You are on page 1of 14

Series

Acute Respiratory Distress Syndrome 2022 2


Acute respiratory distress syndrome in adults: diagnosis,
outcomes, long-term sequelae, and management
Ellen A Gorman, Cecilia M O’Kane, Daniel F McAuley

Acute respiratory distress syndrome (ARDS) is characterised by acute hypoxaemic respiratory failure with bilateral Published Online
infiltrates on chest imaging, which is not fully explained by cardiac failure or fluid overload. ARDS is defined by the September 4, 2022
https://doi.org/10.1016/
Berlin criteria. In this Series paper the diagnosis, management, outcomes, and long-term sequelae of ARDS are S0140-6736(22)01439-8
reviewed. Potential limitations of the ARDS definition and evidence that could inform future revisions are considered. This is the second in a Series of
Guideline recommendations, evidence, and uncertainties in relation to ARDS management are discussed. The future three papers about acute
of ARDS strives towards a precision medicine approach, and the framework of treatable traits in ARDS diagnosis and respiratory distress syndrome
management is explored. (paper 3 appears in The Lancet
Respiratory Medicine). All papers
in the Series are available at
Introduction Diagnosis thelancet.com/series/ARDS-2022
Acute respiratory distress syndrome (ARDS) is The utility of diagnostic tools in health care is their ability Wellcome-Wolfson Institute
characterised by acute hypoxaemic respiratory failure to inform clinical decision making and identify popu- for Experimental Medicine,
with bilateral infiltrates on chest imaging, which is not lations of patients with common sets of characteristics, Queen’s University
Belfast, Belfast, UK
fully explained by cardiac failure or fluid overload.1 It is outcomes, and treatment responsiveness.17 ARDS is a (E A Gorman MB BCh BAO,
precipitated by a predisposing risk factor such as syndrome that as an entity has progressed through Prof C M O’Kane PhD,
pneumonia, non-pulmonary sepsis, gastric aspiration, various iterations of diagnostic criteria since its first Prof D F McAuley MD)
trauma, pancreatitis, burns, inhalation injury, drug description by Ashbaugh and colleagues in 1967.18 There Correspondence to:
overdose, multiple transfusions, or shock.2 The pre­ were no uniformly accepted criteria for ARDS until the Prof Daniel F McAuley,
Wellcome-Wolfson Institute
cipitating insult triggers a cascade of dysregulated development of the American–European Consensus
for Experimental Medicine,
inflammatory responses and cytokine activation. Injury Conference definition in 1994.19 Addressing limitations of Queen’s University Belfast,
to the alveolar epithelial–endothelial barrier could that definition, the current Berlin definition (panel 2) Belfast BT9 7BL, UK
occur directly due to pulmonary insults, with primary was developed in 2012, by consensus of an expert panel.1 d.f.mcauley@qub.ac.uk
damage to the lung epithelium, or indirectly due to One benefit of a standardised definition for ARDS has
extra­pulmonary insults, with primary damage to the been to facilitate recruitment into clinical trials, which
vascular endothelium as a result of systemic has led to the development of effective supportive
inflammation.3 However, notably, the majority of therapies. A standardised definition also allows clinicians
patients with ARDS will most likely have features of to readily understand the population recruited to a
both direct and indirect lung injury.3 The resultant clinical trial and therefore apply the evidence generated
disruption of the alveolar epithelial–endothelial barrier from those trials to inform the treatment of the
results in accumulation of a protein-rich pulmonary appropriate patients in clinical care.20
oedema, surfactant dysfunction, and impaired gas Patients with ARDS represent a subset of a broader
exchange.4 ARDS can be associated with physiological population of acute hypoxaemic respiratory failure. The
derange­ment, including decreased respiratory system key difference between ARDS and acute hypoxaemic
compliance, increased physiological dead space, and respiratory failure is the requirement for bilateral
increased shunting, along with histological features of infiltrates on chest imaging. Evidence from the LUNG
lung oedema, inflammation, hyaline membranes, and SAFE study21 indicates that outcomes are similar for
alveolar haemorrhage.2 Classically, the histological
hallmark of ARDS was described as diffuse alveolar
damage; however, in autopsy data this damage has been Search strategy and selection criteria
reported to be present in less than half of patients with References for this Series paper were identified through
ARDS.5 searches of PubMed for articles published from the database
In this Series we discuss the diagnosis, management, inception to July 15, 2022, by use of the terms “ARDS”,
out­comes, and long-term sequelae of ARDS. As the “diagnosis”, “outcomes”, “ventilation”, “management”,
majority of critically ill patients with severe COVID-19 and “guidelines”. Relevant references cited in papers
are likely to fulfil the criteria for ARDS,6 evidence identified were also reviewed. We focused on clinical studies,
related to COVID-19 ARDS is considered throughout and the final list of cited articles was selected on the basis of
this Series. Panel 1 contextualises our current their relevance to the aims of this Series paper. ClinicalTrials.
understanding of the similarities and differences gov was also searched using the term “ARDS” and a selection
between COVID-19 ARDS and ARDS due to other of trials were selected.
causes.7–9,11

www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8 1


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

Panel 1: COVID-19 ARDS compared with non-COVID-19 Panel 2: The Berlin definition of acute respiratory distress
ARDS—summary of key features syndrome1
• Timing: onset could be more than 7 days from • Timing: within 1 week of a known clinical insult or new or
SARS-COV-2 infection and symptom onset7,8 worsening respiratory symptoms
• Demographics: disparities reported for non-White ethnic • Chest imaging (ie, chest radiograph or CT scan): bilateral
populations and health-care workers9 opacities not fully explained by effusions, lobar or lung
• Thoracic imaging: overlapping thoracic CT findings during collapse, or nodules
the acute phase, but predominance of diffuse pattern with • Origin of oedema: respiratory failure not fully explained
ground glass opacities in COVID-19 ARDS;8 evidence of by cardiac failure or fluid overload; need objective
persistent interstitial lung disease but uncertainty regarding assessment (eg, echocardiography) to exclude hydrostatic
how this finding compares with non-COVID-19 ARDS10 oedema if no risk factor present
• Respiratory mechanics: respiratory system mechanics • Oxygenation (if altitude is higher than 1000 m, the
(including compliance, plateau pressure, and driving correction factor should be calculated, using the ratio
pressure) reported to be similar7–9,11 between partial pressure of arterial oxygen [PaO2] and
• Biomarkers: lower total white cell count (predominantly fraction of inspired oxygen [FiO2] as:
due to a lower neutrophil count) in patients with [PaO2/FiO2 × (barometric pressure/760)])
COVID-19 ARDS;11 reduced IL-6 reported,9 but similarities • Mild: 200 mm Hg > PaO2/FiO2 ≤300 mm Hg with
in other markers of systemic inflammation and positive end-expiratory pressure or continuous
extrapulmonary organ dysfunction;11 hyperinflammatory positive airway pressure ≥5 cm H2O (this can be
phenotype less prevalent12 delivered non-invasively in the mild acute respiratory
• Coagulation: higher platelet count and fibrinogen, lower distress syndrome group)
prothrombin time, and activated partial thromboplastin • Moderate: 100 mm Hg > PaO2/FiO2 ≤200 mm Hg with
time in COVID-19 patients than in non-COVID-19 positive end-expiratory pressure ≥5 cm H2O
patients;11 D-dimer higher in non-COVID-19 ARDS than • Severe: PaO2/FiO2 ≤ 100 mm Hg with positive
COVID-19 ARDS;11 higher incidence of deep venous end-expiratory pressure ≥5 cm H2O
thrombosis in COVID-19 ARDS13
• Pharmacological agents: pharmacological agents
recommended for severe COVID-19 (including COVID-19 of several aspects of the current syndromic definition
ARDS) include corticosteroids, IL-6 receptor blockers, of ARDS, which might usefully be addressed in an
or baricitinib (Janus Kinase inhibitor);14 no specific updated ARDS definition, is timely. Figure 1 summarises
pharmacological agents are recommended for ARDS due how ARDS criteria have evolved over time and how these
to other causes criteria could evolve in the future.
• Adjunctive therapy: paucity of randomised control trial Chest radiographic criteria for ARDS are recognised to
evidence directly relevant to COVID-19 populations; use have suboptimal interobserver reliability and are under-
of recruitment manoeuvres, prone position, and recognised in clinical settings.22 In research settings tools
neuromuscular blockade reported to be higher in have been investigated to improve the reliability of ARDS
COVID-19 ARDS populations than in ARDS populations diagnosis. The Radiographic Assessment of Lung
without COVID-19 7,11 Oedema score, using a visual assessment of four
• Critical care outcomes: prolonged duration of ventilation quadrant consolidation and infiltrate density, has been
in patients with COVID-19 has been reported;9 mortality shown to have good intraobserver reliability and high
outcomes similar7,8,11 diagnostic accuracy for ARDS.23 A similar visual
• Long-term outcomes: no difference in incidence of new assessment score has been shown to correlate with
disability at 6 months;15 similar health-related quality of important clinical outcomes including mortality and
life, psychological, and cognitive function at 6 months;15 duration of intensive care unit stay.24 Artificial intelligence
in survivors of COVID-19 an increased return to work at technology (using deep convolutional neural networks
6 months15 and 1 year has been reported16 that can be trained to recognise findings on imaging),
For more on the ARDS Detection such as the ARDS Detection Tool, is another tool that has
tool see https://ardsdetect.com been shown to accurately identify bilateral airspace
patients with unilateral or bilateral infiltrates, suggesting consolidation consistent with ARDS in research settings
that bilateral infiltrates might not be needed as part of but requires further validation before clinical use.25
the syndromic definition of ARDS. Further research is Ultrasound imaging is emerging as a safe,
needed to understand the similarities and differences in inexpensive, bedside tool for the evaluation of ARDS
the clinical and biological characteristics of patients with although the need for training is recognised to be
ARDS and acute hypoxaemic respiratory failure. essential before this tool can be implemented as a
With the evolution of clinical care and increasing diagnostic imaging modality for pulmonary infiltrates.
recognition of the global burden of ARDS, consideration A ratio between oxygen saturation, measured by pulse

2 www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

Previous criteria Current criteria Short-term Future areas of research


AECC Berlin possible revisions
1994 2012 2022+ Onwards
Timing Acute, not specified New or worsening within 7 days

Chest Bilateral infiltrates on chest Bilateral infiltrates on Ultrasound


Data science
imaging radiograph chest radiograph (or CT)

Oxygenation PaO2/FiO2 PaO2/FiO2 SpO2/FiO2 ratio


Acute lung injury <300 mm Hg Mild >200 mm Hg to ≤300 mm Hg Biomarker
ARDS <200 mm Hg Moderate >100 mm Hg to ≤200 mm Hg development
Severe ≤100 mm Hg Treatable
traits
PEEP Not specfied Minimum PEEP 5 cm H2O (continuous High-flow nasal Integrated
positive airway pressure in mild oxygen, no multiomics
ARDS) requirement for
minimum PEEP
Point-of-care development
Origin of oedema Pulmonary artery wedge Not fully explained by cardiac failure
pressure ≤17 mm Hg

Biological markers None None

Figure 1: Timeline of ARDS criteria and future directions


The first consensus ARDS criteria were the AECC in 1994, followed by the Berlin Consensus Criteria in 2012. ARDS criteria are being revised and potential revisions are
illustrated. The future of ARDS in the era of precision medicine strives towards identifying treatable traits. AEEC=American–European Consensus Criteria. PaO2=partial
pressure of arterial oxygen. FiO2=fraction of inspired oxygen. SpO2=oxygen saturation. ARDS=acute respiratory distress syndrome. PEEP=positive end expiratory
pressure.

oximetry, and fraction of inspired oxygen (SpO2/FiO2) is hypoxaemic respiratory failure and bilateral infiltrates
an attractive alternative to a ratio between partial pressure cannot meet the definition of ARDS, despite having
of arterial oxygen and FiO2 (PaO2/FiO2) ratio due to its similar biological characteristics to patients with these
availability and safety. These simple bedside tools could be characteristics on positive-pressure ventilation. Given
useful in resource-limited settings, and outside the the increasing use of high-flow nasal oxygen, which
traditional intensive care unit.26–28 Evidence from might deliver low levels of PEEP as one of its physiological
retrospective analysis supports the ability of the SpO2/FiO2 benefits,34 there is interest in including high-flow nasal
ratio to predict outcomes in patients with ARDS.26 In oxygen in future definitions of ARDS.35 A modification to
resource-limited settings, in which mechanical ventilation, include high-flow nasal oxygen in the ARDS definition
blood gas analysis, and chest imaging might not be would allow early identification of patients with ARDS
available, the Kigali modification using the SpO2/FiO2 and facilitate recruitment to clinical trials at an earlier
ratio and lung ultrasound has been suggested and has timepoint in the clinical course of ARDS. There are
been useful to evaluate ARDS.29,30 Additionally the Kigali potential limitations of this modification. For instance, in
modification removes the requirement for positive end a single centre prospective study of 148 patients,
expiratory pressure (PEEP) as ventilator resources might PaO2/FiO2 was found to vary substantially after a change
not be available.29 in respiratory support from high-flow nasal oxygen to
Lung ultrasound and SpO2/FiO2 ratio have limitations invasive ventilation.36 Furthermore, Ranieri and
as diagnostic tools in ARDS. Lung ultrasound could colleagues37 showed that patients fulfilling ARDS criteria
overestimate ARDS. Vercesi and colleagues31 reported a on high-flow nasal oxygen who transition to invasive
high rate of false positives when comparing the Kigali mechanical ventilation might have lower mortality rates
modification with the Berlin criteria for ARDS in a than patients on non-invasive ventilation who transition
single centre observational study in the Netherlands. to invasive mechanical ventilation.
The high false positive rate was attributed to the No ARDS biomarker is recommended in clinical
sensitivity of lung ultrasound in detecting interstitial practice. The Berlin Task Force considered biomarkers
infiltrates and consolidative changes.32 In addition, pulse for inclusion in the previous ARDS revision but found
oximetry could cause disparities in the identification of they lacked sensitivity and specificity as a diagnostic tool.2
occult hypoxaemia due to skin colour.33 Further Bos and colleagues38 have recently reviewed potential
prospective studies are required to determine optimal biomarkers, which could inform the diagnostic criteria of
SpO2/FiO2 thresholds for severity, which should account ARDS. Markers of endothelial or epithelial injury, protein
for differences in race and ethnic origin. rich pulmonary oedema, and systemic or alveolar host
The requirement for positive-pressure ventilation response could be considered. A novel approach to assess
means many patients with non-cardiogenic acute for the presence of protein rich pulmonary oedema

www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8 3


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

The Intensive Care Society The French Intensive Care The American Thoracic Society, WHO living guideline
and the Faculty of Intensive Society50 European Society of Intensive (COVID-19 ARDS)14
Care Medicine49 Care Medicine, and the Society
of Critical Care Medicine51
Non-invasive ventilation ·· ·· ·· Conditional recommendation
in mild ARDS
Lung protective ventilation Recommended Recommended Recommended Recommended
Prone positioning Recommended in moderate- Recommended PaO2/FiO2 Recommended in severe ARDS Recommended PaO2/FiO2 ratio
to-severe ARDS ratio <150 mm Hg. <150 mm Hg
High positive end Recommended in moderate- Recommended in moderate- Recommended in moderate-to- Conditional recommendation
expiratory pressure to-severe ARDS. to-severe ARDS severe ARDS for moderate-to-severeARDS
strategy
Driving pressure ·· No recommendation due to Research recommendation Consider driving pressure as
insufficient evidence part of an individualised
positive end expiratory
pressure titration strategy
Spontaneous ventilation ·· No recommendation due to Research recommendation ··
insufficient evidence
Recruitment manoeuvres ·· Not recommended Not routinely recommended ··
High-frequency oscillatory Not recommended Not recommended Not recommended ··
ventilation
Extracorporeal membrane Recommended in severe Recommended when Research recommendation Conditional recommendation
oxygenation ARDS PaO2/FiO2 ratio is <80 mm Hg for when PaO2/FiO2 ratio is
or lung protective ventilation <80 mm Hg despite lung
is not possible protective ventilation
Extracorporeal carbon Research recommendation No recommendation Research recommendation ··
dioxide removal due to insufficient evidence
Conservative fluid strategy Recommended ·· ·· Recommended
Neuromuscular blockade Recommended in early Recommended in early ARDS ·· Not routinely recommended
moderate to severe ARDS with a PaO2/FiO2 ratio of for all patients
<150 mm Hg
Inhaled vasodilators Not recommended Can be used when ·· ··
hypoxaemia persists despite
lung protective ventilation
and prone position, and
before extracorporeal
membrane oxygenation
Corticosteroids Research recommendation ·· ·· Recommended
Other pharmacological ·· ·· ·· IL-6 receptor blockers (eg,
agents tocilizumab or sarilumab) or
baricitinib (Janus kinase
inhibitor) is a strong
recommendation; monoclonal
antibodies (casirivimab and
imdevimab) is a conditional
recommendation for patients
who are seronegative
ARDS=acute respiratory distress syndrome. PaO2=partial pressure of arterial oxygen. FiO2=fraction of inspired oxygen.

Table: Guideline recommendations for ARDS management

includes evaluation of fluid from a heat moisture identified biological phenotypes that could have
exchange filter; however, this technique still requires differential treatment responsiveness. The PHIND study
validation with important outcomes in ARDS.39,40 (NCT04009330) aims to evaluate the ability of a point-of-
There is increasing recognition of the limitations of a care assay to prospectively identify these phenotypes at
syndromic definition, which ignores the substantial the bedside. ARDS phenotypes have also been identified
biological and physiological heterogeneity within ARDS, by machine learning models using routinely available
has driven a new paradigm focusing on the recognition clinical data,45 which could prove a useful tool to
of identifiable and treatable biological traits.41 As a result incorporate into electronic health systems to phenotype
populations of patients could be identified by treatable patients in real time. Biological phenotypes, along with
traits rather than a syndromic definition of ARDS in the emerging data from multiomic studies46 and
future. Pioneering work by Calfee and colleagues42–44 has immunophenotyping,47 might inform treatable traits in

4 www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

ARDS that could be incorporated into future ARDS


criteria. The UK National Institute of Health has recently Inflammasone activation Epithelial injury Inflammation

issued a call for funding applications to form a IL-18, IL-1beta, and Ferritin sRAGE and SP-D IL-6 and sTNFR1
IL-1 inhibitors Solnatide Corticosteroids
collaborative ARDS, Pneumonia, and Sepsis Phenotyping Mesenchymal stromal cells
NLRP3 inhibitors
Consortium to understand the heterogeneity and Disulfiram Simvastatin
underlying mechanisms of critical illness. This initiative IL-6 inhibitors
Epithelial
is an important step towards precision medicine for Inflammatory damage
pulmonary oedema Platelet activation
ARDS. These biological phenotypes and treatable traits
also might be present beyond ARDS and be common to Cytokine
Neutrophil
release
other clinical syndromes seen in patients who are recruitment
critically ill.48 Activated BALF neutrophils
Migrating Neutrophil
neutrophil
neutrophil extracellular traps
ARDS management formation
Cellular
The table summarises evidence-based guidelines for the debris Cathepsin S
inhibitor
management of ARDS published by the UK Faculty of Inflammasome
Ulinastatin
activation
Intensive Care Medicine and Intensive Care Society,49 the
French Intensive Care Society,50 jointly by the American
Thoracic Society, European Society of Intensive Care Inflamed
Medicine, and the Society of Critical Care Medicine,51 and Mitochondrial Mitochondrial endothelial cells Endothelial
dysfunction dysfunction injury
WHO living guidelines for ARDS due to COVID-19.14 A Mitochondrial DNA and reactive
Gap formation
Ang-2
recent report by an expert panel in the UK suggested that Microthrombi
Mesenchymal oxygen species
formation Recombinant ACE2
supportive management of patients with ARDS due to stromal cells Citrulline
COVID-19 should follow existing evidence-based ARDS
guidelines.52 Here we consider the evidence regarding Figure 2: Potential treatable traits in ARDS, a pathophysiological model
ventilation strategies, prone positioning, extracorporeal Treatable traits could be identified by biomarkers (in italics), which align with underlying pathophysiological
mechanisms and could be targeted by specific therapeutics or interventions. Biomarkers illustrated are biological
support, neuromuscular blockade, and corticosteroids in markers (ie, obtained from biological samples). Alternative biomarkers might also include imaging, physiology,
the management of ARDS. Future directions for and clinical data, when they reflect an underlying pathophysiological process that could be responsive to therapy.
therapeutic interventions in ARDS are also discussed The pathophysiological model illustrated here probably does not account for the complexities of interactions
(figure 2). between pathophysiological mechanisms and individual patient responses. Integration of multiple modalities of
information (eg, clinical features, imaging, physiology, biological tests, and multiomics data) might delineate
further subphenotypes that could more reliably predict responsiveness to therapies and interventions.
Ventilation strategies BALF=bronchoalveolar lavage fluid.
Lung protective ventilation (ie, tidal volumes of <6 ml/kg,
predicted body weight, and plateau pressure ≤30 mm Hg) strategies and recruitment manoeuvres in moderate-to-
is a key recommendation based on the findings of the severe ARDS.60 The key finding of this analysis was that a
landmark ARMA trial,20 with reduced mortality and higher PEEP strategy without a lung recruitment
increased days free of ventilation. Randomised controlled manoeuvre was most likely to be beneficial compared
trials (RCTs) of novel ventilatory strategies have with a lower PEEP strategy (probability of mortality
continued to reinforce the benefit of lung protective benefit was 99%), whereas a prolonged recruitment
ventilation.53–55 Clinical trials investigating alternative manoeuvre with a higher PEEP strategy probably caused
ventilation strategies have not shown additional benefit harm compared with a higher PEEP strategy without a
compared with lung protective ventilation.56–58 Airway recruitment maneouvre (probability of increased
pressure release ventilation is an innovative pressure- mortality was 99%).60 Heterogeneity of individual patient
controlled mode of ventilation, which delivers a responses to PEEP strategies is recognised,61 and there is
prolonged high level of pressure with intermittent, time increasing interest in personalised PEEP strategies,
cycled pressure release to a low level of pressure.59 Meta- although to date these have not shown additional benefit
analysis of airway pressure release ventilation in acute over conventional PEEP strategies.62,63
hypoxaemic respiratory failure (330 patients in five Driving pressure (ie, plateau pressure minus end
studies, three of which were in patients with ARDS) expiratory pressure) might be an independent predictor
suggests a benefit in hospital mortality, ventilation free of survival in patients with ARDS.53 Amato and
days, and intensive care unit length of stay.59 However, colleagues53 showed that driving pressure was the key
studies conducted to date have had methodological mediator of the benefits of PEEP and tidal volume
limitations,59 and a robust clinical trial is required to strategies. An upper limit of 15 cm H2O for driving
address the role of airway pressure release ventilation in pressure is recommended, above which there is
ARDS. considerable lung stress64 and mortality could increase.22,53
Guideline recommendations reserve high PEEP for Conversely, two clinical trials have now found increased
patients with moderate-to-severe ARDS. A recent mortality in the setting of lower driving pressure (ie,
Bayesian network meta-analysis evaluated PEEP ≤15 cm H2O), suggesting driving pressure might not be

www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8 5


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

as useful as originally expected to predict mortality.63,65 areas of alveolar overdistension, thus ensuring more
Clinical trials that are underway to investigate PEEP homogeneous aeration of the lung and potentially
strategies targeted to driving pressure (including the reducing ventilator-induced lung injury.73 On the basis of
STAMINA trial NCT04972318) will provide important data from the PROSEVA multicentre RCT,74 patients who
data to inform this debate. are mechanically ventilated and have severe ARDS, defined
Another novel concept that has been applied in the as a PaO2/FiO2 ratio of less than 150 mm Hg with an FiO2
context of ARDS is mechanical power. Mechanical power of at least 0·6 and PEEP of at least 5 cm H2O after an initial
is the amount of energy transferred from a mechanical period of stabilisation for a minimum of 12 h, should be
ventilator to the respiratory system per unit of time and is ventilated in the prone position for at least 16 h daily until
determined by the combined effects of applied tidal clinical improvement. Prone ventilation should be
volume, driving pressure, respiratory rate, inspiratory flow, instituted early and ideally within 36 h of meeting these
and PEEP, as well as determinants of mechanical criteria. Notably, prone ventilation should be used
properties of the lung (eg, respiratory system elastance and alongside a lung protective ventilatory strategy and typically
airway resistance).66 Mechanical power can be calculated patients should be receiving a high PEEP strategy.74 Prone
as: Power (J/min)=0·098 × tidal volume × respiratory position was associated with an improvement in mortality
rate × [PEEP + (0·5 × driving pressure) + (peak pressure– at day 28 (16% vs 33%, p<0·0001), which persisted at day 90
plateau pressure)].54 Mechanical power might be a better (24% vs 41%, p<0·0001).74 Cumulative evidence from
driver of lung protective ventilation, compared with clinical trials done before PROSEVA does not support the
individual ventilator parameters, as it considers the universal application of prone position for patients with
balance of these parameters as a whole.66 Reanalyses of less severe ARDS.49,51 Despite evidence of benefit in the
clinical trial and observational data have shown mechanical PROSEVA trial, the APRONET75 prospective international
power is associated with mortality outcomes.54,67–70 In a observational prevalence study found only a third of
retrospective analysis, including 8207 patients, a consistent patients with severe ARDS were treated in the prone
increase in the risk of death was shown with mechanical position. The low incidence of prone position could in part
power greater than 17 J/min.69 The complexity of be explained by concerns regarding adverse events such as
interpreting mechanical power limits its clinical use, endotracheal tube obstruction, pressure sores, and loss of
although recently Costa and colleagues54 found the venous access.51 In the setting of COVID-19 ARDS, prone
component variables of mechanical power most predictive positioning has been safely and widely implemented for
of mortality were in fact the driving pressure and patients who are ventilated.7
respiratory rate. As these variables can be easily measured In the setting of COVID-19, the use of prone positioning
at the bedside, the additional benefit of mechanical power has been extended to awake patients who are unintubated,
remains uncertain. but there remains uncertainty with conflicting findings
Heterogeneity of treatment effect is apparent in studies from recent trials.76,77 The duration of prone position in
investigating ventilatory strategies in ARDS and suggesting these studies has been shorter than that found to be
there could be phenotypes (or treatable traits) to direct beneficial in patients who are sedated and paralysed
personalised ventilatory strategies towards. Costa and receiving invasive mechanical ventilation, and was often
colleagues54 found patients with low respiratory system limited by patient comfort.76,77 There is no evidence to
compliance might be more likely to benefit from low tidal inform the use of prone positioning in awake patients who
volumes and low driving pressures, whereas patients with are unintubated with acute hypoxaemic respiratory failure
high compliance were predicted to benefit more from low not due to COVID-19 and a clinical trial is required to
respiratory rates. In a re-analysis of the EPVent-2 trial answer this question. The role of prone position as an
investigating oesophageal pressure-guided PEEP in ARDS adjunct to extracorporeal membrane oxygenation (ECMO)
there was a differential effect on mortality dependent on therapy also remains uncertain and is the subject of
disease severity as determined by the APACHE-II score.71 ongoing clinical trials (NCT04139733 and NCT04607551).
Hyperinflammatory and hypo­
inflammatory
subphenotypes of ARDS have also been reported to Neuromuscular blockade
respond differentially to PEEP strategies.42 The findings Previous guidelines made a weak recommendation for
from the LIVE trial,72 which investigated personalised early neuromuscular blockade in patients with
mechanical ventilation tailored to lung morphology in moderate-to-severe ARDS, although methodological
patients with ARDS, highlight the need to align phenotypes limitations in the available evidence were noted.49,50
with personalised ventilation strategies. In this study, in More recently, the ROSE (Early Neuromuscular
patients who received a ventilator strategy misaligned with Blockade for ARDS) trial78 investigated the role of
lung morphology, mortality was substantially increased.72 neuromuscular blockade with deep sedation compared
with usual care with light sedation in patients with
Prone positioning moderate-to-severe ARDS. The trial was stopped for
Prone positioning in patients with ARDS improves futility (after enrolling 1006 patients) and showed no
oxygenation, increases recruitment potential, and reduces difference between groups in 90-day mortality.78 On the

6 www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

basis of this evidence, the routine use of continuous ECMO use in a cohort of 7345 patients with COVID-19
neuromuscular blockade is not recommended in an across five countries.85 ECMO therapy was delivered in
unselected population of patients with ARDS. The 844 patients, and an analysis including patients with a
ROSE trial78 did not specifically evaluate ventilator PaO2/FiO2 ratio of less than 80 mm Hg indicated ECMO
dyssynchrony, and 17% of patients in the control group was associated with reduced mortality compared with
received neuromuscular blockade during the first 48 h, conventional therapy (RR 0·78, 95% CI 0·75 to 0·82).85
so there could be patients who still benefit from neuro­ Although confirmation in an RCT would be desirable,
muscular blockade to manage ventilator dyssynchrony. these findings provide reassurance regarding the use of
Furthermore, the number of patients receiving prone ECMO in a selected population of patients with severe
position and ECMO in the ROSE trial was low, and COVID-19. Extracorporeal carbon dioxide removal has
whether neuromuscular blockade interacts to facilitate been of interest in research settings to facilitate low-tidal-
the benefit associated with these interventions is volume ventilation.65 The REST trial,65 which investigated
unknown. extracorporeal carbon dioxide removal in patients with
acute hypoxaemic respiratory failure found no difference
Extracorporeal support in 90-day mortality, and there was an increased incidence
In the era of lung protective ventilation, two RCTs have of serious adverse events, including clinically important
investigated the role of veno-venous ECMO for patients haemorrhage, in the extracorporeal carbon dioxide
with severe ARDS.79,80 The CESAR trial79 was a multicentre removal group. On this basis the use of extracorporeal
RCT that compared conventional management of ARDS carbon dioxide removal for the treatment of ARDS is not
with referral to an ECMO centre for consideration of the recommended outside RCTs.
treatment. Of patients randomised to referral for ECMO,
only 76% (68 of 90) received it. In the group randomised to Corticosteroids
ECMO referral, an improvement in the primary outcome The role of corticosteroids in ARDS management has
of quality-adjusted life years was seen at 6 months.79 The been a longstanding controversy.18 Steroids have potent
CESAR trial had substantial methodological limitations. anti-inflammatory effects that could be of benefit in
However, these limitations were addressed in the EOLIA ARDS. A substantial development during the COVID-19
trial,80 which was an international RCT comparing ECMO pandemic has been the benefits found with corticosteroid
to conventional treatment, with the option of ECMO as a therapy for patients with severe COVID-19.86–88
rescue therapy if required. Although the primary outcome Corticosteroids are now the standard of care for patients
of mortality included a potentially beneficial treatment with COVID-19 ARDS, and there has been renewed
effect (relative risk [RR] 0·76, 95% CI 0·55 to 1·04, p=0·09), interest in their role in non-COVID-19 ARDS. Before the
it did not achieve statistical significance.80 A post-hoc COVID-19 pandemic, numerous studies had investigated
Bayesian analysis showed a high probability that early the role of corticosteroids in ARDS.89 Unfortunately
ECMO was of benefit.81 Furthermore, in a subsequent steroid regimes differed between studies (eg, different
individual patient data meta-analysis including both the types, doses, and duration) and there were differences in
CESAR and EOLIA RCTs, the precision of the treatment the patient populations investigated (eg, early vs late
effect was improved (combined data for 429 patients) and a ARDS and some studies were done before lung protective
statistically significant benefit in mortality at day 90 was ventilation). Most recently, the DEXA-ARDS trial90
shown in the ECMO group (RR 0·75, 95% CI 0·6 to 0·94, investigated a high dose (ie, 20 mg once daily for 5 days)
p=0·013).82,83 Together, on the basis of these data, patients followed by a lower dose (ie, 10 mg once daily for 5 days)
with severe ARDS (defined as a PaO2/FiO2 ratio of of dexamethasone in patients with moderate-to-severe
<50 mm Hg for >3 h, a PaO2/FiO2 ratio of <80 mm Hg for ARDS. In the dexamethasone group there was an increase
>6 h, or severe hypercapnic respiratory failure [ie, a pH in ventilator free days (between group difference 4·8 days,
of <7·25 with a PaCO2 ≥60 mm Hg for >6 h]) are 95% CI 2·57 to 7·03, p<0·0001) and a reduced 60-day
recommended to be treated with ECMO. Notably, patients mortality (21% vs 36%, between-group difference –15·3%,
receiving ECMO should receive an overall management 95% CI –25·9 to –4·9, p=0·0047). A subsequent meta-
strategy similar to that used in the EOLIA trial. The analysis (999 patients from eight RCTs with
provision of ECMO is complex and organisational non-COVID-19 ARDS) found a mortality benefit in favour
characteristics of ECMO centres should be consistent with of corticosteroid use (RR 0·71, 95% CI 0·54 to 0·92).89
the organisations that delivered it in the EOLIA trial or There was evidence of an association with hyperglycaemia,
comply with the criteria for ECMO centres defined by but no certain evidence supported concerns regarding
expert groups.84 other adverse events including neuromuscular weakness,
Evidence of benefit of ECMO has been extrapolated to a gastrointestinal bleeding, or infection. Notably, however,
COVID-19 population, and ECMO has been delivered an early trial investigating methylprednisolone in
with reported mortality rates similar to that of a general persistent ARDS found an association with increased risk
population of patients with non-COVID-19 ARDS.83 A of late mortality (ie, day 60 and day 180) when steroids
recent large comparative effectiveness study showed were initiated beyond day 14 of ARDS onset.91

www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8 7


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

Misestimation of predicted control event rates and background, the I-SPY COVID-19 trial114 established a
treatment effects (used to inform sample size phase 2 platform trial for the investigation of novel
calculations) is increasingly recognised as common in therapeutics in COVID-19. To capitalise on these
ARDS RCTs, and might have contributed to the developments, it is important to start an international
uncertainty of evidence related to corticosteroids in precision medicine platform phase 2 trial, which can
ARDS.92 Using Bayesian methods, Saha and colleagues92 incorporate treatable traits, to test new therapies for
showed the strength of evidence for corticosteroids in patients with ARDS. Collaboration between the global
ARDS favours benefit and supports the prioritisation of scientific community (including experts in ARDS,
future clinical trials investigating corticosteroids in precision medicine, and adaptive trial design) will be
patients with non-COVID-19 ARDS. needed to drive this research agenda for precision
medicine in ARDS forward.115
Future directions for therapeutic interventions
in ARDS Outcomes in patients with ARDS
Many clinical trials investigating pharmacological The LUNG SAFE study22 was the largest observational
interventions in patients with ARDS have not shown study of the epidemiology, patterns of care, and clinical
benefit. Specific pharmacological agents that have not outcomes of ARDS, done in a cohort of 29 144 patients
shown benefit have included inhaled prostaglandins,93 admitted to 459 intensive care units from 50 countries
statins,94,95 aspirin,96 surfactant,97 activated protein-C,98,99 across five continents. In the LUNG SAFE study, 23% of
and Sivelestat.100 Other agents including β-2 agonists101,102 patients requiring mechanical ventilation had ARDS.
and keratinocyte growth factor,103 which had promising Patients with ARDS had a median duration of ventilation
preclinical data, had potential for harm. of 8 days, an intensive care unit stay of 10 days, and a
One novel therapeutic of interest in ARDS is hospital stay of 17 days. 28-day mortality was 35% and
mesenchymal stromal cells. These multipotent, plastic increased with severity of hypoxia, with rates greater than
adherent cells can be derived from multiple sources 40% in patients with severe ARDS. Considering geo-
including bone marrow, umbilical cord, or adipose tissue economic variation, PROVENT-iMiC116 (an international
and have pleiotropic immunomodulatory, reparative, and multicentre prospective study of patients who were
antimicrobial actions.104 RCTs investigating mesenchymal invasively ventilated in intensive care units from 10 Asian
stromal cells in ARDS and COVID-19 have supported middle income countries) showed a lower proportion of
their safety, however efficacy has not yet been patients had ARDS at the initiation of ventilation (7%)
established.105–109 Although attractive in targeting multiple compared with the LUNG SAFE study.22 Ventilation
therapeutic pathways in ARDS, mesenchymal stromal practices were similar, but there were differences in
cells are known to respond to their biological outcomes including a higher mortality rate (45%), shorter
microenvironment and could also be subject to duration of ventilation (median 4 days), and shorter
heterogeneous treatment effects in different patient duration of intensive care unit stay (median 5 days) than
phenotypes.110 in the LUNG SAFE study.116 Disparities could relate to
The interest in biological phenotypes and treatable geoeconomic variation in the case mix, or might relate to
traits in patients with ARDS, which might respond resource availability. Similarly, excess mortality in
differently to therapeutic interventions, has been patients with ARDS has been shown in other low-income
supported by promising findings from re-analyses of and middle-income countries.117,118
previous clinical trial datasets. In a re-analysis of the Outcomes of patients in clinical trials provide another
HARP-2 (simvastatin for ARDS) trial43 patients with a perspective on ARDS outcomes. A systematic review of
hyperinflammatory phenotype treated with simvastatin 28-day mortality in control groups of ARDS clinical trials
were found to have significantly improved 28-day showed a mortality rate of 29% (from 26 trials with
survival. 2766 patients between 2016–20).119 Discrepancies between
Figure 2 illustrates how therapeutic interventions outcomes for patients with ARDS in RCTs and
could align with treatable traits in this future era of observational studies are recognised; RCTs are more
precision medicine for ARDS. There is a need for restrictive in their patient selection, and lung protective
continued translational research to identify novel ventilation is usually more rigidly implemented.120
treatable traits and therapeutic agents targeting these Furthermore, heterogeneity of outcomes shown in clinical
traits.111–113 There might also be a role for a personalised trials occurs (eg, Juschten and colleagues121 showed 28-day
approach to existing interventions in ARDS, and existing mortality ranging from 10% to 67% in a systematic review
datasets could prove useful to identify populations that of 67 RCTs between 2000 and 2019), and this might limit
are most responsive. the generalisability of trial findings to the clinical setting.
Alongside the recent identification of biological Although data from a pre-COVID-19 era show few
phenotypes and treatable traits, the benefits of adaptive patients with ARDS die from irreversible respiratory
platform trials in establishing effective therapeutics for failure (estimates vary depending on definition but are
COVID-19 have been clearly shown. Building on this shown to be between <1% and 9%),122,123 ARDS has a

8 www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

direct and measurable effect on patient mortality.124 Kamdar and colleagues139,140 have shown that 44% of
Compared with patients in intensive care units who do previously employed survivors of ARDS were jobless at
not have ARDS, ARDS increases the mortality rate 1 year, and at 5 years 30% had never returned to work. Su
by 15%.124 In the setting of ARDS related to sepsis, the and colleagues141 found that one in five survivors of ARDS
attributable mortality rate has been shown to be up who had returned to work were subsequently unable to
to 37%.125 In a retrospective cohort of 127 patients with sustain work. In another study, Brown and colleagues138
ARDS, pulmonary dysfunction was shown as a leading showed the percentage of survivors of ARDS living
cause of death in 28% (other reported causes included independently at home at 6 months reduced from a
sepsis in 29%, neurological dysfunction in 17%, and pre-existing baseline of 91% to 45% at 6 months. Use of
cardiac dysfunction in 10%).123 health-care resources is increased in survivors of ARDS,
Epidemiological trends and outcomes for patients with with hospital re-admissions reported in up to 40% of
ARDS are unlikely to remain static.126 In the setting of the survivors, up to a third of whom require re-admission to
COVID-19 pandemic, estimates of ARDS incidence have intensive care units.136,142 Survivors of ARDS already have
been considerably greater than previously reported, and a high risk of mortality during their acute illness, but the
outcomes have varied over time, and between risk of mortality persists in the long term. At 1 year,
settings.127–129 Ethnic and racial disparities in the mortality rates of 11% have been reported, increasing
epidemiology and outcomes of COVID-19 have been to 20–34% at 5 years.133,143 In a comparison of patients
apparent.130,131 Whether there has been practice creep with with acute hypoxaemic respiratory failure (many of
extrapolation of therapies proven to be of benefit in the whom are most likely to have ARDS) to matched non-
setting of COVID-19 to non-COVID-19 ARDS, even when hospitalised adults, patients with acute hypoxaemic
an evidence gap for these therapies exists in other causes respiratory failure had a 1·9 fold increase in late
of acute respiratory failure, also remains unclear. mortality.144 The risk of late mortality was primarily
Furthermore, COVID-19 has had an immeasurable attributable to the acute inciting event, although
impact on health-care systems, particularly the delivery approximately 30% of the mortality risk was associated
of respiratory and critical care services.132 Such practice with hypoxaemic respiratory failure.144
change could alter the epidemiology and outcomes of Reports of longer-term outcomes of survivors of
ARDS and future large population-based observational ARDS with COVID-19 are emerging and provide
studies of ARDS will be required to inform these evidence of persistent physical, mental, and cognitive
uncertainties in the post-COVID-19 era. deficits.15,16,145–148 Evidence of persistent interstitial lung
disease in patients with ARDS related to COVID-19 who
Sequelae of ARDS were mechanically ventilated has been reported, but
New or worsening problems in physical, cognitive, or there remains uncertainty as to how relevant this
mental health status is common in survivors of ARDS. evidence is to a population of patients with non-
Herein, we discuss evidence related to long-term COVID-19 ARDS.10,149,150 Of note, some studies have
sequelae for non-COVID-19 ARDS, followed by emerging shown better health-related quality of life,16 and higher
evidence in COVID-19 ARDS. Physical features return to work rates in patients with COVID-19 ARDS
commonly seen in survivors of ARDS without COVID-19 compared with a non-COVID-19 ARDS population.15,16,148
include respiratory symptoms (eg, dyspnoea, cough, and However, patients with COVID-19 ARDS self-report
sputum production) and reduced exercise capacity.133 lower disability and higher health-related quality of life
Respiratory symptoms do not usually correlate with the before intensive care unit hospitalisation and therefore
degree of impairment of pulmonary function or with the might have a greater capacity for improvement following
extent of radiological abnormalities.134 Survivors of ARDS critical illness than patients with non-COVID-19
continue to experience a spectrum of physical disorders ARDS.15 When adjusted for potential confounders at
related to sequelae of their critical illness (eg, tracheal baseline, Hodgson and colleagues15 show the incidence
stenosis, vocal cord dysfunction, dental damage, and of new disability, health-related quality of life,
scarring related to interventions).133 The prevalence of psychological function, and cognitive function was
post-traumatic stress disorder is high in survivors of similar at 6 months between patients who are critically
ARDS, with reported rates of up to one in four patients at ill with and without COVID-19. The emerging effect of
8 years.135 Persistent psychiatric symptoms of depression long COVID,151 and long-term effects of COVID-19 on
and anxiety are reported in up to half of patients.133,135 organ function (including interstitial lung disease10,149,150
Survivors of ARDS have been shown to have impairments and a variety of cardiac sequelae152) raises concerns
of cognitive function, which include executive function, regarding the full spectrum of morbidity in survivors of
verbal reasoning, memory, and attention deficits.136 ARDS related to COVID-19.
Given the burden of physical, mental, and cognitive Identification of interventions, both during critical care
symptoms experienced by survivors of ARDS, there is and after critical care, which improve recovery from
considerable decline in quality of life, employment, critical illness has been recognised as a priority by
societal participation, and residential status.133,136–140 patients and their caregivers.153 In a survey of UK hospital

www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8 9


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

sites delivering intensive care services, 74% provided consultancy fees from INSMED, unrelated to this work, and fees for
outpatient follow-up services for survivors of critical care participation in grant panels for the Californian Institute of Regenerative
Medicine, unrelated to this Series paper. CMO’K declares a spouse who
and 18% provided physical rehabilitation programmes.154 has received grants from Wellcome Trust, the Northern Ireland Health
Internationally, reports of follow-up after discharge from and Social Care Research and Development Fund, NIHR, Innovate UK,
intensive care unit vary considerably and there is no the Medical Research Council, and Novavax; consultancy fees unrelated
consensus approach on how these services should be to this work from Bayer, GlaxoSmithKline, Boehringer Ingelheim,
Novartis, and Eli Lilly; payments from GlaxoSmithKline as an educational
configured.155,156 Survivors from intensive care units have seminar speaker; is a member of the data and safety monitoring board
reported benefits of intensive care unit recovery for Vir Biotechnology, Inc, and Faron Pharmaceuticals; has a patent for a
programmes and peer support,157,158 but to date there are novel treatment for an inflammatory disease; is a director of research for
no evidence-based interventions to improve long-term the Intensive Care Society and Director of the Efficacy and Mechanism
Evaluation programme for the Medical Research Council and NIHR.
outcomes for patients with ARDS.159,160 Critical care
survivorship remains an area of active research with Acknowledgments
Figures included in this manuscript were created with BioRender.
ongoing clinical trials investigating potential
interventions occuring post-intensive care unit stay, and References
1 Ranieri VM, Rubenfeld GD, Thompson BT, et al. Acute respiratory
the mode of delivery in terms of face-to-face or virtual distress syndrome: the Berlin definition. JAMA 2012; 307: 2526–33.
approaches to provide programmes to support recovery. 2 Ferguson ND, Fan E, Camporota L, et al. The Berlin definition of
Crucially, future clinical trials in ARDS should also ARDS: an expanded rationale, justification, and supplementary
material. Intensive Care Med 2012; 38: 1573–82.
include multidimensional long-term outcomes. 3 Shaver CM, Bastarache JA. Clinical and biological heterogeneity in
acute respiratory distress syndrome: direct versus indirect lung
Conclusion injury. Clin Chest Med 2014; 35: 639–53.
4 Huppert LA, Matthay MA, Ware LB. Pathogenesis of acute respiratory
ARDS is a clinical syndrome defined by acute hypoxaemic distress syndrome. Semin Respir Crit Care Med 2019; 40: 31–39.
respiratory failure with bilateral infiltrates on chest 5 Thille AW, Esteban A, Fernández-Segoviano P, et al. Comparison of
imaging. Entering the post-pandemic era, there is the Berlin definition for acute respiratory distress syndrome with
uncertainty regarding the epidemiological landscape of autopsy. Am J Respir Crit Care Med 2013; 187: 761–67.
6 Brown SM, Peltan ID, Barkauskas C, et al. What does acute
ARDS; however, patients with ARDS continue to have respiratory distress syndrome mean during the COVID-19
considerable morbidity and mortality in both the short pandemic? Ann Am Thorac Soc 2021; 18: 1948–50.
and long term. ARDS criteria are under revision, and 7 Ferrando C, Suarez-Sipmann F, Mellado-Artigas R, et al. Clinical
features, ventilatory management, and outcome of ARDS caused by
limitations that could be addressed have been considered. COVID-19 are similar to other causes of ARDS. Intensive Care Med
The evidence base for optimal supportive care and 2020; 46: 2200–11.
interventions in ARDS continues to evolve to address 8 Brault C, Zerbib Y, Kontar L, et al. COVID-19- versus non-COVID-
19-related acute respiratory distress syndrome: differences and
areas of uncertainty. As we enter an era of precision similarities. Am J Respir Crit Care Med 2020; 202: 1301–04.
medicine in critical illness, the future of ARDS 9 Bain W, Yang H, Shah FA, et al. COVID-19 versus non-COVID-19
management strives towards identification of biological acute respiratory distress syndrome: comparison of demographics,
physiologic parameters, inflammatory biomarkers, and clinical
phenotypes and treatable traits and delivery of outcomes. Ann Am Thorac Soc 2021; 18: 1202–10.
personalised therapeutic interventions. 10 Vijayakumar B, Tonkin J, Devaraj A, et al. CT lung abnormalities
Contributors after COVID-19 at 3 months and 1 year after hospital discharge.
EAG prepared the first draft of the manuscript and figures. DFM and Radiology 2022; 303: 444–54.
CMO’K reviewed and contributed to subsequent versions of the 11 Sjoding MW, Admon AJ, Saha AK, et al. Comparing clinical
manuscript. All authors have reviewed and approved the final version. features and outcomes in mechanically ventilated patients with
COVID-19 and acute respiratory distress syndrome.
Declaration of interests Ann Am Thorac Soc 2021; 18: 1876–85.
EAG declares funding from the Wellcome Trust for work on a study 12 Sinha P, Calfee CS, Cherian S, et al. Prevalence of phenotypes of
investigating acute respiratory distress syndrome (ARDS) and COVID-19 acute respiratory distress syndrome in critically ill patients with
and scholarship support from the American Thoracic Society to attend an COVID-19: a prospective observational study. Lancet Respir Med
American Thoracic Society meeting. DFM declares grants as an 2020; 8: 1209–18.
investigator in ARDS and COVID-19 studies from Wellcome Trust, the 13 Cui N, Jiang C, Yang C, Zhang L, Feng X. Comparison of deep vein
National Institute for Health and Care Research (NIHR), Innovate UK, thrombosis risks in acute respiratory distress syndrome caused by
the Medical Research Council, Novavax, and the Northern Ireland Health COVID-19 and bacterial pneumonia: a retrospective cohort study.
and Social Care Research and Development Fund; consultancy fees Thromb J 2022; 20: 27.
unrelated to this work from Bayer, GlaxoSmithKline, 14 WHO. Living guidance for clinical management of COVID-19.
Boehringer Ingelheim, Novartis, and Eli Lilly; and payments from Nov 23, 2021. https://www.who.int/publications/i/item/WHO-2019-
nCoV-clinical-2021-2 (accessed July 7, 2022).
GlaxoSmithKline as an educational seminar speaker. DFM is a member
of the data and safety monitoring board for Vir Biotechnology and Faron 15 Hodgson CL, Higgins AM, Bailey MJ, et al. Comparison of 6-month
outcomes of survivors of COVID-19 versus non-COVID-19 critical
Pharmaceuticals; has a patent for a novel treatment for an inflammatory
illness. Am J Respir Crit Care Med 2022; 205: 1159–68.
disease; is a director of research for the Intensive Care Society and
16 Latronico N, Peli E, Calza S, et al. Physical, cognitive and mental
director of the Efficacy and Mechanism Evaluation programme for the
health outcomes in 1-year survivors of COVID-19-associated ARDS.
Medical Research Council and NIHR; declares a spouse who has received Thorax 2022; 77: 300–03.
grants from Wellcome Trust and the Northern Ireland Health and Social
17 Bossuyt PM, Reitsma JB, Linnet K, Moons KG. Beyond diagnostic
Care Research and Development Fund; consultancy fees from INSMED accuracy: the clinical utility of diagnostic tests. Clin Chem 2012;
and from the California Institute for Regenerative Medicine, unrelated to 58: 1636–43.
this Series paper. CMO’K declares grants as an investigator on studies 18 Ashbaugh DG, Bigelow DB, Petty TL, Levine BE. Acute respiratory
related to ARDS and COVID-19 from Wellcome Trust and the Northern distress in adults. Lancet 1967; 2: 319–23.
Ireland Health and Social Care Research and Development Fund;

10 www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

19 Bernard GR, Artigas A, Brigham KL, et al. The American-European 40 McNeil JB, Shaver CM, Kerchberger VE, et al. Novel method for
Consensus Conference on ARDS. Definitions, mechanisms, noninvasive sampling of the distal airspace in acute respiratory
relevant outcomes, and clinical trial coordination. distress syndrome. Am J Respir Crit Care Med 2018; 197: 1027–35.
Am J Respir Crit Care Med 1994; 149: 818–24. 41 Maslove DM, Tang B, Shankar-Hari M, et al. Redefining critical
20 Brower RG, Matthay MA, Morris A, Schoenfeld D, illness. Nat Med 2022; 28: 1141–48.
Taylor Thompson B, Wheeler A. Ventilation with lower tidal 42 Calfee CS, Delucchi K, Parsons PE, Thompson BT, Ware LB,
volumes as compared with traditional tidal volumes for acute lung Matthay MA. Subphenotypes in acute respiratory distress syndrome:
injury and the acute respiratory distress syndrome. N Engl J Med latent class analysis of data from two randomised controlled trials.
2000; 342: 1301–08. Lancet Respir Med 2014; 2: 611–20.
21 Pham T, Pesenti A, Bellani G, et al. Outcome of acute hypoxaemic 43 Calfee CS, Delucchi KL, Sinha P, et al. Acute respiratory distress
respiratory failure: insights from the LUNG SAFE Study. syndrome subphenotypes and differential response to simvastatin:
Eur Respir J 2021; 57: 2003317. secondary analysis of a randomised controlled trial. Lancet Respir Med
22 Bellani G, Laffey JG, Pham T, et al. Epidemiology, patterns of care, 2018; 6: 691–98.
and mortality for patients with acute respiratory distress syndrome 44 Famous KR, Delucchi K, Ware LB, et al. Acute respiratory distress
in intensive care units in 50 countries. JAMA 2016; 315: 788–800. syndrome subphenotypes respond differently to randomized fluid
23 Zimatore C, Pisani L, Lippolis V, et al. Accuracy of the radiographic management strategy. Am J Respir Crit Care Med 2017;
assessment of lung edema score for the diagnosis of ARDS. 195: 331–38.
Front Physiol 2021; 12: 672823. 45 Maddali MV, Churpek M, Pham T, et al. Validation and utility of
24 Mason SE, Dieffenbach PB, Englert JA, et al. Semi-quantitative ARDS subphenotypes identified by machine-learning models using
visual assessment of chest radiography is associated with clinical clinical data: an observational, multicohort, retrospective analysis.
outcomes in critically ill patients. Respir Res 2019; 20: 218. Lancet Respir Med 2022; 10: 367–77.
25 Sjoding MW, Taylor D, Motyka J, et al. Deep learning to detect 46 Du M, Garcia JGN, Christie JD, et al. Integrative omics provide
acute respiratory distress syndrome on chest radiographs: biological and clinical insights into acute respiratory distress
a retrospective study with external validation. Lancet Digit Health syndrome. Intensive Care Med 2021; 47: 761–71.
2021; 3: e340–48. 47 Roussel M, Ferrant J, Reizine F, et al. Comparative immune profiling
26 Leopold SJ, Ghose A, Plewes KA, et al. Point-of-care lung of acute respiratory distress syndrome patients with or without
ultrasound for the detection of pulmonary manifestations of SARS-CoV-2 infection. Cell Rep Med 2021; 2: 100291.
malaria and sepsis: an observational study. PLoS One 2018; 48 Reddy K, Calfee CS, McAuley DF. Acute respiratory distress
13: e0204832. syndrome subphenotypes beyond the syndrome: a step toward
27 Pisani L, De Nicolo A, Schiavone M, et al. Lung ultrasound for treatable traits? Am J Respir Crit Care Med 2021; 203: 1449–51.
detection of pulmonary complications in critically ill obstetric 49 Griffiths MJD, McAuley DF, Perkins GD, et al. Guidelines on the
patients in a resource-limited setting. Am J Trop Med Hyg 2020; management of acute respiratory distress syndrome.
104: 478–86. BMJ Open Respir Res 2019; 6: e000420.
28 Bass CM, Sajed DR, Adedipe AA, West TE. Pulmonary ultrasound 50 Papazian L, Aubron C, Brochard L, et al. Formal guidelines:
and pulse oximetry versus chest radiography and arterial blood gas management of acute respiratory distress syndrome.
analysis for the diagnosis of acute respiratory distress syndrome: Ann Intensive Care 2019; 9: 69.
a pilot study. Crit Care 2015; 19: 282. 51 Fan E, Del Sorbo L, Goligher EC, et al. An official American
29 Riviello ED, Kiviri W, Twagirumugabe T, et al. Hospital incidence Thoracic Society/European Society of Intensive Care Medicine/
and outcomes of the acute respiratory distress syndrome using the Society of Critical Care Medicine Clinical Practice guideline:
Kigali modification of the Berlin definition. mechanical ventilation in adult patients with acute respiratory
Am J Respir Crit Care Med 2016; 193: 52–59. distress syndrome. Am J Respir Crit Care Med 2017; 195: 1253–63.
30 Kwizera A, Kateete DP, Ssenyonga R, et al. Acute respiratory 52 Griffiths M, Meade S, Summers C, et al. RAND appropriateness
distress syndrome in an African intensive care unit setting: panel to determine the applicability of UK guidelines on the
a prospective study of prevalence and outcomes. Ann Am Thorac Soc management of acute respiratory distress syndrome (ARDS) and
2022; 19: 691–94. other strategies in the context of the COVID-19 pandemic. Thorax
31 Vercesi V, Pisani L, van Tongeren PSI, et al. External confirmation 2022; 77: 129–35.
and exploration of the Kigali modification for diagnosing moderate 53 Amato MBP, Meade MO, Slutsky AS, et al. Driving pressure and
or severe ARDS. Intensive Care Med 2018; 44: 523–24. survival in the acute respiratory distress syndrome. N Engl J Med
32 Xirouchaki N, Magkanas E, Vaporidi K, et al. Lung ultrasound in 2015; 372: 747–55.
critically ill patients: comparison with bedside chest radiography. 54 Costa ELV, Slutsky AS, Brochard LJ, et al. Ventilatory variables and
Intensive Care Med 2011; 37: 1488–93. mechanical power in patients with acute respiratory distress
33 Valbuena VSM, Seelye S, Sjoding MW, et al. Racial bias and syndrome. Am J Respir Crit Care Med 2021; 204: 303–11.
reproducibility in pulse oximetry among medical and surgical 55 Beitler JR, Walkey AJ. The staying power of pressure- and volume-
inpatients in general care in the Veterans Health Administration 2013- limited ventilation in acute respiratory distress syndrome.
19: multicenter, retrospective cohort study. BMJ 2022; 378: e069775. Am J Respir Crit Care Med 2021; 204: 247–49.
34 Parke RL, Bloch A, McGuinness SP. Effect of very-high-flow nasal 56 Hodgson CL, Cooper DJ, Arabi Y, et al. Maximal recruitment open
therapy on airway pressure and end-expiratory lung impedance in lung ventilation in acute respiratory distress syndrome (PHARLAP).
healthy volunteers. Respir Care 2015; 60: 1397–403. A phase II, multicenter randomized controlled clinical trial.
35 Matthay MA, Thompson BT, Ware LB. The Berlin definition of Am J Respir Crit Care Med 2019; 200: 1363–72.
acute respiratory distress syndrome: should patients receiving high- 57 Ferguson ND, Cook DJ, Guyatt GH, et al. High-frequency oscillation
flow nasal oxygen be included? Lancet Respir Med 2021; 9: 933–36. in early acute respiratory distress syndrome. N Engl J Med 2013;
36 Hultström M, Hellkvist O, Covaciu L, et al. Limitations of the ARDS 368: 795–805.
criteria during high-flow oxygen or non-invasive ventilation: 58 Young D, Lamb SE, Shah S, et al. High-frequency oscillation for
evidence from critically ill COVID-19 patients. Crit Care 2022; 26: 55. acute respiratory distress syndrome. N Engl J Med 2013;
37 Ranieri VM, Tonetti T, Navalesi P, et al. High-flow nasal oxygen for 368: 806–13.
severe hypoxemia: oxygenation response and outcome in patients 59 Carsetti A, Damiani E, Domizi R, et al. Airway pressure release
with COVID-19. Am J Respir Crit Care Med 2022; 205: 431–39. ventilation during acute hypoxemic respiratory failure: a systematic
38 Bos LDJ, Laffey JG, Ware LB, et al. Towards a biological definition review and meta-analysis of randomized controlled trials.
of ARDS: are treatable traits the solution? Intensive Care Med Exp Ann Intensive Care 2019; 9: 44.
2022; 10: 8. 60 Dianti J, Tisminetzky M, Ferreyro BL, et al. Association of positive
39 Bastarache JA, McNeil JB, Plosa EJ, et al. Standardization of end-expiratory pressure and lung recruitment selection strategies
methods for sampling the distal airspace in mechanically ventilated with mortality in acute respiratory distress syndrome: a systematic
patients using heat moisture exchange filter fluid. review and network meta-analysis. Am J Respir Crit Care Med 2022;
Am J Physiol Lung Cell Mol Physiol 2021; 320: L785–90. 205: 1300–10.

www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8 11


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

61 Goligher EC, Kavanagh BP, Rubenfeld GD, et al. Oxygenation 80 Combes A, Hajage D, Capellier G, et al. Extracorporeal membrane
response to positive end-expiratory pressure predicts mortality in oxygenation for severe acute respiratory distress syndrome.
acute respiratory distress syndrome. A secondary analysis of the N Engl J Med 2018; 378: 1965–75.
LOVS and ExPress trials. Am J Respir Crit Care Med 2014; 190: 70–76. 81 Goligher EC, Tomlinson G, Hajage D, et al. Extracorporeal membrane
62 Beitler JR, Sarge T, Banner-Goodspeed VM, et al. Effect of titrating oxygenation for severe acute respiratory distress syndrome and
positive end-expiratory pressure (PEEP) with an esophageal posterior probability of mortality benefit in a post hoc bayesian
pressure-guided strategy vs an empirical high PEEP-Fio2 strategy analysis of a randomized clinical trial. JAMA 2018; 320: 2251–59.
on death and days free from mechanical ventilation among patients 82 Combes A, Peek GJ, Hajage D, et al. ECMO for severe ARDS:
with acute respiratory distress syndrome: a randomized clinical systematic review and individual patient data meta-analysis.
trial. JAMA 2019; 321: 846–57. Intensive Care Med 2020; 46: 2048–57.
63 Cavalcanti AB, Suzumura ÉA, Laranjeira LN, et al. Effect of lung 83 Camporota L, Meadows C, Ledot S, et al. Consensus on the referral
recruitment and titrated positive end-expiratory pressure (PEEP) vs and admission of patients with severe respiratory failure to the
low PEEP on mortality in patients with acute respiratory distress NHS ECMO service. Lancet Respir Med 2021; 9: e16–17.
syndrome: a randomized clinical trial. JAMA 2017; 318: 1335–45. 84 Combes A, Brodie D, Bartlett R, et al. Position paper for the
64 Chiumello D, Carlesso E, Brioni M, Cressoni M. Airway driving organization of extracorporeal membrane oxygenation programs for
pressure and lung stress in ARDS patients. Crit Care 2016; 20: 276. acute respiratory failure in adult patients. Am J Respir Crit Care Med
65 McNamee JJ, Gillies MA, Barrett NA, et al. Effect of lower tidal 2014; 190: 488–96.
volume ventilation facilitated by extracorporeal carbon dioxide 85 Urner M, Barnett AG, Bassi GL, et al. Venovenous extracorporeal
removal vs standard care ventilation on 90-day mortality in patients membrane oxygenation in patients with acute COVID-19 associated
with acute hypoxemic respiratory failure: the REST randomized respiratory failure: comparative effectiveness study. BMJ 2022;
clinical trial. JAMA 2021; 326: 1013–23. 377: e068723.
66 Gattinoni L, Tonetti T, Cressoni M, et al. Ventilator-related causes of 86 Horby P, Lim WS, Emberson JR, et al. Dexamethasone in hospitalized
lung injury: the mechanical power. Intensive Care Med 2016; patients with COVID-19. N Engl J Med 2021; 384: 693–704.
42: 1567–75. 87 Angus DC, Derde L, Al-Beidh F, et al. Effect of hydrocortisone on
67 Coppola S, Caccioppola A, Froio S, et al. Effect of mechanical power mortality and organ support in patients with severe COVID-19:
on intensive care mortality in ARDS patients. Crit Care 2020; 24: 246. the REMAP-CAP COVID-19 corticosteroid domain randomized
68 Guérin C, Papazian L, Reignier J, Ayzac L, Loundou A, Forel JM. clinical trial. JAMA 2020; 324: 1317–29.
Effect of driving pressure on mortality in ARDS patients during 88 Sterne JAC, Murthy S, Diaz JV, et al. Association between
lung protective mechanical ventilation in two randomized administration of systemic corticosteroids and mortality among
controlled trials. Crit Care 2016; 20: 384. critically ill patients with COVID-19: a meta-analysis. JAMA 2020;
69 Serpa Neto A, Deliberato RO, Johnson AEW, et al. Mechanical 324: 1330–41.
power of ventilation is associated with mortality in critically ill 89 Chaudhuri D, Sasaki K, Karkar A, et al. Corticosteroids in
patients: an analysis of patients in two observational cohorts. COVID-19 and non-COVID-19 ARDS: a systematic review and
Intensive Care Med 2018; 44: 1914–22. meta-analysis. Intensive Care Med 2021; 47: 521–37.
70 Zhang Z, Zheng B, Liu N, Ge H, Hong Y. Mechanical power 90 Villar J, Ferrando C, Martínez D, et al. Dexamethasone treatment
normalized to predicted body weight as a predictor of mortality in for the acute respiratory distress syndrome: a multicentre,
patients with acute respiratory distress syndrome. randomised controlled trial. Lancet Respir Med 2020; 8: 267–76.
Intensive Care Med 2019; 45: 856–64. 91 Steinberg KP, Hudson LD, Goodman RB, et al. Efficacy and safety
71 Sarge T, Baedorf-Kassis E, Banner-Goodspeed V, et al. Effect of of corticosteroids for persistent acute respiratory distress syndrome.
esophageal pressure-guided positive end-expiratory pressure on N Engl J Med 2006; 354: 1671–84.
survival from acute respiratory distress syndrome: a risk-based and 92 Saha R, Assouline B, Mason G, Douiri A, Summers C,
mechanistic reanalysis of the EPVent-2 trial. Shankar-Har M. The impact of sample size misestimations on the
Am J Respir Crit Care Med 2021; 204: 1153–63. interpretation of ARDS trials: systematic review and meta-analysis.
72 Constantin J-M, Jabaudon M, Lefrant J-Y, et al. Personalised Chest 2022; published online May 26. https://doi.org/10.1016/j.
mechanical ventilation tailored to lung morphology versus low chest.2022.05.018.
positive end-expiratory pressure for patients with acute respiratory 93 Fuller BM, Mohr NM, Skrupky L, Fowler S, Kollef MH, Carpenter
distress syndrome in France (the LIVE study): a multicentre, single- CR. The use of inhaled prostaglandins in patients with ARDS:
blind, randomised controlled trial. Lancet Respir Med 2019; 7: 870–80. a systematic review and meta-analysis. Chest 2015; 147: 1510–22.
73 Guérin C, Albert RK, Beitler J, et al. Prone position in ARDS 94 McAuley DF, Laffey JG, O’Kane CM, et al. Simvastatin in the acute
patients: why, when, how and for whom. Intensive Care Med 2020; respiratory distress syndrome. N Engl J Med 2014; 371: 1695–703.
46: 2385–96.
95 Truwit JD, Bernard GR, Steingrub J, et al. Rosuvastatin for sepsis-
74 Guérin C, Reignier J, Richard J-C, et al. Prone positioning in severe associated acute respiratory distress syndrome. N Engl J Med 2014;
acute respiratory distress syndrome. N Engl J Med 2013; 370: 2191–200.
368: 2159–68.
96 Toner P, Boyle AJ, McNamee JJ, et al. Aspirin as a treatment for
75 Guérin C, Beuret P, Constantin JM, et al. A prospective acute respiratory distress syndrome: a randomised placebo
international observational prevalence study on prone positioning controlled clinical trial. Chest 2021; 161: 1275–84.
of ARDS patients: the APRONET (ARDS Prone Position Network)
97 Willson DF, Truwit JD, Conaway MR, Traul CS, Egan EE. The adult
study. Intensive Care Med 2018; 44: 22–37.
calfactant in acute respiratory distress syndrome trial. Chest 2015;
76 Alhazzani W, Parhar KKS, Weatherald J, et al. Effect of awake prone 148: 356–64.
positioning on endotracheal intubation in patients with COVID-19
98 Liu KD, Levitt J, Zhuo H, et al. Randomized clinical trial of activated
and acute respiratory failure: a randomized clinical trial. JAMA
protein C for the treatment of acute lung injury.
2022; 327: 2104–13.
Am J Respir Crit Care Med 2008; 178: 618–23.
77 Ehrmann S, Li J, Ibarra-Estrada M, et al. Awake prone positioning
99 Cornet AD, Groeneveld AB, Hofstra JJ, et al. Recombinant human
for COVID-19 acute hypoxaemic respiratory failure: a randomised,
activated protein C in the treatment of acute respiratory distress
controlled, multinational, open-label meta-trial. Lancet Respir Med
syndrome: a randomized clinical trial. PLoS One 2014; 9: e90983.
2021; 9: 1387–95.
100 Iwata K, Doi A, Ohji G, et al. Effect of neutrophil elastase inhibitor
78 Moss M, Huang DT, Brower RG, et al. Early neuromuscular
(sivelestat sodium) in the treatment of acute lung injury (ALI) and
blockade in the acute respiratory distress syndrome. N Engl J Med
acute respiratory distress syndrome (ARDS): a systematic review and
2019; 380: 1997–2008.
meta-analysis. Intern Med 2010; 49: 2423–32.
79 Peek GJ, Mugford M, Tiruvoipati R, et al. Efficacy and economic
101 Gates S, Perkins GD, Lamb SE, et al. Beta-Agonist Lung injury
assessment of conventional ventilatory support versus
TrIal-2 (BALTI-2): a multicentre, randomised, double-blind, placebo-
extracorporeal membrane oxygenation for severe adult respiratory
controlled trial and economic evaluation of intravenous infusion of
failure (CESAR): a multicentre randomised controlled trial. Lancet
salbutamol versus placebo in patients with acute respiratory distress
2009; 374: 1351–63.
syndrome. Health Technol Assess 2013; 17: v–vi, 1–87.

12 www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

102 Matthay MA, Brower RG, Carson S, et al. Randomized, placebo- 122 Bosch NA, Lee M-M, LeSieur MN, Law AC, Walkey AJ. Death due
controlled clinical trial of an aerosolized β₂-agonist for treatment of to irreversible hypoxemic respiratory failure in ARDSnet clinical
acute lung injury. Am J Respir Crit Care Med 2011; 184: 561–68. trials. J Crit Care 2022; 67: 85–87.
103 McAuley DF, Cross LM, Hamid U, et al. Keratinocyte growth factor 123 Ketcham SW, Sedhai YR, Miller HC, et al. Causes and
for the treatment of the acute respiratory distress syndrome (KARE): characteristics of death in patients with acute hypoxemic respiratory
a randomised, double-blind, placebo-controlled phase 2 trial. failure and acute respiratory distress syndrome: a retrospective
Lancet Respir Med 2017; 5: 484–91. cohort study. Crit Care 2020; 24: 391.
104 Gorman E, Millar J, McAuley D, O’Kane C. Mesenchymal stromal 124 Torres LK, Hoffman KL, Oromendia C, et al. Attributable mortality
cells for acute respiratory distress syndrome (ARDS), sepsis, and of acute respiratory distress syndrome: a systematic review, meta-
COVID-19 infection: optimizing the therapeutic potential. analysis and survival analysis using targeted minimum loss-based
Expert Rev Respir Med 2021; 15: 301–24. estimation. Thorax 2021; 76: 1176–85.
105 Matthay MA, Calfee CS, Zhuo H, et al. Treatment with allogeneic 125 Auriemma CL, Zhuo H, Delucchi K, et al. Acute respiratory distress
mesenchymal stromal cells for moderate to severe acute respiratory syndrome-attributable mortality in critically ill patients with sepsis.
distress syndrome (START study): a randomised phase 2a safety Intensive Care Med 2020; 46: 1222–31.
trial. Lancet Respir Med 2019; 7: 154–62. 126 Ingraham NE, Vakayil V, Pendleton KM, et al. Recent trends in
106 Bellingan G, Jacono F, Bannard-Smith J, et al. Safety and efficacy of admission diagnosis and related mortality in the medically critically
multipotent adult progenitor cells in acute respiratory distress ill. J Intensive Care Med 2022; 37: 185–94.
syndrome (MUST-ARDS): a multicentre, randomised, double-blind, 127 Wick KD, McAuley DF, Levitt JE, et al. Promises and challenges of
placebo-controlled phase 1/2 trial. Intensive Care Med 2022; personalized medicine to guide ARDS therapy. Crit Care 2021;
48: 36–44. 25: 404.
107 Dilogo IH, Aditianingsih D, Sugiarto A, et al. Umbilical cord 128 Tzotzos SJ, Fischer B, Fischer H, Zeitlinger M. Incidence of ARDS
mesenchymal stromal cells as critical COVID-19 adjuvant therapy: and outcomes in hospitalized patients with COVID-19: a global
a randomized controlled trial. Stem Cells Transl Med 2021; 10: 1279–87. literature survey. Crit Care 2020; 24: 516.
108 Lanzoni G, Linetsky E, Correa D, et al. Umbilical cord mesenchymal 129 Johnson SW, Garcia MA, Sisson EKQ, et al. Hospital variation in
stem cells for COVID-19 acute respiratory distress syndrome: management and outcomes of acute respiratory distress syndrome
a double-blind, phase 1/2a, randomized controlled trial. due to COVID-19. Crit Care Explor 2022; 10: e0638.
Stem Cells Transl Med 2021; 10: 660–73. 130 Magesh S, John D, Li WT, et al. Disparities in COVID-19 outcomes
109 Monsel A, Hauw-Berlemont C, Mebarki M, et al. Treatment of by race, ethnicity, and socioeconomic status: a systematic review
COVID-19-associated ARDS with mesenchymal stromal cells: and meta-analysis. JAMA Netw Open 2021; 4: e2134147.
a multicenter randomized double-blind trial. Crit Care 2022; 26: 48. 131 Romano SD, Blackstock AJ, Taylor EV, et al. Trends in racial and
110 Dutra Silva J, Su Y, Calfee CS, et al. Mesenchymal stromal cell ethnic disparities in COVID-19 hospitalizations, by region—
extracellular vesicles rescue mitochondrial dysfunction and improve United States, March–December 2020.
barrier integrity in clinically relevant models of ARDS. Eur Respir J MMWR Morb Mortal Wkly Rep 2021; 70: 560–65.
2021; 58: 2002978. 132 Arabi YM, Azoulay E, Al-Dorzi HM, et al. How the COVID-19
111 McKelvey MC, Abladey AA, Small DM, et al. Cathepsin S pandemic will change the future of critical care. Intensive Care Med
contributes to lung inflammation in acute respiratory distress 2021; 47: 282–91.
syndrome. Am J Respir Crit Care Med 2022; 205: 769–82. 133 Herridge MS, Tansey CM, Matté A, et al. Functional disability
112 Mehta P, Samanta R, Wick K, et al. Elevated ferritin is associated 5 years after acute respiratory distress syndrome. N Engl J Med 2011;
with systemic inflammation, inflammasome activation and 364: 1293–304.
mortality in acute respiratory distress syndrome (ARDS). 134 Wilcox ME, Patsios D, Murphy G, et al. Radiologic outcomes at
ERJ Open Res 2022; 8 (suppl 8): 118. 5 years after severe ARDS. Chest 2013; 143: 920–26.
113 Boyle AJ, Ferris P, Bradbury I, et al. Baseline plasma IL-18 may 135 Davydow DS, Desai SV, Needham DM, Bienvenu OJ. Psychiatric
predict simvastatin treatment response in patients with ARDS: morbidity in survivors of the acute respiratory distress syndrome:
a secondary analysis of the HARP-2 randomised clinical trial. a systematic review. Psychosom Med 2008; 70: 512–19.
Crit Care 2022; 26: 164.
136 Hashem MD, Hopkins RO, Colantuoni E, et al. Six-month and
114 Calfee CS, Liu KD, Asare AL, et al. Clinical trial design during and 12-month patient outcomes based on inflammatory subphenotypes
beyond the pandemic: the I-SPY COVID trial. Nat Med 2022; in sepsis-associated ARDS: secondary analysis of SAILS-ALTOS
28: 9–11. trial. Thorax 2022; 77: 22–30.
115 Beitler JR, Thompson BT, Baron RM, et al. Advancing precision 137 Needham DM, Dinglas VD, Bienvenu OJ, et al. One year outcomes
medicine for acute respiratory distress syndrome. Lancet Respir Med in patients with acute lung injury randomised to initial trophic or
2022; 10: 107–20. full enteral feeding: prospective follow-up of EDEN randomised
116 Pisani L, Algera AG, Serpa Neto A, et al. Epidemiological trial. BMJ 2013; 346: f1532.
characteristics, ventilator management, and clinical outcome in 138 Brown SM, Wilson EL, Presson AP, et al. Understanding patient
patients receiving invasive ventilation in intensive care units from outcomes after acute respiratory distress syndrome: identifying
10 Asian middle-income countries (PRoVENT-iMiC): subtypes of physical, cognitive and mental health outcomes. Thorax
an international, multicenter, prospective study. Am J Trop Med Hyg 2017; 72: 1094–103.
2021; 104: 1022–33.
139 Kamdar BB, Huang M, Dinglas VD, et al. Joblessness and lost
117 Azevedo LCP, Park M, Salluh JIF, et al. Clinical outcomes of patients earnings after acute respiratory distress syndrome in a 1-year
requiring ventilatory support in Brazilian intensive care units: a national multicenter study. Am J Respir Crit Care Med 2017;
multicenter, prospective, cohort study. Crit Care 2013; 17: R63. 196: 1012–20.
118 Gupta E, Hossen S, Grigsby MR, et al. Risk factors for the 140 Kamdar BB, Sepulveda KA, Chong A, et al. Return to work and lost
development of acute respiratory distress syndrome in mechanically earnings after acute respiratory distress syndrome: a 5-year
ventilated adults in Peru: a multicenter observational study. Crit Care prospective, longitudinal study of long-term survivors. Thorax 2018;
2019; 23: 398. 73: 125–33.
119 Saha R, Assouline B, Mason G, Douiri A, Summers C, 141 Su H, Hopkins RO, Kamdar BB, et al. Association of imbalance
Shankar-Hari M. Impact of differences in acute respiratory distress between job workload and functional ability with return to work in
syndrome randomised controlled trial inclusion and exclusion ARDS survivors. Thorax 2022; 77: 123–28.
criteria: systematic review and meta-analysis. Br J Anaesth 2021;
142 Ruhl AP, Huang M, Colantuoni E, et al. Healthcare utilization and
127: 85–101.
costs in ARDS survivors: a 1-year longitudinal national US
120 Pais FM, Sinha P, Liu KD, Matthay MA. Influence of clinical factors multicenter study. Intensive Care Med 2017; 43: 980–91.
and exclusion criteria on mortality in ARDS observational studies
143 Dinglas VD, Aronson Friedman L, Colantuoni E, et al. Muscle
and randomized controlled trials. Respir Care 2018; 63: 1060–69.
weakness and 5-year survival in acute respiratory distress
121 Juschten J, Tuinman PR, Guo T, et al. Between-trial heterogeneity in syndrome survivors. Crit Care Med 2017; 45: 446–53.
ARDS research. Intensive Care Med 2021; 47: 422–34.

www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8 13


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.
Series

144 Prescott HC, Sjoding MW, Langa KM, Iwashyna TJ, McAuley DF. 153 George M, Hernandez C, Smith S, et al. Nursing research priorities
Late mortality after acute hypoxic respiratory failure. Thorax 2017; in critical care, pulmonary, and sleep: international Delphi survey of
73: 618–25. nurses, patients, and caregivers. Ann Am Thorac Soc 2020; 17: 1–10.
145 Eberst G, Claudé F, Laurent L, et al. Result of one-year, prospective 154 Connolly B, Milton-Cole R, Adams C, et al. Recovery, rehabilitation
follow-up of intensive care unit survivors after SARS-CoV-2 and follow-up services following critical illness: an updated UK
pneumonia. Ann Intensive Care 2022; 12: 23. national cross-sectional survey and progress report. BMJ Open 2021;
146 Zangrillo A, Belletti A, Palumbo D, et al. One-year 11: e052214.
multidisciplinary follow-up of patients with COVID-19 requiring 155 Bakhru RN, Davidson JF, Bookstaver RE, et al. Implementation of
invasive mechanical ventilation. J Cardiothorac Vasc Anesth 2022; an ICU recovery clinic at a tertiary care academic center.
36: 1354–63. Crit Care Explor 2019; 1: e0034.
147 Heesakkers H, van der Hoeven JG, Corsten S, et al. Clinical 156 Cook K, Bartholdy R, Raven M, et al. A national survey of intensive
outcomes among patients with 1-year survival following intensive care follow-up clinics in Australia. Aust Crit Care 2020; 33: 533–37.
care unit treatment for COVID-19. JAMA 2022; 327: 559–65. 157 McPeake J, Boehm LM, Hibbert E, et al. Key components of ICU
148 Hodgson CL, Higgins AM, Bailey MJ, et al. The impact of recovery programs: what did patients report provided benefit?
COVID-19 critical illness on new disability, functional outcomes Crit Care Explor 2020; 2: e0088.
and return to work at 6 months: a prospective cohort study. 158 McPeake J, Iwashyna TJ, Boehm LM, et al. Benefits of peer support
Crit Care 2021; 25: 382. for intensive care unit survivors: sharing experiences, care
149 Stewart I, Jacob J, George PM, et al. Interstitial lung damage debriefing, and altruism. Am J Crit Care 2021; 30: 145–49.
following COVID-19 hospitalisation: an interim analysis of the 159 Schofield-Robinson OJ, Lewis SR, Smith AF, McPeake J,
UKILD Post-COVID study. medRxiv 2022: published online Alderson P. Follow-up services for improving long-term outcomes
March 16. https://doi.org/10.1101/2022.03.10.22272081 (preprint). in intensive care unit (ICU) survivors. Cochrane Database Syst Rev
150 McGroder CF, Zhang D, Choudhury MA, et al. Pulmonary fibrosis 2018; 11: CD012701.
4 months after COVID-19 is associated with severity of illness and 160 Connolly B, Salisbury L, O’Neill B, et al. Exercise rehabilitation
blood leucocyte telomere length. Thorax 2021; 76: 1242–45. following intensive care unit discharge for recovery from critical
151 Yang C, Zhao H, Tebbutt SJ. A glimpse into long COVID and illness. Cochrane Database Syst Rev 2015; 2015: CD008632.
symptoms. Lancet Respir Med 2022; published online June 10.
https://doi.org/10.1016/S2213-2600(22)00217-X. Copyright © 2022 Elsevier Ltd. All rights reserved.
152 Xie Y, Xu E, Bowe B, Al-Aly Z. Long-term cardiovascular outcomes
of COVID-19. Nat Med 2022; 28: 583–90.

14 www.thelancet.com Published online September 4, 2022 https://doi.org/10.1016/S0140-6736(22)01439-8


Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 16, 2022. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2022. Elsevier Inc. Todos los derechos reservados.

You might also like