Abstract
Preventing, treating, and promoting recovery from critical illness due to pulmonary disease are foundational goals of the critical care community and the NHLBI. Decades of clinical research in acute respiratory distress syndrome, acute respiratory failure, pneumonia, and sepsis have yielded improvements in supportive care, which have translated into improved patient outcomes. Novel therapeutics have largely failed to translate from promising preclinical findings into improved patient outcomes in late-phase clinical trials. Recent advances in personalized medicine, “big data,” causal inference using observational data, novel clinical trial designs, preclinical disease modeling, and understanding of recovery from acute illness promise to transform the methods of pulmonary and critical care clinical research. To assess the current state of, research priorities for, and future directions in adult pulmonary and critical care research, the NHLBI assembled a multidisciplinary working group of investigators. This working group identified recommendations for future research, including 1) focusing on understanding the clinical, physiological, and biological underpinnings of heterogeneity in syndromes, diseases, and treatment response with the goal of developing targeted, personalized interventions; 2) optimizing preclinical models by incorporating comorbidities, cointerventions, and organ support; 3) developing and applying novel clinical trial designs; and 4) advancing mechanistic understanding of injury and recovery to develop and test interventions targeted at achieving long-term improvements in the lives of patients and families. Specific areas of research are highlighted as especially promising for making advances in pneumonia, acute hypoxemic respiratory failure, and acute respiratory distress syndrome.
Keywords: acute respiratory failure, sepsis, pneumonia, mechanical ventilation, clinical trials
For 50 years, caring for patients with acute pulmonary disease has been a defining element of critical care (1, 2). Identifying interventions to prevent, treat, and facilitate recovery from pneumonia, acute hypoxemic respiratory failure (AHRF), and acute respiratory distress syndrome (ARDS) has been a priority for the critical care community and the NHLBI (3). Preclinical research (4, 5) and clinical trials (6) have yielded major improvements in supportive care, which have translated into improved patient outcomes (7, 8). However, novel therapeutics have largely failed to improve outcomes when tested in late-phase clinical trials (9–13).
To assess the current state of, research priorities for, and future directions in adult pulmonary and critical care research, in September 2018, the NHLBI assembled a working group for clinical research in adult pulmonary and critical care. This multidisciplinary group included leaders in clinical, translational, and basic science research in pulmonary, critical care, and emergency medicine. Specific areas of content expertise (pneumonia, AHRF, ARDS, and sepsis) and methodological expertise (molecular biology, preclinical disease models, early-phase clinical trials, late-phase clinical trials, long-term outcomes, health services research, and biostatistics) were represented. The meeting was divided into two types of sessions: 1) sessions addressing principles applicable to the design of clinical research in adult pulmonary and critical care and 2) sessions addressing specific disease processes. Each session was followed by group discussion of the common themes and priorities.
The first section of this report describes the predominant themes that emerged as having the greatest potential for contributing to progress in clinical research in adult pulmonary and critical care (Table 1). The second section outlines research priorities for three specific conditions highlighted for discussion by the working group: pneumonia, AHRF, and ARDS.
Table 1.
Summary of Overarching Recommendations for Future Critical Care Research
| Heterogeneity of patients, conditions, and treatment effect | For critical illness syndromes such as sepsis or ARDS, research should focus on understanding the clinical, physiological, and biological underpinnings of heterogeneity in syndromes, diseases, and treatment response to improve the development of targeted interventions and the design of clinical trials. |
| Preclinical research and early-phase clinical trials | Optimizing preclinical models by incorporating comorbidities, cointerventions, and organ failure and support and improving the use of biomarkers and surrogate outcomes in early-phase clinical trials can close the gap between mechanistic discovery and successful late-phase clinical trials. |
| Clinical study designs | Adaptive and platform designs may increase the efficiency of evaluating novel therapies for a disease or subphenotypes of a disease. Cluster-level trials and trials leveraging electronic health records may increase the efficiency of comparing the effectiveness of common therapies for broad groups of patients. |
| Long-term patient-important outcomes | Future research should incorporate measurement of long-term patient-important outcomes and potential surrogate outcomes into observational and interventional studies to advance mechanistic understanding of injury, recovery, and adaptation. Trials should test interventions targeting these mechanisms to improve the lives of patients and their caregivers. |
Definition of abbreviation: ARDS = acute respiratory distress syndrome.
Overarching Recommendations for Future Critical Care Research
Heterogeneity of Patients, Conditions, and Treatment Effect
A fundamental challenge for critical care research today is to identify which critically ill patients are likely to experience improved clinical outcomes with a given therapy. Meeting this challenge will require understanding three types of risk: 1) a patient’s overall risk of an outcome, 2) the proportion of a patient’s overall risk of an outcome that is attributable to the disease of interest, and 3) the proportion of the patient’s overall risk of an outcome that is amenable to treatment with the therapy being evaluated (Figures 1 and E1 in the online supplement).
Figure 1.

Relationships between overall risk, disease-attributable risk, and treatment-responsive risk. The x-axis displays, for a theoretical population of critically ill patients with a disease of interest, each patient’s predicted risk of death, ranging from 0% (will almost certainly survive) to 100% (will almost certainly die). The y-axis displays the observed percentage of patients who die, ranging from 0% (no patients died) to 100% (all patients died). The light gray shading denotes the deaths that were caused by the disease of interest (disease-attributable risk). The medium gray shading denotes deaths that were caused by comorbidities or concurrent organ failures rather than the disease of interest. The dark gray denotes deaths prevented by treatment. (A) A disease-specific treatment will generally be able to prevent more deaths among patients who have a greater overall risk of death (prognostic enrichment) and whose risk of death is primarily due to the disease being treated rather than comorbidities or concurrent organ failures. (B) A specific therapy may have a greater effect on outcomes in a highly treatable disease compared with a poorly treatable disease. Which patients respond to a specific treatment depends on the mechanism of action and the biology of the disease. (C) The probability of responding to a specific treatment might be greatest among patients at high overall risk or low overall risk, or it might be completely independent of overall risk. Disease-specific treatments would generally be expected to prevent deaths resulting from the disease of interest (light gray), whereas some supportive therapies might be expected to prevent deaths resulting from the disease of interest (light gray) and deaths resulting from comorbidities or concurrent organ failures (medium gray).
Overall risk
Within a population of critically ill patients, individual patients may differ greatly in their risk of an outcome (14). Patients at higher risk of a specific outcome have the potential to experience greater absolute reductions in that outcome with treatment (when disease-attributable and treatment-responsive risk are constant) (Figure 1) (14, 15). Patients at low risk of a specific outcome may have little potential to benefit from the treatment but may still experience its adverse effects (14). Enrolling patients at high risk of an outcome (prognostic enrichment) in critical care trials can ensure a high absolute event rate in the control group, potentially decreasing sample size, increasing power, and improving benefit–risk balance.
Attributable risk
Critical illness is complex, and frequently only a portion of a patient’s overall risk of an outcome is due to the disease of interest (attributable risk), whereas much of the patient’s risk of an outcome is due to comorbidities or concurrent organ failures (16–19) (Figure 1). For example, the overall in-hospital mortality among patients with ARDS is 30–40% (7). As much as 80% of the mortality risk among patients with ARDS, however, may be attributable to the underlying disease or concurrent organ failures rather than to ARDS itself (20). Thus, preventing or treating ARDS may produce only a small decrease in overall mortality. Hence, moribund patients, who have a high overall risk of death, of which only a small portion is attributable to the disease of interest, are frequently considered unlikely to benefit from study treatments and thus are excluded from clinical trials of ARDS therapies. Historically, critical care trials have selected sample sizes using overall rates of an outcome (21). Using more conservative estimates focused on the risk attributable to the disease, rather than the patient’s overall risk, may allow more realistic sample size estimations (Figure 1A).
Treatment-responsive risk
The difference between an individual’s outcome with a treatment versus without the treatment is the “treatment-responsive risk” (Figure 1C). Patients more likely to respond to a particular treatment may be identified using demographic characteristics (22), pathophysiology (23), prior response to the treatment (24), or a disease characteristic related to the treatment’s mechanism of action (e.g., genomic or proteomic factor) (25). Selectively enrolling patients in critical care trials who are more likely to respond to the treatment (predictive enrichment) can produce greater absolute and relative risk reductions, decrease sample size, and improve the benefit–risk balance for patients (23, 26–28).
Critical Illness Syndromes, Phenotypes, and Endotypes
Identifying patients likely to respond to a disease-specific treatment requires understanding the biology of the disease, the likely mechanistic effect of the treatment, and the interaction between the two. Historically, many critical illnesses (e.g., sepsis and ARDS) have been considered syndromes, defined by observable signs and symptoms, but not necessarily specific to underlying biological processes. Ideally, such syndromic definitions would be replaced with increasingly precise disease phenotypes (29), subphenotypes, and endotypes (Table 2). Critical care may learn from the fields of oncology and asthma, which have used biological pathways to define disease, identify therapeutic targets, and design efficient clinical trials (25, 30–32). This will require critical care research to develop valid proxy measures of mechanism in the causal pathway between intervention and outcome.
Table 2.
Approaches to Grouping Patients for Research
| Term | Definition | Example |
|---|---|---|
| Syndrome | A set of medical signs and symptoms that are correlated with each other but may or may not be directly related to a specific disease | Asthma |
| Phenotype | Observable characteristics (clinical, physiological, morphological, biological, or response to treatment) without known relation to a pathophysiological mechanism | Early-onset allergic asthma |
| Subphenotype | A subset of a phenotype that is characteristic of a subset of a population | Early-onset allergic asthma with sputum eosinophilia |
| Endotype | A subset of a disease or syndrome defined by a distinct functional or pathophysiological mechanism | Th2 cytokines and allergen-specific IgE mediate atopy and eosinophilic inflammation responsive to corticosteroids and antibodies to IL-13 and IL-5 |
Definition of abbreviation: Th2 = T-helper cell type 2.
Large observational and interventional datasets with standardized clinical phenotyping, physiological measures, and biological samples from both acute illness and postacute recovery should be used to propel understanding of phenotypes, subphenotypes, and their mechanisms. Partnerships between academia, industry, and the U.S. Food and Drug Administration will be required to develop and validate approaches to rapid phenotyping and theragnostics (strategies that combine a diagnostic test which identifies patients likely to respond and targeted therapy based on that test’s results) for use in clinical trials. Markers of subphenotypes may be “static,” such as baseline plasma cytokine concentration (33), or “dynamic,” such as a patient’s physiological response to a change in positive end-expiratory pressure at the beginning of an ARDS trial (34). Ultimately, new interventions in critical care may be developed and tested using biological criteria rather than clinical syndromes, including potentially testing treatments in subphenotypes of patients that cut across historically separate clinical syndromes.
Preclinical Research
Major advances that have improved clinical outcomes for patients with ARDS began with preclinical animal studies. Research in small- and large-animal models established that higher Vts and high airway pressures were injurious to the normal and injured lung, paving the way for clinical trials to identify a marked reduction in mortality with use of lower Vts (4–6). Similarly, the potential benefit of a conservative approach to fluid management in ARDS was based on animal models of acute lung injury that demonstrated less pulmonary edema with reduced pulmonary vascular hydrostatic pressure (35).
Studies involving preclinical models must be designed with a specific aim and must be understood within that specific framework. For some preclinical models, the aim will be purely to advance mechanistic understanding (e.g., examining lung epithelial tissue factor–knockout mice to understand determinants of alveolar–capillary barrier permeability). With many preclinical models, the aim is to examine a new therapeutic and establish a mechanism of benefit and a proof of principle that the new therapy might have potential value in the clinical setting. To accomplish this objective, there is substantial need for preclinical models that have more direct clinical relevance. For example, a recent international expert consensus initiative provided recommendations for more representative preclinical models of sepsis (many of which apply to other critical illnesses) (36). Overall, preclinical models need to
-
1.
Examine model systems beyond rodents (e.g., large animals [37], ex vivo human lungs [38], and LPS inhalation in human volunteers [39]);
-
2.
Include covariates and comorbidities in animal models (e.g., age, diabetes, and immunosuppression);
-
3.
Induce sepsis in a manner similar to the onset of sepsis in humans (e.g., microorganisms common in human sepsis [40], sites other than the peritoneal cavity [41], and prior microbial exposure to establish immunological memory and trained immunity [42]);
-
4.
Include organ dysfunction and organ support in animal models (e.g., mechanical ventilation for respiratory failure);
-
5.
Administer cointerventions similarly to current clinical care for human patients with sepsis (e.g., administration of antimicrobials and crystalloid solutions after, rather than before, sepsis onset);
-
6.
Use clinically relevant outcomes over an appropriate time scale.
Ideally, research using animal models should evaluate the full spectrum of critical illness from prevention and early treatment through established disease and the resolution and recovery phases. In addition to more clinically relevant preclinical models, preclinical research should leverage highly controlled and replicable circumstances to understand not just whether a therapy works but also the circumstances under which it does not work, as well as the effects of specific aspects of the disease on the model and the therapy. An American Thoracic Society consensus document provides detailed recommendations for how animal studies of acute lung injury can be designed to encompass key elements of ARDS (43).
Another important issue with animal studies in sepsis and ARDS is the widespread lack of transparent, quality reporting of study design and implementation, including reporting randomization, blinding, and statistical power (44, 45). Ideally, preclinical research in pulmonary and critical care should move to adopt the Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines (46). Optimizing preclinical models by incorporating randomization and blinding may improve the eventual translation of promising novel therapeutics from preclinical research into human clinical trials.
Early-Phase Clinical Trials and Surrogate Outcomes
Early-phase clinical trials are highly controlled studies enrolling small numbers of carefully selected patients. The aim of early-phase clinical trials is to advance understanding of pathophysiology (47, 48); define the route, dose, and side effects of a novel therapy (49); or assess the effect of a novel therapy on surrogate outcomes (50). Identifying surrogate outcomes for use in early-phase clinical trials is an urgent unmet need for critical care research (51). A surrogate outcome is a laboratory-, survey-, or performance-based measurement or a physical sign used as a substitute for a clinical outcome. For a surrogate outcome to be valid, the effect of the therapy on the surrogate outcome must predict the effect of the therapy on the clinical outcome. Development of validated, short-term surrogate outcomes could smooth the transition from preclinical models to early-phase clinical trials, increase the efficiency of adaptive trials, and facilitate the incorporation of long-term, patient-important outcomes into a broader range of trials. For long-term outcomes specifically, biological (e.g., S100B), structural (e.g., cortical thickness on magnetic resonance imaging), or functional (e.g., neuronal activity on functional magnetic resonance imaging) biomarkers would ideally eventually predict cognitive outcomes (52–54), assess the short-term efficacy of interventions (55), and elucidate the mechanisms of injury and recovery in critical illness (54, 56).
Clinical Study Design
In addition to the challenges summarized above regarding heterogeneity of patients, conditions, and treatment effects, the therapeutic window for many critical care interventions is narrow and early. Patients are often vulnerable or cognitively impaired, which complicates participation in research (57–59). To address these challenges, critical care research should 1) develop and apply novel trial designs (e.g., adaptive trials and platform trials), 2) leverage electronic health records (EHRs) to conduct pragmatic trials and observational research, 3) advance the understanding of and approaches to informed consent in acute care research, and 4) apply advanced observational research methods to inform both the design of trials and clinical care.
Adaptive and platform trials
Adaptive and platform trial designs hold the potential to minimize exposure of critically ill adults to ineffective therapies while shepherding effective therapies from development to clinical practice quickly and cost-effectively. An adaptive trial is defined as a trial designed to allow prospectively planned modifications to trial features (e.g., number of study groups, sample size per group, enrollment criteria, and dose or frequency of the intervention) based on accumulating data from subjects in the trial (Figure E2). Adaptive designs may gain efficiency from adaptive enrichment of the study population, early discontinuation of worse-performing treatments, and seamless transition from early- to late-phase trials.
A platform trial is defined as a trial using a single master protocol and research infrastructure to simultaneously evaluate multiple interventions and/or disease subpopulations in multiple substudies (Figure E2). Platform trials gain efficiencies from shared control groups, adaptive borrowing of information from similar groups of patients, and shared infrastructure and governance. However, there may be challenges, including considerable pretrial planning (e.g., statistical modeling, patient selection, organization of study arms, and within-trial adaptations), funding (e.g., traditional funding mechanisms may not easily handle a trial with no fixed sample size; a trial that tests multiple experimental agents owned by multiple companies cannot easily be sponsored by a single company), study monitoring, and consent (e.g., communicating risks and benefits to patients for each separate agent being tested).
Trials that combine features of adaptive and platform designs (adaptive platform trials) are currently addressing choice of antibiotic, use of steroids, and optimal ventilator strategy in community-acquired pneumonia (NCT02735707) and evaluating interventions to improve outcomes of elective surgery (NCT03861767). Additional efficiency may be gained by integrating adaptive platform trials with the EHR in the form of randomized, embedded, multifactorial, adaptive platform (“REMAP”) trials (60).
Adaptive and platform trials in critical care face specific design challenges and potential limitations. Methods developed for oncology trials with long enrollment windows, interventions implemented and monitored over long durations, and biologically established short-term surrogate outcomes may not extrapolate straightforwardly to critical care trials. The complexity of critical illness and the significant potential for interactions between multiple interventions and cointerventions require careful consideration during the design and sample size estimation of adaptive and platform trials.
Pragmatic trials
Pragmatic trials aim to evaluate the effectiveness of a treatment under “real-world” conditions by incorporating broad eligibility criteria, administration of the intervention by treating clinicians, and simplified data collection for patient-important outcomes (61). Pragmatic trial designs are well suited to critical care comparative effectiveness questions for which the required sample size is large, generalizability is a primary focus, or the intervention is more reliably delivered by clinical personnel than by study personnel (e.g., a trial of tracheal intubation vs. laryngeal mask airway during out-of-hospital cardiac arrest when research personnel are not present and the intervention is urgent). In the unpredictable environments of the prehospital setting, emergency department, and ICU, executing traditional study procedures (e.g., screening, consenting, enrolling, randomizing, and delivering the intervention) during a narrow therapeutic window may be challenging, disruptive to clinical care, or impossible. For questions in which early intervention is paramount, specific pragmatic designs may improve representative patient enrollment and reliable delivery of interventions. For example, cluster-randomized, cluster-crossover, or stepped-wedge trials, in which providers or units are randomized, may facilitate examination of urgent or emergent critical care interventions that would be challenging to rigorously evaluate using patient-level randomization (62, 63). Pragmatic trials may provide the sample size and breadth of patients necessary to examine not only the average effect of an intervention among all patients to whom the intervention might be applied in clinical practice but also the estimated effects of the intervention for individual patients (64).
Leveraging EHRs
Trials with traditional, adaptive, platform, or pragmatic designs (and observational research) may all potentially gain efficiency by leveraging the EHR. Applications that interface directly or indirectly with the EHR may facilitate multiple steps of a traditional randomized trial, including the following:
-
1.
Screening for eligible patients (e.g., an ARDS “sniffer” [65])
-
2.
Prompting and documenting informed consent (e.g., point-of-care consent for patients and clinicians [66])
-
3.
Generating study group assignment (e.g., EHR-embedded randomization [67])
-
4.
Delivering the intervention (e.g., via order sets or an advisor [68])
-
5.
Monitoring treatment compliance and adverse events (69)
-
6.
Providing data on cointerventions and short- and long-term outcomes (70)
Challenges to be addressed include improving quality and standardization of data entry in the EHR, developing registries and reproducible approaches for automatically extracting validated EHR data, building systems for real-time monitoring of adverse events, embedding machine learning models for phenotyping patients using EHR data, addressing data security and data-sharing concerns in multicenter trials, and collaborating with third-party and EHR vendors for support of clinical research. A potential benefit of integrating clinical research into the delivery of critical care through the EHR is to facilitate implementation of new evidence into practice as a part of a learning healthcare system (71).
Informed consent
Critical care research faces unique challenges regarding informed consent. Critical care research occurs in complex environments with patients who frequently lack decision-making capacity and need interventions on a time scale of minutes or hours rather than days. Current federal regulations specify two approaches to informed consent applicable to critical care research: 1) enrollment with prospective informed consent from patients who have full decision-making capacity or from an available legally authorized representative or 2) enrollment with waiver of or exception from informed consent when obtaining informed consent is impracticable (e.g., because of patients’ illness) and obtaining informed consent from a legally authorized representative is impracticable (e.g., because of the urgency of the intervention). Alternative approaches to prospective informed consent could be optimized for patients who are acutely ill but awake (e.g., abbreviated consent with informed refusal), sedated or in severe pain (e.g., abbreviated consent, assent, or dissent), or clinically unstable but possibly aware (e.g., simple disclosure) (72). Federal regulators and stakeholders in critical care research should evaluate alternative approaches to informed consent in the specific context of prehospital, emergency medicine, and critical care research. Simply denying current and future critically ill patients the opportunity to benefit from new research because traditional models of informed consent do not work well is unacceptable. Instead, models of informed participation in research are needed that reflect the realities of the context in which critical care is practiced and recognize the obligation of healthcare professionals and institutions to continuously improve the quality of clinical care through research (73). Achieving these aims will be facilitated by involving patients, families, and community members in each stage of critical care research from design to dissemination (74).
Advanced methods for observational research
For clinically important questions in critical care that cannot be readily answered in a clinical trial, analyses of observational data provide an important source of causal inference. Observational data can quantify the incidence of the disease (75), estimate the frequency of outcomes (76), identify subphenotypes (29), provide risk-adjusted associations between interventions and outcomes (77), and provide a sustainable context into which interventional studies are integrated (78). Increasingly, modern analytic techniques (79) applied to well-curated datasets may address many sources of confounding and either mirror the results of randomized trials (80) or provide generalizable information about patients underrepresented in traditional explanatory trials. The advent of “big data,” from “-omics” to million-patient EHR registries, provides the opportunity for complex, dynamic, multilevel datasets to be gathered, curated, and integrated across ICUs, hospitals, and regions. Priorities for future critical care research using big data are summarized in Table 3.
Table 3.
Recommendations for Critical Care Research Using “Big Data”
| 1. Promote collaboration across health centers, disciplines, and federal institutes |
| 2. Revise funding models for information technology proposals to keep pace with rapidity of technological advancement |
| 3. Incentivize standardized data entry in the electronic health record during clinical care to facilitate research |
| 4. Implement a common data model and data curation to minimize missing data and optimize the quality of real-world data derived from electronic health records, health plan data, and patient-reported outcomes |
| 5. Build distributed data networks and promote a standard framework for securely sharing and exchanging patient information and ensuring application interoperability |
| 6. Link electronic health record registries with biospecimen repositories |
| 7. Explore unsupervised approaches to phenotype derivation |
| 8. Employ infrastructure developed for observational research to facilitate the conduct of pragmatic trials; phase IV postmarketing studies for drug safety and effectiveness; and data collection during pandemics, emergencies, and natural disasters |
Long-Term, Patient-Important Outcomes
Core outcomes and measures
Over the past decade, substantial progress has been made in understanding which outcomes after critical illness are important to patients and how these outcomes should be measured (81). A program of research (www.improvelto.com)—including systematic reviews (82); international surveys of patients, caregivers, clinicians, and researchers (83); and an international modified Delphi consensus process (84, 85)—produced a recommended core outcome set and core outcome measurement set for all studies evaluating outcomes after hospital discharge for survivors of acute respiratory failure. Statistical methods have been developed for analyzing such functional outcomes while addressing truncation due to mortality (86). To facilitate interpretation of results across trials, future research evaluating patient-important outcomes after hospital discharge should include the established core outcome set in addition to study-specific outcomes. The core outcome set consists of survival, health-related quality of life, mental health, pain, cognition, physical function, muscle and/or nerve function, and pulmonary function (84). The established core outcome measurement set (85) should be used, and measurement instruments for core outcomes currently lacking consensus should be evaluated (87).
Long-term outcomes in randomized trials
Long-term, patient-important outcomes within a randomized trial may serve as the primary outcome (capturing a causal process affected by the intervention with a measurable effect size) (88, 89), secondary outcomes (important explanatory variables for which the study may have insufficient power) (90), safety outcomes (measures of harm through the primary causal mechanism or other pathways), or descriptive outcomes (characterizing the trajectory of the cohort over time, regardless of differences between groups). Specific, long-term, patient-important outcomes should be considered as the primary outcome for future critical care trials in which the mechanistic effect of the intervention is anticipated to affect such outcomes and as safety outcomes in many critical care trials in which the effect of the intervention on such outcomes is uncertain.
Knowledge gaps about long-term outcomes
Several knowledge gaps currently limit the broader use of long-term, patient-important outcomes in critical care research. First, limited progress has been made in understanding and measuring patients’ preillness baseline function and trajectory (which is a prerequisite to understanding how much disability is induced by critical illness and might be preventable with intervention) (91). A standardized approach to retrospectively ascertaining preillness conditions and trajectories could be developed, validated, and applied to observational and interventional critical care research.
Second, understanding the mechanism and timing of injury and recovery is a prerequisite to designing trials with the right intervention, dose, timing, frequency, intensity, and duration to improve long-term, patient-important outcomes. Large-cohort studies should advance mechanistic understanding of injury and recovery for patients and families by collecting clinical and biological samples before, during, and after acute illness. Contemporaneously, we should endeavor to understand adaptation to injuries that cannot be prevented or healed but whose impact on health and function can be mitigated.
Third, future research should evaluate the effects of critical illness not only on patients but also on families, caregivers, and healthcare professionals. Interventions should be evaluated that target a family-centered model of care, including enhanced social and community support, mental health interventions, or respite care. Engaging patients and families in critical care research may provide new insights into questions, methods, and outcomes and ensure accountability and a patient- and family-centered research focus (74). Improving long-term outcomes for critically ill patients may also require evaluating interventions to prevent and treat “burnout” among healthcare professionals, either organizationally (e.g., shift scheduling) or personally (e.g., mindfulness training) (92).
Research in Specific Diseases
Pneumonia
Pneumonia is the most common cause of both sepsis and ARDS and is a leading cause of death in the United States (6, 93, 94). Decreasing the incidence, severity, and sequelae of pneumonia is a priority for research in adult pulmonary and critical care. Improving understanding of and treatment for pneumonia faces several challenges. First, pneumonia is falsely considered to be merely an acute illness. Patients with pneumonia experience residual effects on cytokine concentrations past hospital discharge (95, 96) and clinical symptoms for months (97). Research should delineate host factors that determine susceptibility to pneumonia and factors that influence resolution of lung injury and long-term sequelae (42, 98–100). Second, pneumonia is incorrectly perceived to be merely a localized illness. Research should examine factors that influence the transition from local infection in the lung (pneumonia) to inflammatory responses elsewhere in the lung (ARDS) or systemically (sepsis). Research should also examine the mechanisms linking pneumonia to cardiac, cerebrovascular, renal, and other nonpulmonary sequelae, independent of sepsis, hypotension, or metastatic infection (101). Third, pneumonia is frequently treated as a syndrome. Cohort studies collecting clinical and biological samples before, during, and after severe pneumonia (98) should identify biological pathways (endotypes) underlying clinical presentations of pneumonia and mediators of injury resolution (102, 103) to target interventions at mechanisms rather than clinical syndromes (104). Specific research priorities for community-acquired pneumonia are presented in Table 4.
Table 4.
Research Priorities for Community-acquired Pneumonia
| Summary of Research Priorities | Illustrative Examples | |
|---|---|---|
| Before pneumonia (prevention) | • Evaluate systemic and pulmonary markers of host susceptibility to pneumonia | • Use imaging and biospecimens to define and differentiate mechanistic signals in “protected” individuals (e.g., young healthy humans and animal models with relevant exposures establishing lung protection) versus more “susceptible” individuals (e.g., humans at high risk for pneumonia and animal models with relevant risk factors); test if those signals calibrate pneumonia risk when measured in human populations or shift pneumonia risk when manipulated in animal models |
| • Examine mechanisms that defend the lung against pneumonia, including the ability to eliminate microbes (immune resistance) and the ability to withstand injury from infection and inflammation (tissue resilience) | ||
| • Develop and test interventions that reverse or slow the progression of increasing pneumonia susceptibility | ||
| • Examine the effects of antecedent respiratory infections on innate immunity in the lung (e.g., alveolar macrophage phenotypes) and local immunological memory in the lung (e.g., resident memory T cells) | ||
| During pneumonia (treatment) | • Advance cross-disciplinary evaluation of host–pathogen interactions | • Identify candidate pharmacologic agents that phenocopy signaling pathways tied to protection or reverse signaling pathways tied to injury and test lead candidates in experimental systems for abilities to bolster immune killing of microbes or resilience of lung and extrapulmonary tissues |
| • Collect pulmonary and nonpulmonary biospecimens in observational and interventional studies to identify biological pathways (endotypes) underlying clinical presentations of pneumonia to target interventions at mechanisms rather than clinical syndromes | ||
| • Examine factors that influence the transition from localized infection (pneumonia) to inflammatory responses elsewhere in the lung (ARDS) or systemically (sepsis) | ||
| • Develop and test interventions that 1) help the host to more efficiently kill microbes, 2) diminish the anatomical and physiological damage from infection and inflammation, and 3) interrupt pneumonia’s nonpulmonary sequelae (e.g., muscle wasting) | ||
| • Leverage new technology for rapid, comprehensive pathogen identification to ensure that evaluation of pneumonia treatments occurs in the context of early and appropriate antibiotic therapy | ||
| • Elucidate the transition from infection and inflammation to resolution and repair (e.g., the role of specialized proresolving mediators) and the implications for immunomodulatory therapy | ||
| After pneumonia (facilitating resolution and recovery) | • Examine the mechanisms linking pneumonia to long-term pulmonary sequalae (e.g., accelerated chronic respiratory disease) and nonpulmonary sequelae (e.g., cardiac and cerebrovascular events and impaired cognition) | • Identify discrete molecular, physiological, or clinical signals that discriminate patients with pneumonia who developed adverse long-term outcomes from others who did not; test those signals for prognostic value in human patients and causation in animal models |
| • Incorporate noninfectious outcomes (e.g., cardiovascular events) into long-term outcomes in pneumonia research | ||
| • Define trajectories of recovery after pneumonia, as well as the biological, clinical, and social factors that influence these trajectories |
Definition of abbreviation: ARDS = acute respiratory distress syndrome.
AHRF
AHRF, characterized by tachypnea and diminished arterial partial pressure of oxygen, occurs in 25–50% of ICU patients (105). A fundamental challenge in AHRF research is that numerous distinct diseases (e.g., cardiogenic pulmonary edema, community-acquired pneumonia, and acute eosinophilic pneumonia) or syndromes (e.g., sepsis and ARDS) may present as AHRF. Research in AHRF should balance the imperative to identify effective, common therapies for AHRF across a broad range of patients with recognition of the reality that identifying targeted, mechanistic therapies may require dividing patients with AHRF by the underlying biological processes, trajectories, and phases of illness. The degree to which the underlying biology and response to therapy are shared between the syndromes of AHRF and ARDS remains unclear, including whether the recently proposed subphenotypes of ARDS (33) have AHRF counterparts. Large, observational cohort studies with detailed phenotyping, endotyping, and genotyping are needed to understand the relationships between pneumonia, AHRF, and ARDS. Examples of important unanswered questions in the clinical care of mechanically ventilated patients with AHRF are provided in Table 5.
Table 5.
Research Questions in Acute Hypoxemic Respiratory Failure with Invasive Mechanical Ventilation
| Examples of Research Questions | |
|---|---|
| Extracorporeal support | • Does venovenous extracorporeal membrane oxygenation improve outcomes for patients with mild-to-moderate ARDS or for patients with AHRF without ARDS? |
| • Should extracorporeal carbon dioxide removal be used to facilitate ultralow-Vt ventilation? | |
| Spontaneous breathing | • Can extracorporeal carbon dioxide removal facilitate safe spontaneous breathing? |
| • Should we encourage spontaneous breathing by early transition to pressure support ventilation? | |
| • Should we encourage spontaneous breathing with airway pressure release ventilation? | |
| • Among spontaneously breathing patients, should we actively limit Vt? | |
| Personalized ventilation | • Should we measure patient effort and titrate ventilatory support to achieve normal levels? |
| • Does limiting driving pressure or mechanical power improve outcomes beyond pressure- or volume-limited ventilation? | |
| • Does minimizing asynchrony improve patient outcomes? | |
| Comparative effectiveness | • How do outcomes compare between higher positive end-expiratory pressure and prone positioning overall and for specific subpopulations of patients? |
Definition of abbreviations: AHRF = acute hypoxemic respiratory failure; ARDS = acute respiratory distress syndrome.
ARDS
ARDS has been an archetypal illness for adult critical care (6, 106, 107). Decades of basic, translational, and clinical research have improved scientific understanding of ARDS (108). Clinical trials have identified strategies for supportive care (many related to minimizing the harms of mechanical ventilation) that reduce systemic inflammation (109) and improve patient outcomes (6, 110). No pharmacological therapy for ARDS has been shown to reduce short-term or long-term mortality. In this way, ARDS exemplifies the challenges and opportunities facing adult pulmonary and critical care research outlined in this summary.
Improving outcomes for patients at risk for ARDS, for those with established ARDS, and for those recovering from ARDS remains a priority for adult pulmonary and critical care research. Each of the overarching recommendations for future critical care research described above applies to future clinical research in ARDS (111), as summarized in Table 6. Meeting these challenges in ARDS research may help clear the path for clinical research in adult pulmonary and critical care more broadly and advance the shared goal of enhancing health, lengthening life, and reducing illness and disability for critically ill adults and their families.
Table 6.
Methodologic Recommendations for Acute Respiratory Distress Syndrome Research
| Summary of Methodologic Recommendations | Illustrative Examples | |
|---|---|---|
| Precision medicine | • Dissect the clinical syndrome of ARDS into the underlying subphenotypes or endotypes and test interventions targeted at biological pathways | • A trial comparing the efficacy of multiple interventions based on biomarkers and clinical outcomes among critically ill adults with a specific ARDS phenotype |
| Preclinical models | • Develop preclinical models of ARDS that better represent human disease | • Blinded, randomized trial of interventions in large-animal models that recapitulate ARDS etiology and biology in humans and subsequent treatment |
| Biomarker development | • Develop reliable biomarkers of prognosis and treatment response to bridge from preclinical research to late-phase clinical trials | • Research using preclinical models and existing or prospective human studies to understand and validate the role of specific molecules in the development, progression, and resolution of ARDS |
| Novel research designs | • Employ novel trial designs and leverage the EHR to improve the efficiency of ARDS clinical trials | • Cluster-randomized trial using electronic health records to identify potential participants, monitor delivery of the intervention, and continuously transmit curated, deidentified data to a coordinating center |
| Long-term outcomes | • Develop and incorporate a core set of long-term patient-important outcomes into ARDS clinical trials to improve understanding of patient trajectory, acute illness, and recovery | • Trial comparing a novel intervention with current clinical care with regard to long-term patient-important outcomes |
Definition of abbreviations: ARDS = acute respiratory distress syndrome; EHR = electronic health record.
Conclusions
This NHLBI working group made several recommendations for future clinical research in adult pulmonary and critical care. Increased focus on understanding the clinical, physiological, and biological underpinnings of heterogeneity in syndromes, diseases, and treatment response will improve the development of targeted interventions to be tested with novel and efficient clinical trials. Optimizing preclinical research by using designs with randomization and blinding and incorporating comorbidities, cointerventions, and organ support into animal models may improve the translation of promising novel therapeutics from preclinical research to late-phase clinical trials. Developing novel biomarkers associated with patient prognosis and treatment response may help ensure that clinical trials evaluate novel therapeutics for the right patients at the right time. Adaptive, platform, and pragmatic trial designs may efficiently identify efficacious new therapies and compare the effectiveness of existing ones. Incorporating long-term, patient-important outcomes into large-scale observational studies and clinical trials will improve understanding of patient trajectories and mechanisms of injury and recovery and allow evaluation of targeted interventions to improve the lives of patients, caregivers, and healthcare professionals. Pneumonia, AHRF, and ARDS offer specific promising research opportunities to improve mechanistic understanding of disease and test available therapies with the potential to improve patient outcomes.
Supplementary Material
Acknowledgments
Acknowledgment
The authors acknowledge the following individuals who attended the working group meeting: Gail Weinmann, M.D.; Mario Stylianou, Ph.D.; and Myron Waclawiw, Ph.D.; of the NHLBI; Sarah Dunsmore, Ph.D., of the National Institute of General Medical Sciences; Chao Jiang, Ph.D., of the National Institute of Allergy and Infectious Diseases; Robert Tamburro, M.D., of the Eunice Kennedy Shriver National Institute of Child Health and Human Development; and Karen Huss, Ph.D., R.N., A.N.P.-B.C., of the National Institute of Nursing Research.
Footnotes
Supported in part by the National Institute for General Medical Sciences (grants P01GM095467 [B.D.L.] and R35GM119519 [C.W.S.]), the NHLBI (grants R35HL140026 [C.S.C.], R24HL111895 [D.M.N.], R35HL135756 and R33HL137081 [J.P.M.], U01HL123004 and R42HL126456 [M.A.M.], K24HL089223 [M.M.], K23HL143053 [M.W.S.], R01HL132887 [R.D.S.], and K12HL138039 [T.J.I.]), the National Institute of Allergy and Infectious Diseases (grants R01AI115053 [J.P.M.] and U19AI135964 [R.G.W.]), the National Center for Complementary and Integrative Health (grant R34AT009181 [M.M.]), the American Thoracic Society (M.M.), the National Institute on Aging (grant R01AG050698 [R.D.S.]), the Intermountain Research and Medical Foundation (R.O.H.), and the Veterans Health Administration (grant I01HX002390 [T.J.I.]). The content of this manuscript is the responsibility of the authors alone and does not necessarily reflect the views or policies of the U.S. Department of Veterans Affairs, the U.S. Department of Health and Human Services, or the U.S. government.
Author Contributions: Drafting of the manuscript: M.W.S., G.R.B., and L.A.R. Critical revision of the manuscript for important intellectual content: all authors.
This article has an online supplement, which is accessible from this issue’s table of contents at www.atsjournals.org.
Originally Published in Press as DOI: 10.1164/rccm.201908-1595WS on March 9, 2020
Author disclosures are available with the text of this article at www.atsjournals.org.
References
- 1.Weil MH, Tang W. From intensive care to critical care medicine: a historical perspective. Am J Respir Crit Care Med. 2011;183:1451–1453. doi: 10.1164/rccm.201008-1341OE. [DOI] [PubMed] [Google Scholar]
- 2.Ibsen B. The anaesthetist’s viewpoint on the treatment of respiratory complications in poliomyelitis during the epidemic in Copenhagen, 1952. Proc R Soc Med. 1954;47:72–74. doi: 10.1177/003591575404700120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Spragg RG, Bernard GR, Checkley W, Curtis JR, Gajic O, Guyatt G, et al. Beyond mortality: future clinical research in acute lung injury. Am J Respir Crit Care Med. 2010;181:1121–1127. doi: 10.1164/rccm.201001-0024WS. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.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. doi: 10.1164/arrd.1974.110.5.556. [DOI] [PubMed] [Google Scholar]
- 5.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. 1984;57:1809–1816. doi: 10.1152/jappl.1984.57.6.1809. [DOI] [PubMed] [Google Scholar]
- 6.Brower RG, Matthay MA, Morris A, Schoenfeld D, Thompson BT, Wheeler A Acute Respiratory Distress Syndrome Network. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N Engl J Med. 2000;342:1301–1308. doi: 10.1056/NEJM200005043421801. [DOI] [PubMed] [Google Scholar]
- 7.Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, et al. LUNG SAFE Investigators; ESICM Trials Group. 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. [Published erratum appears in JAMA 316:350.] [DOI] [PubMed] [Google Scholar]
- 8.Kaukonen KM, Bailey M, Suzuki S, Pilcher D, Bellomo R. Mortality related to severe sepsis and septic shock among critically ill patients in Australia and New Zealand, 2000-2012. JAMA. 2014;311:1308–1316. doi: 10.1001/jama.2014.2637. [DOI] [PubMed] [Google Scholar]
- 9.Bernard GR, Wheeler AP, Russell JA, Schein R, Summer WR, Steinberg KP, et al. Ibuprofen in Sepsis Study Group. The effects of ibuprofen on the physiology and survival of patients with sepsis. N Engl J Med. 1997;336:912–918. doi: 10.1056/NEJM199703273361303. [DOI] [PubMed] [Google Scholar]
- 10.ARDS Network. Ketoconazole for early treatment of acute lung injury and acute respiratory distress syndrome: a randomized controlled trial. JAMA. 2000;283:1995–2002. doi: 10.1001/jama.283.15.1995. [Published errata appear in JAMA 284:2450; 286:1578; 284:2597.] [DOI] [PubMed] [Google Scholar]
- 11.ARDS Clinical Trials Network; National Heart, Lung, and Blood Institute, National Institutes of Health. Randomized, placebo-controlled trial of lisofylline for early treatment of acute lung injury and acute respiratory distress syndrome. Crit Care Med. 2002;30:1–6. doi: 10.1097/00003246-200201000-00001. [DOI] [PubMed] [Google Scholar]
- 12.Gao Smith F, Perkins GD, Gates S, Young D, McAuley DF, Tunnicliffe W, et al. BALTI-2 study investigators. Effect of intravenous β-2 agonist treatment on clinical outcomes in acute respiratory distress syndrome (BALTI-2): a multicentre, randomised controlled trial. Lancet. 2012;379:229–235. doi: 10.1016/S0140-6736(11)61623-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ranieri VM, Thompson BT, Barie PS, Dhainaut JF, Douglas IS, Finfer S, et al. PROWESS-SHOCK Study Group. Drotrecogin alfa (activated) in adults with septic shock. N Engl J Med. 2012;366:2055–2064. doi: 10.1056/NEJMoa1202290. [DOI] [PubMed] [Google Scholar]
- 14.Iwashyna TJ, Burke JF, Sussman JB, Prescott HC, Hayward RA, Angus DC. Implications of heterogeneity of treatment effect for reporting and analysis of randomized trials in critical care. Am J Respir Crit Care Med. 2015;192:1045–1051. doi: 10.1164/rccm.201411-2125CP. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.McKown AC, Huerta LE, Rice TW, Semler MW Pragmatic Critical Care Research Group. Heterogeneity of treatment effect by baseline risk in a trial of balanced crystalloids versus saline. Am J Respir Crit Care Med. 2018;198:810–813. doi: 10.1164/rccm.201804-0680LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Heyland DK, Cook DJ, Griffith L, Keenan SP, Brun-Buisson C Canadian Critical Trials Group. The attributable morbidity and mortality of ventilator-associated pneumonia in the critically ill patient. Am J Respir Crit Care Med. 1999;159:1249–1256. doi: 10.1164/ajrccm.159.4.9807050. [DOI] [PubMed] [Google Scholar]
- 17.van Vught LA, Klein Klouwenberg PMC, Spitoni C, Scicluna BP, Wiewel MA, Horn J, et al. MARS Consortium. Incidence, risk factors, and attributable mortality of secondary infections in the intensive care unit after admission for sepsis. JAMA. 2016;315:1469–1479. doi: 10.1001/jama.2016.2691. [DOI] [PubMed] [Google Scholar]
- 18.Klein Klouwenberg PMC, Zaal IJ, Spitoni C, Ong DSY, van der Kooi AW, Bonten MJM, et al. The attributable mortality of delirium in critically ill patients: prospective cohort study. BMJ. 2014;349:g6652. doi: 10.1136/bmj.g6652. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Wood KA, Huang D, Angus DC. Improving clinical trial design in acute lung injury. Crit Care Med. 2003;31(Suppl):S305–S311. doi: 10.1097/01.CCM.0000057908.11686.B3. [DOI] [PubMed] [Google Scholar]
- 20.Rubenfeld GD. Who cares about preventing acute respiratory distress syndrome? Am J Respir Crit Care Med. 2015;191:255–260. doi: 10.1164/rccm.201408-1574CP. [DOI] [PubMed] [Google Scholar]
- 21.Harhay MO, Wagner J, Ratcliffe SJ, Bronheim RS, Gopal A, Green S, et al. Outcomes and statistical power in adult critical care randomized trials. Am J Respir Crit Care Med. 2014;189:1469–1478. doi: 10.1164/rccm.201401-0056CP. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Taylor AL, Ziesche S, Yancy C, Carson P, D’Agostino R, Jr, Ferdinand K, et al. African-American Heart Failure Trial Investigators. Combination of isosorbide dinitrate and hydralazine in blacks with heart failure. N Engl J Med. 2004;351:2049–2057. doi: 10.1056/NEJMoa042934. [DOI] [PubMed] [Google Scholar]
- 23.Constantin JM, Jabaudon M, Lefrant JY, Jaber S, Quenot JP, Langeron O, et al. AZUREA Network. Personalised mechanical ventilation tailored to lung morphology versus low positive end-expiratory pressure for patients with acute respiratory distress syndrome in France (the LIVE study): a multicentre, single-blind, randomised controlled trial. Lancet Respir Med. 2019;7:870–880. doi: 10.1016/S2213-2600(19)30138-9. [DOI] [PubMed] [Google Scholar]
- 24.Echt DS, Liebson PR, Mitchell LB, Peters RW, Obias-Manno D, Barker AH, et al. Mortality and morbidity in patients receiving encainide, flecainide, or placebo—the Cardiac Arrhythmia Suppression Trial. N Engl J Med. 1991;324:781–788. doi: 10.1056/NEJM199103213241201. [DOI] [PubMed] [Google Scholar]
- 25.Park JW, Liu MC, Yee D, Yau C, van ’t Veer LJ, Symmans WF, et al. I-SPY 2 Investigators. Adaptive randomization of neratinib in early breast cancer. N Engl J Med. 2016;375:11–22. doi: 10.1056/NEJMoa1513750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Guérin C, Reignier J, Richard JC, Beuret P, Gacouin A, Boulain T, et al. PROSEVA Study Group. Prone positioning in severe acute respiratory distress syndrome. N Engl J Med. 2013;368:2159–2168. doi: 10.1056/NEJMoa1214103. [DOI] [PubMed] [Google Scholar]
- 27.Papazian L, Forel JM, Gacouin A, Penot-Ragon C, Perrin G, Loundou A, et al. ACURASYS Study Investigators. Neuromuscular blockers in early acute respiratory distress syndrome. N Engl J Med. 2010;363:1107–1116. doi: 10.1056/NEJMoa1005372. [DOI] [PubMed] [Google Scholar]
- 28.National Heart, Lung, and Blood Institute PETAL Clinical Trials Network. Early neuromuscular blockade in the acute respiratory distress syndrome. N Engl J Med. 2019;380:1997–2008. doi: 10.1056/NEJMoa1901686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Seymour CW, Kennedy JN, Wang S, Chang CH, Elliott CF, Xu Z, et al. Derivation, validation, and potential treatment implications of novel clinical phenotypes for sepsis. JAMA. 2019;321:2003–2017. doi: 10.1001/jama.2019.5791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Woodruff PG, Modrek B, Choy DF, Jia G, Abbas AR, Ellwanger A, et al. T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med. 2009;180:388–395. doi: 10.1164/rccm.200903-0392OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wenzel SE. Asthma phenotypes: the evolution from clinical to molecular approaches. Nat Med. 2012;18:716–725. doi: 10.1038/nm.2678. [DOI] [PubMed] [Google Scholar]
- 32.Wenzel S, Ford L, Pearlman D, Spector S, Sher L, Skobieranda F, et al. Dupilumab in persistent asthma with elevated eosinophil levels. N Engl J Med. 2013;368:2455–2466. doi: 10.1056/NEJMoa1304048. [DOI] [PubMed] [Google Scholar]
- 33.Calfee CS, Delucchi K, Parsons PE, Thompson BT, Ware LB, Matthay MA NHLBI ARDS Network. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Goligher EC, Kavanagh BP, Rubenfeld GD, Adhikari NKJ, Pinto R, Fan E, et al. Oxygenation response to positive end-expiratory pressure predicts mortality in acute respiratory distress syndrome: a secondary analysis of the LOVS and ExPress trials. Am J Respir Crit Care Med. 2014;190:70–76. doi: 10.1164/rccm.201404-0688OC. [DOI] [PubMed] [Google Scholar]
- 35.Prewitt RM, McCarthy J, Wood LD. Treatment of acute low pressure pulmonary edema in dogs: relative effects of hydrostatic and oncotic pressure, nitroprusside, and positive end-expiratory pressure. J Clin Invest. 1981;67:409–418. doi: 10.1172/JCI110049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Osuchowski MF, Ayala A, Bahrami S, Bauer M, Boros M, Cavaillon JM, et al. Minimum quality threshold in pre-clinical sepsis studies (MQTiPSS): an international expert consensus initiative for improvement of animal modeling in sepsis. Intensive Care Med Exp. 2018;6:26. doi: 10.1186/s40635-018-0189-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Asmussen S, Ito H, Traber DL, Lee JW, Cox RA, Hawkins HK, et al. Human mesenchymal stem cells reduce the severity of acute lung injury in a sheep model of bacterial pneumonia. Thorax. 2014;69:819–825. doi: 10.1136/thoraxjnl-2013-204980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Park J, Kim S, Lim H, Liu A, Hu S, Lee J, et al. Therapeutic effects of human mesenchymal stem cell microvesicles in an ex vivo perfused human lung injured with severe E. coli pneumonia. Thorax. 2019;74:43–50. doi: 10.1136/thoraxjnl-2018-211576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Moazed F, Burnham EL, Vandivier RW, O’Kane CM, Shyamsundar M, Hamid U, et al. Cigarette smokers have exaggerated alveolar barrier disruption in response to lipopolysaccharide inhalation. Thorax. 2016;71:1130–1136. doi: 10.1136/thoraxjnl-2015-207886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gotts JE, Chun L, Abbott J, Fang X, Takasaka N, Nishimura SL, et al. Cigarette smoke exposure worsens acute lung injury in antibiotic-treated bacterial pneumonia in mice. Am J Physiol Lung Cell Mol Physiol. 2018;315:L25–L40. doi: 10.1152/ajplung.00405.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lefrançais E, Mallavia B, Zhuo H, Calfee CS, Looney MR. Maladaptive role of neutrophil extracellular traps in pathogen-induced lung injury. JCI Insight. 2018;3:e98178. doi: 10.1172/jci.insight.98178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Quinton LJ, Walkey AJ, Mizgerd JP. Integrative physiology of pneumonia. Physiol Rev. 2018;98:1417–1464. doi: 10.1152/physrev.00032.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Matute-Bello G, Downey G, Moore BB, Groshong SD, Matthay MA, Slutsky AS, et al. Acute Lung Injury in Animals Study Group. An official American Thoracic Society workshop report: features and measurements of experimental acute lung injury in animals. Am J Respir Cell Mol Biol. 2011;44:725–738. doi: 10.1165/rcmb.2009-0210ST. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kilkenny C, Parsons N, Kadyszewski E, Festing MFW, Cuthill IC, Fry D, et al. Survey of the quality of experimental design, statistical analysis and reporting of research using animals. PLoS One. 2009;4:e7824. doi: 10.1371/journal.pone.0007824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Avey MT, Moher D, Sullivan KJ, Fergusson D, Griffin G, Grimshaw JM, et al. Canadian Critical Care Translational Biology Group. The devil is in the details: incomplete reporting in preclinical animal research. PLoS One. 2016;11:e0166733. doi: 10.1371/journal.pone.0166733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol. 2010;8:e1000412. doi: 10.1371/journal.pbio.1000412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Gattinoni L, Caironi P, Cressoni M, Chiumello D, Ranieri VM, Quintel M, et al. Lung recruitment in patients with the acute respiratory distress syndrome. N Engl J Med. 2006;354:1775–1786. doi: 10.1056/NEJMoa052052. [DOI] [PubMed] [Google Scholar]
- 48.Chen L, Del Sorbo L, Grieco DL, Junhasavasdikul D, Rittayamai N, Soliman I, et al. Potential for lung recruitment estimated by the recruitment-to-inflation ratio in acute respiratory distress syndrome: a clinical trial. Am J Respir Crit Care Med. 2020;201:178–187. doi: 10.1164/rccm.201902-0334OC. [DOI] [PubMed] [Google Scholar]
- 49.McIntyre LA, Stewart DJ, Mei SHJ, Courtman D, Watpool I, Granton J, et al. Canadian Critical Care Trials Group; Canadian Critical Care Translational Biology Group. Cellular immunotherapy for septic shock: a phase I clinical trial. Am J Respir Crit Care Med. 2018;197:337–347. doi: 10.1164/rccm.201705-1006OC. [DOI] [PubMed] [Google Scholar]
- 50.Russell JA, Vincent JL, Kjølbye AL, Olsson H, Blemings A, Spapen H, et al. Selepressin, a novel selective vasopressin V1A agonist, is an effective substitute for norepinephrine in a phase IIa randomized, placebo-controlled trial in septic shock patients. Crit Care. 2017;21:213. doi: 10.1186/s13054-017-1798-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Young P, Hodgson C, Dulhunty J, Saxena M, Bailey M, Bellomo R, et al. ANZICS Clinical Trials Group. End points for phase II trials in intensive care: recommendations from the Australian and New Zealand Clinical Trials Group consensus panel meeting. Crit Care Resusc. 2012;14:211–215. [PubMed] [Google Scholar]
- 52.Gunther ML, Morandi A, Krauskopf E, Pandharipande P, Girard TD, Jackson JC, et al. VISIONS Investigation, VISualizing Icu SurvivOrs Neuroradiological Sequelae. The association between brain volumes, delirium duration, and cognitive outcomes in intensive care unit survivors: the VISIONS cohort magnetic resonance imaging study. Crit Care Med. 2012;40:2022–2032. doi: 10.1097/CCM.0b013e318250acc0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Morandi A, Rogers BP, Gunther ML, Merkle K, Pandharipande P, Girard TD, et al. VISIONS Investigation, VISualizing Icu SurvivOrs Neuroradiological Sequelae. The relationship between delirium duration, white matter integrity, and cognitive impairment in intensive care unit survivors as determined by diffusion tensor imaging: the VISIONS prospective cohort magnetic resonance imaging study. Crit Care Med. 2012;40:2182–2189. doi: 10.1097/CCM.0b013e318250acdc. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hughes CG, Patel MB, Brummel NE, Thompson JL, McNeil JB, Pandharipande PP, et al. Relationships between markers of neurologic and endothelial injury during critical illness and long-term cognitive impairment and disability. Intensive Care Med. 2018;44:345–355. doi: 10.1007/s00134-018-5120-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jackson JC, Morandi A, Girard TD, Merkle K, Graves AJ, Thompson JL, et al. VISualizing Icu SurvivOrs Neuroradiological Sequelae (VISIONS) Investigation. Functional brain imaging in survivors of critical illness: a prospective feasibility study and exploration of the association between delirium and brain activation patterns. J Crit Care. 2015;30:653, e1–e7. doi: 10.1016/j.jcrc.2015.01.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zenaide PV, Gusmao-Flores D. Biomarkers in septic encephalopathy: a systematic review of clinical studies. Rev Bras Ter Intensiva. 2013;25:56–62. doi: 10.1590/S0103-507X2013000100011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Martel ML, Klein LR, Miner JR, Cole JB, Nystrom PC, Holm KM, et al. A brief assessment of capacity to consent instrument in acutely intoxicated emergency department patients. Am J Emerg Med. 2018;36:18–23. doi: 10.1016/j.ajem.2017.06.043. [DOI] [PubMed] [Google Scholar]
- 58.Verceles AC, Bhatti W. The ethical concerns of seeking consent from critically ill, mechanically ventilated patients for research: a matter of possessing capacity or surrogate insight. Clin Ethics. 2018;13:107–111. doi: 10.1177/1477750918762350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Netzer G, Sullivan DR. Recognizing, naming, and measuring a family intensive care unit syndrome. Ann Am Thorac Soc. 2014;11:435–441. doi: 10.1513/AnnalsATS.201309-308OT. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Angus DC. Fusing randomized trials with big data: the key to self-learning health care systems? JAMA. 2015;314:767–768. doi: 10.1001/jama.2015.7762. [DOI] [PubMed] [Google Scholar]
- 61.Loudon K, Treweek S, Sullivan F, Donnan P, Thorpe KE, Zwarenstein M. The PRECIS-2 tool: designing trials that are fit for purpose. BMJ. 2015;350:h2147. doi: 10.1136/bmj.h2147. [DOI] [PubMed] [Google Scholar]
- 62.Wang HE, Schmicker RH, Daya MR, Stephens SW, Idris AH, Carlson JN, et al. Effect of a strategy of initial laryngeal tube insertion vs endotracheal intubation on 72-hour survival in adults with out-of-hospital cardiac arrest: a randomized clinical trial. JAMA. 2018;320:769–778. doi: 10.1001/jama.2018.7044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Self WH, Semler MW, Wanderer JP, Wang L, Byrne DW, Collins SP, et al. SALT-ED Investigators. Balanced crystalloids versus saline in noncritically ill adults. N Engl J Med. 2018;378:819–828. doi: 10.1056/NEJMoa1711586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Segal JB, Weiss C, Varadhan R. Understanding heterogeneity of treatment effects in pragmatic trials with an example of a large, simple trial of a drug treatment for osteoporosis [accessed 2019 Apr 12] Available from: http://www.cmtpnet.org/docs/resources/Segal-Heterogeneity-in-Pragmatic-Trials.pdf.
- 65.Wayne MT, Valley TS, Cooke CR, Sjoding MW. Electronic “sniffer” systems to identify the acute respiratory distress syndrome. Ann Am Thorac Soc. 2019;16:488–495. doi: 10.1513/AnnalsATS.201810-715OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Fiore LD, Brophy M, Ferguson RE, D’Avolio L, Hermos JA, Lew RA, et al. A point-of-care clinical trial comparing insulin administered using a sliding scale versus a weight-based regimen. Clin Trials. 2011;8:183–195. doi: 10.1177/1740774511398368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Semler MW, Weavind L, Hooper MH, Rice TW, Gowda SS, Nadas A, et al. An electronic tool for the evaluation and treatment of sepsis in the ICU: a randomized controlled trial. Crit Care Med. 2015;43:1595–1602. doi: 10.1097/CCM.0000000000001020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Semler MW, Self WH, Wanderer JP, Ehrenfeld JM, Wang L, Byrne DW, et al. SMART Investigators and the Pragmatic Critical Care Research Group. Balanced crystalloids versus saline in critically ill adults. N Engl J Med. 2018;378:829–839. doi: 10.1056/NEJMoa1711584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Institute for Healthcare Improvement. Cambridge, MA: Institute for Healthcare Improvement [accessed 2019 Feb 28]; IHI intensive care unit (ICU) adverse event trigger tool. Available from: http://www.ihi.org:80/resources/Pages/Tools/ICUAdverseEventTriggerTool.aspx. [Google Scholar]
- 70.Lanspa MJ, Gong MN, Schoenfeld DA, Lee KT, Grissom CK, Hou PC, et al. National Heart, Lung, and Blood Institute Prevention and Early Treatment of Acute Lung Injury (PETAL) Clinical Trials Network. Prospective assessment of the feasibility of a trial of low-tidal volume ventilation for patients with acute respiratory failure. Ann Am Thorac Soc. 2019;16:356–362. doi: 10.1513/AnnalsATS.201807-459OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Grumbach K, Lucey CR, Johnston SC. Transforming from centers of learning to learning health systems: the challenge for academic health centers. JAMA. 2014;311:1109–1110. doi: 10.1001/jama.2014.705. [DOI] [PubMed] [Google Scholar]
- 72.Biros M. Capacity, vulnerability, and informed consent for research. J Law Med Ethics. 2018;46:72–78. doi: 10.1177/1073110518766021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Faden RR, Kass NE, Goodman SN, Pronovost P, Tunis S, Beauchamp TL. An ethics framework for a learning health care system: a departure from traditional research ethics and clinical ethics. Hastings Cent Rep. 2013;43(Spec No):S16–S27. doi: 10.1002/hast.134. [DOI] [PubMed] [Google Scholar]
- 74.Burns KEA, Devlin JW, Hill NS. Patient and family engagement in designing and implementing a weaning trial: a novel research paradigm in critical care. Chest. 2017;152:707–711. doi: 10.1016/j.chest.2017.06.028. [DOI] [PubMed] [Google Scholar]
- 75.Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med. 2013;369:840–851. doi: 10.1056/NEJMra1208623. [DOI] [PubMed] [Google Scholar]
- 76.Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3) JAMA. 2016;315:801–810. doi: 10.1001/jama.2016.0287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Seymour CW, Gesten F, Prescott HC, Friedrich ME, Iwashyna TJ, Phillips GS, et al. Time to treatment and mortality during mandated emergency care for sepsis. N Engl J Med. 2017;376:2235–2244. doi: 10.1056/NEJMoa1703058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.PEPTIC Investigators for the Australian and New Zealand Intensive Care Society Clinical Trials Group, Alberta Health Services Critical Care Strategic Clinical Network, and the Irish Critical Care Trials Group. Effect of stress ulcer prophylaxis with proton pump inhibitors vs histamine-2 receptor blockers on in-hospital mortality among ICU patients receiving invasive mechanical ventilation: the PEPTIC randomized clinical trial. JAMA. 2020;323:616–626. doi: 10.1001/jama.2019.22190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hernán MA, Robins JM. Using big data to emulate a target trial when a randomized trial is not available. Am J Epidemiol. 2016;183:758–764. doi: 10.1093/aje/kwv254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Gershon AS, Jafarzadeh SR, Wilson KC, Walkey AJ. Clinical knowledge from observational studies: everything you wanted to know but were afraid to ask. Am J Respir Crit Care Med. 2018;198:859–867. doi: 10.1164/rccm.201801-0118PP. [DOI] [PubMed] [Google Scholar]
- 81.Dinglas VD, Faraone LN, Needham DM. Understanding patient-important outcomes after critical illness: a synthesis of recent qualitative, empirical, and consensus-related studies. Curr Opin Crit Care. 2018;24:401–409. doi: 10.1097/MCC.0000000000000533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Hashem MD, Nallagangula A, Nalamalapu S, Nunna K, Nausran U, Robinson KA, et al. Patient outcomes after critical illness: a systematic review of qualitative studies following hospital discharge. Crit Care. 2016;20:345. doi: 10.1186/s13054-016-1516-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Dinglas VD, Chessare CM, Davis WE, Parker A, Friedman LA, Colantuoni E, et al. Perspectives of survivors, families and researchers on key outcomes for research in acute respiratory failure. Thorax. 2018;73:7–12. doi: 10.1136/thoraxjnl-2017-210234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Turnbull AE, Sepulveda KA, Dinglas VD, Chessare CM, Bingham CO, III, Needham DM. Core domains for clinical research in acute respiratory failure survivors: an international modified Delphi consensus study. Crit Care Med. 2017;45:1001–1010. doi: 10.1097/CCM.0000000000002435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Needham DM, Sepulveda KA, Dinglas VD, Chessare CM, Friedman LA, Bingham CO, III, et al. Core outcome measures for clinical research in acute respiratory failure survivors: an international modified Delphi consensus study. Am J Respir Crit Care Med. 2017;196:1122–1130. doi: 10.1164/rccm.201702-0372OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Colantuoni E, Scharfstein DO, Wang C, Hashem MD, Leroux A, Needham DM, et al. Statistical methods to compare functional outcomes in randomized controlled trials with high mortality. BMJ. 2018;360:j5748. doi: 10.1136/bmj.j5748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Connolly B, Denehy L, Hart N, Pattison N, Williamson P, Blackwood B. Physical Rehabilitation Core Outcomes In Critical illness (PRACTICE): protocol for development of a core outcome set. Trials. 2018;19:294. doi: 10.1186/s13063-018-2678-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Morris PE, Berry MJ, Files DC, Thompson JC, Hauser J, Flores L, et al. Standardized rehabilitation and hospital length of stay among patients with acute respiratory failure: a randomized clinical trial. JAMA. 2016;315:2694–2702. doi: 10.1001/jama.2016.7201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Moss M, Nordon-Craft A, Malone D, Van Pelt D, Frankel SK, Warner ML, et al. A randomized trial of an intensive physical therapy program for patients with acute respiratory failure. Am J Respir Crit Care Med. 2016;193:1101–1110. doi: 10.1164/rccm.201505-1039OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Huang DT, Angus DC, Moss M, Thompson BT, Ferguson ND, Ginde A, et al. Reevaluation of Systemic Early Neuromuscular Blockade Protocol Committee and the National Institutes of Health National Heart, Lung, and Blood Institute Prevention and Early Treatment of Acute Lung Injury Network Investigators. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Prescott HC, Carmichael AG, Langa KM, Gonzalez R, Iwashyna TJ. Paths into sepsis: trajectories of presepsis healthcare use. Ann Am Thorac Soc. 2019;16:116–123. doi: 10.1513/AnnalsATS.201806-391OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Cox CE, Hough CL, Jones DM, Ungar A, Reagan W, Key MD, et al. Effects of mindfulness training programmes delivered by a self-directed mobile app and by telephone compared with an education programme for survivors of critical illness: a pilot randomised clinical trial. Thorax. 2019;74:33–42. doi: 10.1136/thoraxjnl-2017-211264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.ProCESS Investigators. A randomized trial of protocol-based care for early septic shock. N Engl J Med. 2014;370:1683–1693. doi: 10.1056/NEJMoa1401602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.El Bcheraoui C, Mokdad AH, Dwyer-Lindgren L, Bertozzi-Villa A, Stubbs RW, Morozoff C, et al. Trends and patterns of differences in infectious disease mortality among US counties, 1980-2014. JAMA. 2018;319:1248–1260. doi: 10.1001/jama.2018.2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Yende S, D’Angelo G, Mayr F, Kellum JA, Weissfeld L, Kaynar AM, et al. GenIMS Investigators. Elevated hemostasis markers after pneumonia increases one-year risk of all-cause and cardiovascular deaths. PLoS One. 2011;6:e22847. doi: 10.1371/journal.pone.0022847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Yende S, D’Angelo G, Kellum JA, Weissfeld L, Fine J, Welch RD, et al. GenIMS Investigators. Inflammatory markers at hospital discharge predict subsequent mortality after pneumonia and sepsis. Am J Respir Crit Care Med. 2008;177:1242–1247. doi: 10.1164/rccm.200712-1777OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Ramirez JA, Wiemken TL, Peyrani P, Arnold FW, Kelley R, Mattingly WA, et al. University of Louisville Pneumonia Study Group. Adults hospitalized with pneumonia in the United States: incidence, epidemiology, and mortality. Clin Infect Dis. 2017;65:1806–1812. doi: 10.1093/cid/cix647. [DOI] [PubMed] [Google Scholar]
- 98.Dela Cruz CS, Wunderink RG, Christiani DC, Cormier SA, Crothers K, Doerschuk CM, et al. Future research directions in pneumonia: NHLBI working group report. Am J Respir Crit Care Med. 2018;198:256–263. doi: 10.1164/rccm.201801-0139WS. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Beura LK, Hamilton SE, Bi K, Schenkel JM, Odumade OA, Casey KA, et al. Normalizing the environment recapitulates adult human immune traits in laboratory mice. Nature. 2016;532:512–516. doi: 10.1038/nature17655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Basil MC, Levy BD. Specialized pro-resolving mediators: endogenous regulators of infection and inflammation. Nat Rev Immunol. 2016;16:51–67. doi: 10.1038/nri.2015.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Musher DM, Rueda AM, Kaka AS, Mapara SM. The association between pneumococcal pneumonia and acute cardiac events. Clin Infect Dis. 2007;45:158–165. doi: 10.1086/518849. [DOI] [PubMed] [Google Scholar]
- 102.Krishnamoorthy N, Abdulnour REE, Walker KH, Engstrom BD, Levy BD. Specialized proresolving mediators in innate and adaptive immune responses in airway diseases. Physiol Rev. 2018;98:1335–1370. doi: 10.1152/physrev.00026.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Serhan CN, Levy BD. Resolvins in inflammation: emergence of the pro-resolving superfamily of mediators. J Clin Invest. 2018;128:2657–2669. doi: 10.1172/JCI97943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Laterre PF, Garber G, Levy H, Wunderink R, Kinasewitz GT, Sollet JP, et al. PROWESS Clinical Evaluation Committee. Severe community-acquired pneumonia as a cause of severe sepsis: data from the PROWESS study. Crit Care Med. 2005;33:952–961. doi: 10.1097/01.ccm.0000162381.24074.d7. [DOI] [PubMed] [Google Scholar]
- 105.Vincent JL, Akça S, De Mendonça A, Haji-Michael P, Sprung C, Moreno R, et al. SOFA Working Group. The epidemiology of acute respiratory failure in critically ill patients. Chest. 2002;121:1602–1609. doi: 10.1378/chest.121.5.1602. [DOI] [PubMed] [Google Scholar]
- 106.Ashbaugh DG, Bigelow DB, Petty TL, Levine BE.Acute respiratory distress in adults Lancet 19672319–323.4143721 [Google Scholar]
- 107.Matthay MA, Zemans RL, Zimmerman GA, Arabi YM, Beitler JR, Mercat A, et al. Acute respiratory distress syndrome. Nat Rev Dis Primers. 2019;5:18. doi: 10.1038/s41572-019-0069-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Thompson BT, Chambers RC, Liu KD. Acute respiratory distress syndrome. N Engl J Med. 2017;377:562–572. doi: 10.1056/NEJMra1608077. [DOI] [PubMed] [Google Scholar]
- 109.Parsons PE, Eisner MD, Thompson BT, Matthay MA, Ancukiewicz M, Bernard GR, et al. NHLBI Acute Respiratory Distress Syndrome Clinical Trials Network. 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. [Discussion, pp. 230–232] [DOI] [PubMed] [Google Scholar]
- 110.National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network. Comparison of two fluid-management strategies in acute lung injury. N Engl J Med. 2006;354:2564–2575. doi: 10.1056/NEJMoa062200. [DOI] [PubMed] [Google Scholar]
- 111.Beitler JR, Goligher EC, Schmidt M, Spieth PM, Zanella A, Martin-Loeches I, et al. ARDSne(x)t Investigators. Personalized medicine for ARDS: the 2035 research agenda. Intensive Care Med. 2016;42:756–767. doi: 10.1007/s00134-016-4331-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
