP, et al. Long-term cognitive impairment after critical illness. N. Engl. J. Med. 2013;369:1306–1316. doi: 10.1056/NEJMoa1301372.
    1. Ferrante LE, et al. Functional trajectories among older persons before and after critical illness. JAMA Intern. Med. 2015;175:523–529. doi: 10.1001/jamainternmed.2014.7889.
    1. Kress JP, Hall JB. ICU-acquired weakness and recovery from critical illness. N. Engl. J. Med. 2014;371:287–288. doi: 10.1056/NEJMc1406274.
    1. Levine S, et al. Rapid disuse atrophy of diaphragm fibers in mechanically ventilated humans. N. Engl. J. Med. 2008;358:1327–1335. doi: 10.1056/NEJMoa070447.
    1. Puthucheary ZA, et al. Acute skeletal muscle wasting in critical illness. JAMA. 2013;310:1591–1600. doi: 10.1001/jama.2013.278481.
    1. Dos Santos C, et al. Mechanisms of chronic muscle wasting and dysfunction after an intensive care unit stay. A pilot study. Am. J. Respir. Crit. Care Med. 2016;194:821–830. doi: 10.1164/rccm.201512-2344OC.
    1. Warren MA, et al. Severity scoring of lung oedema on the chest radiograph is associated with clinical outcomes in ARDS. Thorax. 2018;73:840–846. doi: 10.1136/thoraxjnl-2017-211280.
    1. Sinha P, et al. Physiologic analysis and clinical performance of the ventilatory ratio in acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 2019;199:333–341. doi: 10.1164/rccm.201804-0692OC.
    1. Langelier C, et al. Integrating host response and unbiased microbe detection for lower respiratory tract infection diagnosis in critically ill adults. Proc. Natl Acad. Sci. USA. 2018;115:E12353–E12362. doi: 10.1073/pnas.1809700115.
    1. Boyle AJ, et al. Extracorporeal carbon dioxide removal for lowering the risk of mechanical ventilation: research questions and clinical potential for the future. Lancet Respir. Med. 2018;6:874–884. doi: 10.1016/S2213-2600(18)30326-6.
    1. Laffey JG, Matthay MA. Fifty years of research in ARDS. Cell-based therapy for acute respiratory distress syndrome. Biology and potential therapeutic value. Am. J. Respir. Crit. Care Med. 2017;196:266–273. doi: 10.1164/rccm.201701-0107CP.
    1. Matthay MA, et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised phase 2a safety trial. Lancet Respir. Med. 2018;7:P154–P162. doi: 10.1016/S2213-2600(18)30418-1.
    1. Matthay MA, Zimmerman GA. Acute lung injury and the acute respiratory distress syndrome: four decades of inquiry into pathogenesis and rational management. Am. J. Respir. Cell Mol. Biol. 2005;33:319–327. doi: 10.1165/rcmb.F305.
    1. Jansing NL, et al. Unbiased quantitation of alveolar type II to alveolar type I cell transdifferentiation during repair after lung injury in mice. Am. J. Respir. Cell Mol. Biol. 2017;57:519–526. doi: 10.1165/rcmb.2017-0037MA.
    1. Zacharias WJ, et al. Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor. Nature. 2018;555:251–255. doi: 10.1038/nature25786.
    1. Liu Y, et al. FoxM1 mediates the progenitor function of type II epithelial cells in repairing alveolar injury induced by Pseudomonas aeruginosa. J. Exp. Med. 2011;208:1473–1484. doi: 10.1084/jem.20102041.
    1. Liang J, et al. Hyaluronan and TLR4 promote surfactant-protein-C-positive alveolar progenitor cell renewal and prevent severe pulmonary fibrosis in mice. Nat. Med. 2016;22:1285–1293. doi: 10.1038/nm.4192.
    1. Rafii S, et al. Platelet-derived SDF-1 primes the pulmonary capillary vascular niche to drive lung alveolar regeneration. Nat. Cell Biol. 2015;17:123–136. doi: 10.1038/ncb3096.

Source: PubMed

3
Předplatit