In this review, we highlight the major scientific output in 2020 related to critical care in American Thoracic Society journals. Scientific focus surrounded acute respiratory distress syndrome (ARDS), sepsis, acute kidney injury (AKI), ICU organization, post-ICU recovery, critical care research, and education. This review focuses predominantly on original research articles and related commentary, but there were also several excellent review articles (1–3) and letters to the editor (4–12) related to critical care. Furthermore, there is a dedicated update on coronavirus disease (COVID-19), which also contains pertinent information related to critical care.
ARDS
ARDS Pathogenesis
The identification of subphenotypes of ARDS has led to a more nuanced approach to clinical trials, but biomarkers to identify subphenotypes are often not available for clinical use. In a secondary analysis of the NHLBI ARDS Network clinical trials, a machine learning technique called “gradient-boosted machines” using routinely available clinical laboratory parameters reliably reidentified hyperinflammatory and hypoinflammatory subphenotypes initially identified using the latent classifier approach (13, 14). Genetic predisposition to ARDS may be an important factor for future clinical trials. In a Mendelian randomization study on two well-phenotyped cohorts of critically ill patients with sepsis and ARDS, plasma sRAGE was genetically determined and identified as a potential causal factor in sepsis-associated ARDS (15). Logical next steps would include studying patients at risk for ARDS (16).
The renin–angiotensin system (RAS) has been implicated in the pathogenesis of ARDS. A normal angiotensin I–to–angiotensin II ratio was inverted in individuals with early ARDS but not in surgical control subjects, and it was associated with poor outcomes in patients with ARDS (17). In addition, biomarkers of the alternative RAS pathway stimulated by neprilysin, Ang 1–7, and Ang 1–5 were higher in patients with ARDS than in control subjects, and RAS dysregulation was noted to persist in later ARDS.
Lung repair after ARDS continued to be a focus in preclinical studies. Regulatory T cells (Tregs) are elevated in human blood and BAL fluid from patients with ARDS; however, the mechanism by which Tregs alter alveolar type II IFN responses remains unclear. Using gene ontology analysis in an animal model of ARDS, gene sets composing type I IFN, intracellular transport, and cellular localization were upregulated in the Treg-depleted samples (18). Tregs may be a potential therapeutic target in the resolution phase of ARDS (19). Permissive hypercapnia may have unintended consequences for cellular metabolism and may slow down wound healing in epithelial monolayers through the inhibition of Rac1/Cofilin via AMPK (20). Upregulation of NF-κB suppressed CXCL12 and activated Rac1 via the binding to CXCR4 and prevented the adverse effects of hypercapnia in an orthotopic tracheal transplantation model of wound healing (20).
Lung-Protective Ventilation and Ventilator-induced Lung Injury
Positive end-expiratory pressure (PEEP) management in ARDS remains controversial, with a heterogenous response being based on lung recruitability. Through a physiologic study in 45 adults with ARDS, a single-breath method to assess recruitability by calculating the recruitment-to-inflation ratio provided a simple way to identify patients who may benefit from a higher-PEEP strategy (21, 22). The addition of expiratory resistance in both spontaneously breathing and mechanically ventilated pigs with lung injury resulted in less heterogeneous expiratory time constants, preventing end-expiratory lung collapse. The method may provide an alternative or adjuvant approach to PEEP management to prevent atelectrauma and regional overdistention in heterogenous lung disease (23, 24).
A pilot study demonstrated that a strategy to limit driving pressure in adult patients with ARDS, compared with conventional low Vt ventilation, was feasible (25). Although providing a foundation for future trials, deep sedation or paralysis was required and may limit its widespread use (26). Mechanistically, stretch-related ventilator-induced lung injury may be decreased by the activation of Piezo1, a nonselective cation channel that allows the influx of calcium and sodium into endothelial cells, stabilizes endothelial adherens junctions and lung vascular barriers (27), and activates calpain (28).
Patient Self-inflicted Lung Injury and Ventilator-induced Diaphragm Dysfunction
Although ventilator-induced lung injury has been well recognized over the last several decades, it is becoming increasingly clear that lung injury can occur when patients have a strong respiratory drive, a term collectively known as patient self-inflicted lung injury (P-SILI). P-SILI is exacerbated when patients achieve high Vts with correspondingly high transpulmonary pressure, particularly when lung compliance is impaired. Strategies have been developed to identify patients with significant respiratory effort who are at high risk of P-SILI, both on invasive and noninvasive mechanical ventilation. It is also important to identify mechanically ventilated patients with very low respiratory effort, as this is a risk factor for ventilator-induced diaphragm dysfunction.
Through secondary analysis of 132 mechanically ventilated patients with moderate-to-severe ARDS, neuromuscular blockade was only beneficial in the subgroup with higher baseline diaphragm thickness and was harmful in the subgroup with lower baseline diaphragm thickness, suggesting that patients with higher baseline thickness may have the strength to generate more injurious transpulmonary pressures and are at higher risk of P-SILI (29).
Airway occlusion maneuvers during expiration or inspiration can be used to estimate patient effort or driving pressure. As compared with esophageal pressure, excessively high or low respiratory effort can be reliably detected using the ventilator-calculated change in pressure during the first 100 ms of a breath during airway occlusion (30, 31). During assisted ventilation on pressure support, driving pressure measured during inspiratory or expiratory airway occlusion maneuvers are highly correlated with one another. Specifically, the negative deflection in airway pressure during an expiratory hold multiplied by 0.75 plus the set pressure support level represents the dynamic driving pressure. This dynamic driving pressure was found to be nearly identical to the static driving pressure measured as the difference between the plateau pressure during an inspiratory hold and the PEEP level, after controlling for the airway resistance.
Tissue doppler imaging of the diaphragm using ultrasound is a novel technique to monitor respiratory effort and identify patients likely to experience weaning failure. The technique was highly reproducible and identified differences between patients with weaning failure and patients with weaning success or healthy volunteers, and parameters were correlated with gold standard measures of respiratory effort, such as the transdiaphragmatic pressure time product (33, 34). Electrical activity of the diaphragm, often used to measure patient effort and gauge patient–ventilator synchrony, may have limitations in detecting ineffective respiratory effort, as compared with esophageal manometry or surface electromyography (35).
In patients with hypoxemic respiratory failure treated with noninvasive ventilation (NIV), strong spontaneous breathing effort (large swings in esophageal pressure) were associated with worsening of lung injury on chest radiographs and an increased risk of intubation. Minimal reduction in esophageal pressure swings (<10 cm H2O) after 2 hours of NIV was more strongly associated with intubation than large expiratory Vts; the Heart Rate, Acidosis, Consciousness, Oxygenation, and Respiratory Rate score; or other physiologic parameters (36, 37). Higher pressure support was strongly associated with decreasing spontaneous breathing effort (38).
In a physiologic crossover study on 15 patients with acute hypoxemic respiratory failure, use of high-PEEP helmet NIV was associated with improved oxygenation and work of breathing compared with use of a high-flow nasal canula (HFNC). Patients with high inspiratory effort during HFNC use were more likely to have improvement with helmet NIV use, but patients with low inspiratory effort during HFNC use had increased transpulmonary pressure swings during helmet NIV use, which may have resulted in larger and harmful Vts (39, 40). Although Vt is generally not measurable during helmet NIV use, it may be accurately measured by using a turbine-driven ventilator connected to the inspiratory port of the helmet by using a single-limb circuit with an intentional calibrated leak from the expiratory port of the helmet (41).
Adjuvant ARDS Therapies and Extracorporeal Membrane Oxygenation
Mesenchymal stem cells have been a recent focus in ARDS. The 1-year and 5-year follow-up results of two patients who had received a single intravenous infusion of allogenic bone marrow–derived mesenchymal stromal cells were reported and showed full resolution of lung parenchymal injuries on computed tomography scans (42). In an animal model of ARDS treated with extracorporeal membrane oxygenation (ECMO), endobronchial infusion of induced pluripotent stem cell–derived human mesenchymal stem cells was associated with decreased lung inflammation, histologic lung injury, and shock severity. However, no differences in oxygenation or respiratory mechanics were observed, and stem cells were associated with pulmonary arterial thromboses and impairment of the membrane oxygenator (43, 44).
Adjuvant therapies for ARDS are used with significant variability among institutions. An observational study conducted in 100 international pediatric ICUs identified that continuous neuromuscular blockade was used in 30% of patients with pediatric ARDS and in 50% of patients with severe pediatric ARDS, whereas the remainder of other adjuvant therapies were used less than 15% of the time. There were significant regional and institutional differences in the application of adjuvant therapies and their order of implementation (45, 46). Some of these institutional differences may relate to comfort with the therapy and training, particularly for prone positioning. According to a survey conducted in 54 acute care hospitals in Massachusetts, only 44% of hospitals were “prone ready,” and the absence of a protocol for prone positioning was associated with the use of other therapies (47).
Data from the Extracorporeal Life Support Organization registry suggested that a relative drop in PaCO2 > 50% in the first 24 hours of ECMO was associated with a 70% increased risk of neurologic complications. These findings suggest that PaCO2 or the related cerebral vasoconstriction should be monitored after ECMO initiation to guide sweep gas flow (48, 49). Interestingly, Bourcier and colleagues (50) reported favorable outcomes in four out of five young patients with refractory obstructive shock secondary to mediastinal compressive tumors with ECMO and chemotherapy.
Translational and Preclinical Studies on the Lung
Electronic cigarette– or vaping-associated lung injury is a recently recognized cause of ARDS in young individuals. Vitamin E acetate caused dose-dependent increases in pulmonary edema as well as organizing and lipoid pneumonia in a mouse model of electronic cigarette– or vaping-associated lung injury through the upregulation of the gene expression of inflammatory pathways and through a direct toxic effect on alveolar type II cells (51). α-Tocopherol, a metabolite of Vitamin E acetate is a free radical scavenger that has been proposed to treat ARDS. Pretreatment of endothelial and epithelial cells with α-tocopherol reduced paracellular permeability induced by Pseudomonas aeruginosa exoenzyme (type III secretion system) and prevented mortality in a mouse model of P. aeruginosa pneumonia (52).
Severe meconium aspiration syndrome (MAS) is a neonatal form of ARDS. A translational study using nonbronchoscopic BAL fluid from neonates with MAS compared with that of control subjects found that MAS was associated with significant changes in the surfactant phospholipid profile, with an increase in lysophosphatidylcholine and surfactant proteins B and C being shown. In addition, MAS was also associated with alteration of surfactants’ nanostructure, possibly due to concurrent lung inflammation (53).
Viral and bacterial products activate the innate immune system through TLR (Toll-like receptor) signaling. TLR8 protein levels were lower in blood samples of patients with ARDS compared with blood samples of healthy control subjects. The authors discovered that E3 ligase RNF216 (ring finger protein 216) ubiquitinates TLR8 in response to microRNAs. Gene expression profiles obtained from patients with ARDS had reduced RNF216. Furthermore, RNA extracted from plasma of human ARDS samples activated TLR8 in a macrophage cell line in vitro, suggesting that TLR8–RNF216 signaling plays an important role in ARDS (54) and that the hyperinflammatory phenotype is perhaps due to reduced RNF216 (55). The time course of immune system activation is critical in the development of ARDS. Harnessing epithelial antiviral properties has the potential to decrease the pulmonary viral burden and hence the occurrence of ARDS. Inhaled Pam2-ODN, a TLR 2/6 and 9 agonist, reduced the lung Sendai virus (the murine form of the human parainfluenza virus) burden and decreased mortality in mice. In addition, Pam2-ODN blocked the increase in CD8+ T cells induced by viral challenge, leading to reduced mortality (56). However, the mechanism for viral inactivation due to Pam2-ODN and its effects on established viral pneumonia are unknown (57).
Respiratory viral infections are common in children and can be associated with poor outcomes. In neonates with respiratory syncytial virus, higher disease severity was related to CD8+ T cells expressing IL-4 (Tc2) and reduced proportions of CD8+ T cells expressing IFNg (Tc1) in nasal aspirates. Less severe disease was associated with greater frequencies of Tc1, CD8+ T cells expressing IL-17 (Tc17), and CD4+ T cells expressing IL-17 (Th17), highlighting the importance of T-cell subsets in the pathogenesis and severity of respiratory syncytial viral infection (58). The nasal washing cytokine composition of pediatric allogeneic hematopoietic cell-transplant patients in the absence of viral respiratory tract infection is different from that of healthy control subjects and produced higher concentrations of IL-4, IL-12p40, and IL-1ra during a viral infection (59).
The lung microbiome was a focus of several investigations. Using droplet digital PCR and bacterial 16S quantification and sequencing in mini-BAL fluid before the receipt of antibiotics, differences in the lung microbiome were predictive of ventilator-free days and were associated with the diagnosis of ARDS (60). Enterobacteriaceae predominance was a risk factor for poor outcomes (61). Similarly, three clusters of endotracheal aspirate flora and two clusters of oral flora were identified in ICU patients with respiratory failure before administration of antibiotics. Individual clusters were associated with bacterial diversity, the prevalence of pathogenic bacteria, inflammation, and outcomes (62). Together, these findings suggest that the alteration of the airway microbiome of ICU patients may be a modifiable factor or therapeutic target (63).
The incidence of bronchopulmonary dysplasia is increasing. Maternal vitamin D deficiency impaired lung development in rats, causing outcomes including a simplified distal lung structure and higher resistance and airway hyperactivity in experimental animals compared with control animals (64). These changes mimicked those seen after postnasal hyperoxia exposure and may be explained by an abnormal reduction of HIF-1α/VEGF signaling (64, 65). Basal autophagy is impaired during neonatal hyperoxia-induced lung injury and in a baboon model of bronchopulmonary dysplasia. Autophagy-deficient mice had reduced alveolarization, increased expression of proinflammatory genes, and enhanced NLRP3 inflammasome activation in alveolar macrophages, increasing their susceptibility for neonatal hyperoxia-induced lung injury (66).
Other Studies of Mechanical Ventilation
Ciliary dyskinesia was common in mechanically ventilated patients suspected of having pneumonia and was associated with the severity of respiratory failure but not with ventilator-associated events or outcomes (67). The prevalence of probable Aspergillus infection using the algorithm proposed by the AspICU investigators (68) was as high as 12% in patients without neutropenia who had suspected ventilator-associated pneumonia and was associated with longer ICU stays but was not associated with higher mortality (69, 70). In contrast, invasive tracheobronchial aspergillosis, as suggested by the presence of endotracheobronchial lesions in patients with Aspergillus infection, was diagnosed in 29% of patients with severe influenza and pulmonary aspergillosis, and their in-ICU mortality rate was 90% despite antifungal treatment (71, 72).
From 2010 to 2017, NIV was used as the first-line treatment of asthma exacerbation in 25% of cases and increased over time. Although it failed in 22% of cases, use of NIV was independently associated with lower risks of intubation and mortality (73, 74). On the basis of administrative diagnosis data of ICU admissions in Australia and New Zealand, it was shown that the percentage of ICU admissions with acute exacerbations of chronic obstructive pulmonary disease increased between 2005 and 2017 but that mortality decreased (75, 76).
Among patients with respiratory failure requiring mechanical ventilation in Taiwan, more than 90% of successful weaning outcomes occurred in the first 30 days of mechanical ventilation, and the probability of death surpassed that of weaning after the 27th ventilator day (77). It remains to be seen whether the online prognosis tool derived from this study informs shared decision-making and whether these results are generalizable (78). The characteristics of high-performing centers specialized in the management of patients with prolonged mechanical ventilation included hospital leadership engagement in quality improvement, adequate staffing, detailed yet flexible protocols to increase the autonomy of bedside providers, and interdisciplinary team meetings. Key factors associated with effective care included the coordination among caregivers to balance aggressiveness and responsiveness and the involvement of patients and their families in decision-making (79, 80).
Delirium is common among ICU patients, particularly among those on mechanical ventilation. In a secondary analysis of two randomized controlled trials on delirium management, five trajectories of delirium, integrating symptom severity and duration, were identified as being associated with patient characteristics and 30-day mortality (81).
In ventilated patients, the optimal nutritional target remains as a subject of debate because delivery of 100% or 70% of the recommended calorie intake in the acute phase of critical illness did not result in significant differences in the long-term mortality, disability, quality of life, or return-to-work rate of survivors after 6 months (82, 83).
Sepsis
Clinical Studies in Sepsis
Hydrocortisone, high-dose ascorbic acid, and thiamine therapy has been considered in septic shock (84). From 2015 to 2018, its use in adults admitted to the ICU for septic shock increased from use in 0.03% of patients to use in over 2.5% of patients. There was marked heterogeneity in its use among centers, with use in up to 20% of patients in some hospitals, especially in patients with more severe organ dysfunction. In a retrospective cohort analysis in adults, its use was associated with worse outcomes despite extensive multivariable modeling and propensity matching (85). In contrast, in a single-institution pediatric study, hydrocortisone, high-dose ascrobic acid, and thiamine therapy were associated with improved clinical outcomes when compared to control therapy after propensity score-matched and inverse probability-weighted analyses (86). Differences in patient selection, criteria for allocation of treatment, and unmeasured confounding likely contribute to the seemingly conflicting results of these two studies. Interestingly, baseline levels of cortisol, aldosterone, or ascorbic acid were not related to the treatment effect of hydrocortisone on mortality, shock resolution, or other clinical outcomes in a secondary analysis of the Adjunctive Corticosteroid Treatment in Critically Ill Patients With Septic Shock (ADRENAL) trial, a large, multinational, randomized, placebo-controlled trial of stress steroids in septic shock (87, 88).
In a secondary analysis of the randomized controlled trial of exogenous angiotensin II in vasodilatory shock (ATHOS-3) (89), the response to treatment was heterogeneous. Higher renin levels were associated with kidney injury and with a higher angiotensin I/II ratio (90). Compared with placebo treatment, treatment with exogenous angiotensin II improved outcomes in the subgroup of patients with higher renin levels (91).
Through a Bayesian reanalysis of the randomized clinical trial comparing peripheral perfusion to a lactate-clearance resuscitation strategy (ANDROMEDA-SHOCK), there was a >90% likelihood that the peripheral perfusion strategy was better, using a wide range of assumptions about the prior probability. Bayesian networks were also used to analyze patterns of change in Sequential Organ Failure Assessment (SOFA) scores, with death as a competing risk (92, 93).
Premorbid (baseline) blood pressure influenced the duration of vasopressor medication use in the ICU (94). Compared with normal premorbid blood pressure, a lower premorbid blood pressure was associated with a longer duration of vasopressor use and higher mortality, whereas higher premorbid blood pressure was associated with a shorter duration of vasopressor use, suggesting that differential blood pressure targets stratified by premorbid blood pressure should be evaluated (95). In addition, the depth and duration of the blood pressure deficit during critical illness compared with a premorbid measurement was associated with risk of a major adverse kidney event (96, 97).
Methods to improve delivery of care to patients with sepsis was a focus. Compliance with a 3-hour sepsis treatment bundle varied between daytime and nighttime and between emergency and nonemergency areas, suggesting the need to incorporate the setting of diagnosis into quality-improvement initiatives (98). In a pilot trial, coated devices (endotracheal tubes, central venous catheters, and urinary catheters) were found to be possibly associated with more days alive and free of antibiotics (99). The provision of palliative care consultation for older patients who died with a diagnosis of septic shock was associated with shorter lengths of stay and lower hospital costs compared with no provision of palliative care (100). Sepsis-associated mortality rates improved in patients with cancer but not in patients without cancer (101), highlighting the need to understand the interplay between cancer and sepsis and the validity of administrative data surrounding cancer and sepsis (101, 102). In selected patients labeled as being allergic to penicillin who had a history of low-risk clinical manifestations, direct oral amoxicillin challenge without prior desensitization or skin testing was shown to be safe and allowed for the removal of inaccurate penicillin allergy labels (103).
Diagnostic tools in sepsis may need modification to achieve optimal performance in resource-limited settings. In a prospective study on patients with suspected infection in Brazil, a quick SOFA (qSOFA) score ≥1 predicted mortality with a sensitivity of 84.9% (vs. 53.9% for a qSOFA score ≥2). In resource-limited settings, using a SOFA score ≥1 or using a qSOFA score ≥1 plus the lactate status to identify any organ dysfunction could be alternatives to the usual method of identification using a qSOFA score ≥2 (104, 105). In patients with sepsis hospitalized in Malawi, sonographic lung B-lines indicating supleural interstitial edema were strongly associated with eventual hypoxemia (area under the curve, 0.86 [95% confidence interval, 0.69–0.97]), with a dose–response increase in the number of B-lines and a dose–response decrease in oxygen saturation occurring with each liter of intravenous crystalloid administered (106).
Translational Studies in Sepsis
Compared with placebo and vasopressin administration, norepinephrine administration increased IL-10 levels in human leukocytes, increased bacterial growth after cecal ligation in a mouse model, and altered the cytokine response to an LPS challenge in healthy patients (107). However, the immune balance of patients admitted with sepsis remains incompletely understood, and large randomized trials have not demonstrated increased harm with norepinephrine administration (108).
In a proof-of-concept study in pediatric patients with sepsis, a microfluidic multianalyte system had acceptable agreement with biomarkers measured with standard ELISA techniques, which were associated with mortality. This may facilitate nearly real-time assessment of biomarker profiles for precision medicine approaches and trials (109).
Analysis of the differential RNA expression in the cortical gray matter of older patients with dementia who died of infectious versus noninfectious causes revealed clusters of gene transcripts that were associated with infection, including classical innate immune-related genes, damage-associated molecular patterns, complement factors, and cytokines (110). These results provide new targets for sepsis-associated brain injury research.
Higher levels of alternative complement pathway activity in the serum of critically ill patients were associated with higher levels of other complement-associated proteins, a hypoinflammatory phenotype, and better outcomes (111). Differential complement activation may represent differing host complement reserves or differential sampling (112).
AKI
Early identification of patients likely to develop AKI may have therapeutic implications. The development of severe kidney injury after ICU admission could be accurately identified through the measurement of admission serum creatinine and the Pediatric Sepsis Biomarker Risk Model (PERSEVERE) biomarker panel within the first 24 hours of sepsis diagnosis. This may help identify patients who may benefit from renal protective interventions (113, 114).
Patients with high serum proenkephalin (a renal biomarker) had a higher mortality rate than those without, but this mortality rate remained lower than that of those with overt AKI, suggesting that proenkephalin may help identify subclinical kidney injury (115, 116). Through secondary analysis of a randomized clinical trial comparing resuscitation strategies for patients at risk of septic shock, the product of tissue inhibitor of metalloproteinases-2 and insulin-like growth factor–binding protein 7 in urine was associated with the up-front severity of illness and was predictive of Kidney Disease Improving Global Outcomes stage 3 kidney injury, renal replacement therapy, or death within 7 days (117, 118). The genetic predisposition for developing AKI, associated with SNPs at the RF2 and TBX1 loci, was not confirmed in a large Finnish ICU population (119), tempering the enthusiasm for genome-wide association studies that do not have rigorous prior evidence for implicated pathways.
Using principles of the Stewart strong ion difference, an extracorporeal mechanism to remove blood chloride successfully decreased blood chloride, improved pH and urinary chloride and ammonium excretion, and was well tolerated in an animal model of respiratory and metabolic acidosis without shock (120). This may represent an alternative treatment for acidosis, although advantages over traditional dialysis are not clear (121).
ICU Organization
Nearly one in six ICU admissions are potentially preventable, with wide variation across U.S. states (122), suggesting that up-front investments in ambulatory care may enable the avoidance of ICU admissions and that preventable ICU admissions could serve as a metric of population health (123). Hospital strain, measured by a novel composite strain index, was strongly associated with ICU admission for patients with sepsis and/or acute respiratory failure, setting up a within-hospital variable to assess the marginal benefit of ICU admission (124). In Australia and New Zealand, ICU discharge delay for more than 6 hours occurred in 25% of patients and was associated with lower ICU readmission and hospital mortality, especially in the subgroup of patients with the highest predicted risk of mortality at ICU admission (125, 126). Identifying factors associated with ICU readmission is important because clinicians had only fair accuracy in predicting it (127). This may reflect the complexity of ICU admission and/or readmission as an organizational construct (128).
The structure and makeup of the ICU may have important implications for patient outcomes. In a before and after cohort study, the rollout of an emergency department–embedded critical care unit was not associated with improved outcomes (129), which puts into question whether the potential benefits are offset by the incurred harm of such units (130, 131). However, nurse staffing models and network relationships of nursing teams (132), as well as interactions between staff members (measured through the analysis of wearable badges) (133), seemed to play an important role in the outcomes of ICU patients. Furthermore, in rural hospitals, initiatives, attitudes, and ownership of change are key determinants for implementing evidence-based practice in the ICU, leading to better program engagement (134). In addition, methods to extend the influence of critical care physicians, including telemedicine approaches that enable physicians to participate as “copilots” guiding resuscitation during in-hospital cardiac arrest, may be beneficial on the wards of community hospitals (135).
Unfortunately, increasing demands on critical care practitioners may have consequences, as nearly 80% of division directors in pulmonary, sleep, and critical care believed that burnout was a problem in their division. Many drivers of burnout were inherent to personality characteristics of individuals or characteristics of the job, but many potentially modifiable factors, such as misalignment between the clinicians and administration, the electronic health records, and the workplace climate, were identified (136).
Disparities have a large impact on patient outcomes. Among critically ill patients, 25% of African American patients and 48% of Hispanic patients were concentrated in only 14 out of 208 U.S. hospitals. Hospital mortality and ICU and hospital stays decreased over time but remained higher in minority-serving hospitals than in non–minority-serving hospitals and did not change in the subgroup of African American patients (137, 138). Another study identified that female survivors of shock or respiratory failure had poorer physical and psychological function than male survivors and were less likely to live at home 3 months after discharge, although most of these differences attenuated after 12 months (139).
Novel therapeutics have improved outcomes for many patients with cancer, although they may have adverse events prompting ICU admission. Patients who received chimeric antigen receptor T-cell therapy for lymphoma had higher rates of ICU admission for neurotoxicity and longer hospital stays than control patients with lymphoma, although ICU lengths of stay were shorter and the 60-day mortality was lower for these patients than for control patients with lymphoma admitted to the ICU (140).
Post-ICU Trajectories and Patient/Family Satisfaction
Critical illness survivors’ priorities evolved during recovery from basic survival concerns (e.g., mobility, participating in self-care) immediately after leaving the ICU to more aspirational concerns (e.g., seeking new experiences) 2 months after discharge (141). Identifying factors associated with post-ICU morbidity has therefore been a focus. In patients older than 65 years of age with severe traumatic brain injury, age and an Injury Severity Score ≥25 were independently associated with functional outcomes, with only 6% of patients recovering functional independence by 6 months (142, 143). Furthermore, at the transition out of the hospital (144), adherence to expert recommendations in the postsepsis setting (medication optimization, screening for common impairments, monitoring for common preventable causes of deterioration, and treatment alignment with patient preferences) (145) occurred in only 11% of patients recovering from sepsis but was associated with an 88% reduced odds of death or hospital readmission (146). Although there is often a concern that opioid use in the ICU may lead to dependence after ICU discharge, persistent opioid use 1 year after hospital discharge was reported in only 2.6% of ICU survivors treated with mechanical ventilation (147, 148).
Family satisfaction is a crucial component of delivering high-quality critical care, as illustrated by the need for a family-centered discharge approach focused on the coordination of care and teaching of emergency preparedness as keys to the safe discharge of tracheostomized children or young adults (149). These themes suggest opportunities to use simulation for education on airway emergencies and to use telemedicine for minimizing the risks of travel (150). During the second and third weeks of patients’ ventilation, both surrogate decision-makers and physicians were less likely to prefer that mechanical ventilation be continued for patients with poor prognoses, but surrogates required until Week 3 of mechanical ventilation before incorporating the prognosis into their decision-making (151). Interestingly, the concepts of futile or inappropriate treatment were difficult to understand for community members, but most were able to recognize inappropriate treatment when presented with a case (152). Inadequate physician–patient communication may underpin the difficulty with the concept, together with fears that the judgment of medical futility could undermine patient/family autonomy. Nevertheless, for patients who died in the hospital, complete ICU or mixed ICU/general-floor care at the end of life was consistently associated with higher family satisfaction, highlighting the importance of ICU care at the end of life (153, 154).
Critical Care Research
Disparities are apparent in the gender breakdown of critical care authorship over the last 10 years. Only 30% of critical care publications have a female first author, with less than 20% having a female senior author, although the presence of a female senior author was associated with a higher likelihood of having a female co-author. This may be contributing to there being fewer women in leadership positions in critical care (155, 156).
Improving methods to perform critical care research was a focus of several investigations. Many collaborative critical care research networks are focused on conducting high-quality randomized controlled trials but are highly dependent on external sources of funding to support network infrastructure (157). The creation of common protocols and approaches is crucial for the success of critical care trials. Recommendations on the maximal blood volume allowable for research and on the safe lower limit of Hb at the time of blood draws for critically ill patients have been proposed (158). Trial recruitment is often challenging in the ICU (159), and a collection of “nudges” presented to surrogate decision-makers as a preconsent survey was not an effective means of increasing enrollment in a sham trial (160). This prompts a need to better understand how surrogates perceive the risk of trial participation.
Administrative data are frequently used in critical care research investigations, and analyses using such data have more accurately identified the use of mechanical ventilation, but not the use of vasoactive medications or acute renal replacement therapy, than clinical chart review (161). Integration of electronic health record data may be required to answer some questions (162).
Education in Critical Care
This past year, the greatest accomplishment in critical care education was helping nonintensivists care for patients in the ICU. Educational content was rapidly created and widely disseminated through publications as well as on social media platforms such as Twitter, which had been widely used by intensivists before the pandemic and became an even more powerful tool for education during the pandemic (163). Much of the educational content aimed at helping care for patients with COVID-19 was published in ATS Scholar, a new open-access journal disseminating medical education focused on pulmonary, critical care, and sleep medicine. A series of videos (164–167) on mechanical ventilation for those with limited or nonexistent training proved to be an invaluable resource for clinicians redeployed to work in ICUs during the pandemic. In light of severely stretched staffing, the concept of “respiratory therapy extenders” was created, and a framework for how to train medical students as such extenders was reported (168). Online resources with quick tips and algorithms for nonintensivists to reference when caring for critically patients (169, 170) as well as methods to keep intensivists up to date on the rapidly changing evidence related to the care of patients with COVID-19, such as the “Fast Literature Assessment and Review Initiative,” were shared widely (171).
Footnotes
Originally Published in Press as DOI: 10.1164/rccm.202102-0336UP on March 15, 2021
Author disclosures are available with the text of this article at www.atsjournals.org.
References
- 1.Minagawa S, Yoshida M, Araya J, Hara H, Imai H, Kuwano K. Regulated necrosis in pulmonary disease: a focus on necroptosis and ferroptosis. Am J Respir Cell Mol Biol. 2020;62:554–562. doi: 10.1165/rcmb.2019-0337TR. [DOI] [PubMed] [Google Scholar]
- 2.Mahida RY, Matsumoto S, Matthay MA. Extracellular vesicles: a new frontier for research in acute respiratory distress syndrome. Am J Respir Cell Mol Biol. 2020;63:15–24. doi: 10.1165/rcmb.2019-0447TR. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Szabo C, Martins V, Liaudet L. Poly(ADP-ribose) polymerase inhibition in acute lung injury: a reemerging concept. Am J Respir Cell Mol Biol. 2020;63:571–590. doi: 10.1165/rcmb.2020-0188TR. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Tsai M, Mallampalli RK. E-cigarette or vaping product use-associated lung injury: opportunities and challenges. Am J Respir Cell Mol Biol. 2020;62:397–398. doi: 10.1165/rcmb.2019-0422LE. [DOI] [PubMed] [Google Scholar]
- 5.Singer BD, Jain M, Budinger GRS, Wunderink RG. A call for rational intensive care in the era of COVID-19. Am J Respir Cell Mol Biol. 2020;63:132–133. doi: 10.1165/rcmb.2020-0151LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Sananez I, Raiden S, Holgado MP, Seery V, De Lillo L, Davenport C, et al. Upregulation of CD32 in T cells from infants with severe respiratory syncytial virus disease: a new costimulatory pathway? Am J Respir Cell Mol Biol. 2020;63:133–136. doi: 10.1165/rcmb.2020-0025LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Migaud M, Gandotra S, Chand HS, Gillespie MN, Thannickal VJ, Langley RJ. Metabolomics to predict antiviral drug efficacy in COVID-19. Am J Respir Cell Mol Biol. 2020;63:396–398. doi: 10.1165/rcmb.2020-0206LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Evans SE, Tseng CK, Scott BL, Höök AM, Dickey BF. Inducible epithelial resistance against coronavirus pneumonia in mice. Am J Respir Cell Mol Biol. 2020;63:540–541. doi: 10.1165/rcmb.2020-0247LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hedrick TL, Murray BP, Hagan RS, Mock JR. COVID-19: clean up on IL-6. Am J Respir Cell Mol Biol. 2020;63:541–543. doi: 10.1165/rcmb.2020-0277LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Renteria AE, Mfuna Endam L, Adam D, Filali-Mouhim A, Maniakas A, Rousseau S, et al. Azithromycin downregulates gene expression of IL-1β and pathways involving TMPRSS2 and TMPRSS11D required by SARS-CoV-2. Am J Respir Cell Mol Biol. 2020;63:707–709. doi: 10.1165/rcmb.2020-0285LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gruber AD, Osterrieder N, Bertzbach LD, Vladimirova D, Greuel S, Ihlow J, et al. Standardization of reporting criteria for lung pathology in SARS-CoV-2-infected hamsters: what matters? Am J Respir Cell Mol Biol. 2020;63:856–859. doi: 10.1165/rcmb.2020-0280LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Murphy RC, Lai Y, Barrow KA, Hamerman JA, Lacy-Hulbert A, Piliponsky AM, et al. Effects of asthma and human rhinovirus A16 on the expression of SARS-CoV-2 entry factors in human airway epithelium. Am J Respir Cell Mol Biol. 2020;63:859–863. doi: 10.1165/rcmb.2020-0394LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Sinha P, Churpek MM, Calfee CS. Machine learning classifier models can identify acute respiratory distress syndrome phenotypes using readily available clinical data. Am J Respir Crit Care Med. 2020;202:996–1004. doi: 10.1164/rccm.202002-0347OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.McNicholas B, Madden MG, Laffey JG. Machine learning classifier models: the future for acute respiratory distress syndrome phenotyping? Am J Respir Crit Care Med. 2020;202:919–920. doi: 10.1164/rccm.202006-2388ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Jones TK, Feng R, Kerchberger VE, Reilly JP, Anderson BJ, Shashaty MGS, et al. Plasma sRAGE acts as a genetically regulated causal intermediate in sepsis-associated acute respiratory distress syndrome. Am J Respir Crit Care Med. 2020;201:47–56. doi: 10.1164/rccm.201810-2033OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Walley KR. Discovering causal mechanistic pathways in sepsis-associated acute respiratory distress syndrome. Am J Respir Crit Care Med. 2020;201:2–4. doi: 10.1164/rccm.201909-1772ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Krenn K, Höbart P, Poglitsch M, Croizé A, Ullrich R. Equilibrium angiotensin metabolite profiling in patients with acute respiratory distress syndrome indicates angiotensin-converting enzyme inhibition. Am J Respir Crit Care Med. 2020;202:1468–1471. doi: 10.1164/rccm.201912-2504LE. [DOI] [PubMed] [Google Scholar]
- 18.Mock JR, Dial CF, Tune MK, Gilmore RC, O’Neal WK, Dang H, et al. Impact of regulatory T cells on type 2 alveolar epithelial cell transcriptomes during resolution of acute lung injury and contributions of IFN-γ. Am J Respir Cell Mol Biol. 2020;63:464–477. doi: 10.1165/rcmb.2019-0399OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Cipolla EM, Alcorn JF. Repair of the lung by regulatory T cells. Am J Respir Cell Mol Biol. 2020;63:405–407. doi: 10.1165/rcmb.2020-0260ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Bharat A, Angulo M, Sun H, Akbarpour M, Alberro A, Cheng Y, et al. High CO2 levels impair lung wound healing. Am J Respir Cell Mol Biol. 2020;63:244–254. doi: 10.1165/rcmb.2019-0354OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.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]
- 22.Gattinoni L, Marini JJ, Quintel M. Recruiting the acutely injured lung: how and why? Am J Respir Crit Care Med. 2020;201:130–132. doi: 10.1164/rccm.201910-2005ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Pellegrini M, Gudmundsson M, Bencze R, Segelsjö M, Freden F, Rylander C, et al. Expiratory resistances prevent expiratory diaphragm contraction, flow limitation, and lung collapse. Am J Respir Crit Care Med. 2020;201:1218–1229. doi: 10.1164/rccm.201909-1690OC. [DOI] [PubMed] [Google Scholar]
- 24.Spadaro S, Volta CA. A physiological point of view on expiratory (Re)action during mechanical ventilation. Am J Respir Crit Care Med. 2020;201:1170–1172. doi: 10.1164/rccm.202003-0645ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Pereira Romano ML, Maia IS, Laranjeira LN, Damiani LP, Paisani DM, Borges MC, et al. Driving pressure-limited strategy for patients with acute respiratory distress syndrome: a pilot randomized clinical trial. Ann Am Thorac Soc. 2020;17:596–604. doi: 10.1513/AnnalsATS.201907-506OC. [DOI] [PubMed] [Google Scholar]
- 26.Hirshberg EL, Majercik S. Targeting driving pressure for the management of ARDS . . . isn’t it just very low tidal volume ventilation? Ann Am Thorac Soc. 2020;17:557–558. doi: 10.1513/AnnalsATS.202002-108ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhong M, Wu W, Kang H, Hong Z, Xiong S, Gao X, et al. Alveolar stretch activation of endothelial Piezo1 protects adherens junctions and lung vascular barrier. Am J Respir Cell Mol Biol. 2020;62:168–177. doi: 10.1165/rcmb.2019-0024OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Heise RL, Farkas L. A cation channel protects the stretched lung. Am J Respir Cell Mol Biol. 2020;62:128–129. doi: 10.1165/rcmb.2019-0292ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Dianti J, Angriman F, Ferreyro BL, Sklar MC, Brochard L, Ferguson ND, et al. Association of mortality with neuromuscular blockade differs according to baseline diaphragm thickness. Am J Respir Crit Care Med. 2020;202:1717–1720. doi: 10.1164/rccm.202004-1157LE. [DOI] [PubMed] [Google Scholar]
- 30.Telias I, Junhasavasdikul D, Rittayamai N, Piquilloud L, Chen L, Ferguson ND, et al. Airway occlusion pressure as an estimate of respiratory drive and inspiratory effort during assisted ventilation. Am J Respir Crit Care Med. 2020;201:1086–1098. doi: 10.1164/rccm.201907-1425OC. [DOI] [PubMed] [Google Scholar]
- 31.Sassoon CS, Younes M. Airway occlusion pressure revisited. Am J Respir Crit Care Med. 2020;201:1027–1028. doi: 10.1164/rccm.202003-0585ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Teggia-Droghi M, Grassi A, Rezoagli E, Pozzi M, Foti G, Patroniti N, et al. Comparison of two approaches to estimate driving pressure during assisted ventilation. Am J Respir Crit Care Med. 2020;202:1595–1598. doi: 10.1164/rccm.202004-1281LE. [DOI] [PubMed] [Google Scholar]
- 33.Soilemezi E, Savvidou S, Sotiriou P, Smyrniotis D, Tsagourias M, Matamis D. Tissue Doppler imaging of the diaphragm in healthy subjects and critically ill patients. Am J Respir Crit Care Med. 2020;202:1005–1012. doi: 10.1164/rccm.201912-2341OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.McCool FD, Tzelepis GE. Tissue Doppler imaging of the diaphragm: a new kid on the block? Am J Respir Crit Care Med. 2020;202:921–922. doi: 10.1164/rccm.202007-2771ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jonkman AH, Roesthuis LH, de Boer EC, de Vries HJ, Girbes ARJ, van der Hoeven JG, et al. Inadequate assessment of patient-ventilator interaction due to suboptimal diaphragm electrical activity signal filtering. Am J Respir Crit Care Med. 2020;202:141–144. doi: 10.1164/rccm.201912-2306LE. [DOI] [PubMed] [Google Scholar]
- 36.Tonelli R, Fantini R, Tabbì L, Castaniere I, Pisani L, Pellegrino MR, et al. Early inspiratory effort assessment by esophageal manometry predicts noninvasive ventilation outcome in de novo respiratory failure: a pilot study. Am J Respir Crit Care Med. 2020;202:558–567. doi: 10.1164/rccm.201912-2512OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Yoshida T. The dark side of spontaneous breathing during non-invasive ventilation: from hypothesis to theory. Am J Respir Crit Care Med. 2020;202:482–484. doi: 10.1164/rccm.202005-1612ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Tuffet S, Mekontso Dessap A, Carteaux G. Noninvasive ventilation for de novo respiratory failure: impact of ventilator setting adjustments. Am J Respir Crit Care Med. 2020;202:769–770. doi: 10.1164/rccm.202005-1565LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Grieco DL, Menga LS, Raggi V, Bongiovanni F, Anzellotti GM, Tanzarella ES, et al. Physiological comparison of high-flow nasal cannula and helmet noninvasive ventilation in acute hypoxemic respiratory failure. Am J Respir Crit Care Med. 2020;201:303–312. doi: 10.1164/rccm.201904-0841OC. [DOI] [PubMed] [Google Scholar]
- 40.Thille AW, Yoshida T. High pressure versus high flow: what should we target in acute respiratory failure? Am J Respir Crit Care Med. 2020;201:265–266. doi: 10.1164/rccm.201911-2196ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Cortegiani A, Accurso G, Ball L, Ippolito M, Ingoglia G, Vitale F, et al. A new configuration for helmet continuous positive airway pressure allowing tidal volume monitoring. Am J Respir Crit Care Med. 2020;202:612–614. doi: 10.1164/rccm.202003-0550LE. [DOI] [PubMed] [Google Scholar]
- 42.Simonson OE, Ståhle E, Hansen T, Wedin JO, Larsson A, Mattsson M, et al. Five-year follow-up after mesenchymal stromal cell-based treatment of severe acute respiratory distress syndrome. Am J Respir Crit Care Med. 2020;202:1051–1055. doi: 10.1164/rccm.202003-0544LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Del Sorbo L, Fan E. Mesenchymal stem cells in acute respiratory distress syndrome supported with extracorporeal membrane oxygenation: lost in translational research? Am J Respir Crit Care Med. 2020;202:314–315. doi: 10.1164/rccm.202004-1139ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Millar JE, Bartnikowski N, Passmore MR, Obonyo NG, Malfertheiner MV, von Bahr V, et al. Combined mesenchymal stromal cell therapy and extracorporeal membrane oxygenation in acute respiratory distress syndrome: a randomized controlled trial in sheep. Am J Respir Crit Care Med. 2020;202:383–392. doi: 10.1164/rccm.201911-2143OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Rowan CM, Klein MJ, Hsing DD, Dahmer MK, Spinella PC, Emeriaud G, et al. Early use of adjunctive therapies for pediatric acute respiratory distress syndrome: a PARDIE study. Am J Respir Crit Care Med. 2020;201:1389–1397. doi: 10.1164/rccm.201909-1807OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Oishi P, Fineman JR. Equipoise and acumen in pediatric acute respiratory distress syndrome research. Am J Respir Crit Care Med. 2020;201:1326–1327. doi: 10.1164/rccm.202003-0746ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Law AC, Forbath N, O’Donoghue S, Stevens JP, Walkey AJ. Hospital-level availability of prone positioning in Massachusetts ICUs. Am J Respir Crit Care Med. 2020;201:1006–1008. doi: 10.1164/rccm.201910-2097LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Cavayas YA, Munshi L, Del Sorbo L, Fan E. The early change in PaCO2 after extracorporeal membrane oxygenation initiation is associated with neurological complications. Am J Respir Crit Care Med. 2020;201:1525–1535. doi: 10.1164/rccm.202001-0023OC. [DOI] [PubMed] [Google Scholar]
- 49.Karagiannidis C, Windisch W, Bein T. Rapid changes in arterial carbon dioxide levels caused by extracorporeal membrane oxygenation: the temptation of a fascinating technology. Am J Respir Crit Care Med. 2020;201:1466–1468. doi: 10.1164/rccm.202004-1060ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Bourcier S, Villie P, Nguyen S, Hékimian G, Demondion P, Bréchot N, et al. Venoarterial extracorporeal membrane oxygenation support rescue of obstructive shock caused by bulky compressive mediastinal cancer. Am J Respir Crit Care Med. 2020;202:1181–1184. doi: 10.1164/rccm.202001-0193LE. [DOI] [PubMed] [Google Scholar]
- 51.Matsumoto S, Fang X, Traber MG, Jones KD, Langelier C, Hayakawa Serpa P, et al. Dose-dependent pulmonary toxicity of aerosolized Vitamin E acetate. Am J Respir Cell Mol Biol. 2020;63:748–757. doi: 10.1165/rcmb.2020-0209OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Wagener BM, Anjum N, Evans C, Brandon A, Honavar J, Creighton J, et al. α-Tocopherol attenuates the severity of Pseudomonas aeruginosa–induced pneumonia. Am J Respir Cell Mol Biol. 2020;63:234–243. doi: 10.1165/rcmb.2019-0185OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Autilio C, Echaide M, Shankar-Aguilera S, Bragado R, Amidani D, Salomone F, et al. Surfactant injury in the early phase of severe meconium aspiration syndrome. Am J Respir Cell Mol Biol. 2020;63:327–337. doi: 10.1165/rcmb.2019-0413OC. [DOI] [PubMed] [Google Scholar]
- 54.Evankovich J, Lear T, Baldwin C, Chen Y, White V, Villandre J, et al. Toll-like receptor 8 stability is regulated by ring finger 216 in response to circulating microRNAs. Am J Respir Cell Mol Biol. 2020;62:157–167. doi: 10.1165/rcmb.2018-0373OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ghimire L, Jeyaseelan S. Kiss of death: ring finger 216 regulates Toll-like receptor 8 stability through ubiquitination. Am J Respir Cell Mol Biol. 2020;62:125–127. doi: 10.1165/rcmb.2019-0272ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wali S, Flores JR, Jaramillo AM, Goldblatt DL, Pantaleón García J, Tuvim MJ, et al. Immune modulation to improve survival of viral pneumonia in mice. Am J Respir Cell Mol Biol. 2020;63:758–766. doi: 10.1165/rcmb.2020-0241OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ballinger MN, Davis IC. CD8+ T cells: exacting a Toll in viral pneumonia. Am J Respir Cell Mol Biol. 2020;63:717–718. doi: 10.1165/rcmb.2020-0378ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Siefker DT, Vu L, You D, McBride A, Taylor R, Jones TL, et al. Respiratory syncytial virus disease severity is associated with distinct CD8+ T-cell profiles. Am J Respir Crit Care Med. 2020;201:325–334. doi: 10.1164/rccm.201903-0588OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Flerlage T, Souquette A, Allen EK, Brahm T, Crawford JC, Tang L, et al. Nasal wash cytokines during respiratory viral infection in pediatric allogeneic hematopoietic cell-transplant recipients. Am J Respir Cell Mol Biol. 2020;63:349–361. doi: 10.1165/rcmb.2020-0014OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Dickson RP, Schultz MJ, van der Poll T, Schouten LR, Falkowski NR, Luth JE, et al. Consortium BAiSIPB: lung microbiota predict clinical outcomes in critically ill patients. Am J Respir Crit Care Med. 2020;201:555–563. doi: 10.1164/rccm.201907-1487OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Cookson WOCM, Moffatt MF. In the wrong place at the wrong time: microbial misplacement and acute respiratory distress syndrome. Am J Respir Crit Care Med. 2020;201:506–507. doi: 10.1164/rccm.202001-0004ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kitsios GD, Yang H, Yang L, Qin S, Fitch A, Wang XH, et al. Respiratory tract dysbiosis is associated with worse outcomes in mechanically ventilated patients. Am J Respir Crit Care Med. 2020;202:1666–1677. doi: 10.1164/rccm.201912-2441OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Ali F, Sweeney DA. In pursuit of microbiome-based therapies for acute respiratory failure. Am J Respir Crit Care Med. 2020;202:1616–1618. doi: 10.1164/rccm.202008-3146ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mandell EW, Ryan S, Seedorf GJ, Gonzalez T, Smith BJ, Fleet JC, et al. Maternal vitamin D deficiency causes sustained impairment of lung structure and function and increases susceptibility to hyperoxia-induced lung injury in infant rats. Am J Respir Cell Mol Biol. 2020;63:79–91. doi: 10.1165/rcmb.2019-0295OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Cornfield DN. Vitamin D: feel it in more than just your bones! Am J Respir Cell Mol Biol. 2020;63:11–12. doi: 10.1165/rcmb.2020-0072ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zhang L, Soni S, Hekimoglu E, Berkelhamer S, Çataltepe S. Impaired autophagic activity contributes to the pathogenesis of bronchopulmonary dysplasia: evidence from murine and baboon models. Am J Respir Cell Mol Biol. 2020;63:338–348. doi: 10.1165/rcmb.2019-0445OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Rosman J, Contou D, Tran Van Nhieu J, Renaud M, Bottier M, Maitre B, et al. Severe ciliary dyskinesia in ventilated patients: a pilot study. Am J Respir Crit Care Med. 2020;201:867–869. doi: 10.1164/rccm.201912-2440LE. [DOI] [PubMed] [Google Scholar]
- 68.Schauwvlieghe AFAD, Rijnders BJA, Philips N, Verwijs R, Vanderbeke L, Van Tienen C, et al. Dutch-Belgian Mycosis Study Group. Invasive aspergillosis in patients admitted to the intensive care unit with severe influenza: a retrospective cohort study. Lancet Respir Med. 2018;6:782–792. doi: 10.1016/S2213-2600(18)30274-1. [DOI] [PubMed] [Google Scholar]
- 69.Loughlin L, Hellyer TP, White PL, McAuley DF, Conway Morris A, Posso RB, et al. Pulmonary aspergillosis in patients with suspected ventilator-associated pneumonia in UK ICUs. Am J Respir Crit Care Med. 2020;202:1125–1132. doi: 10.1164/rccm.202002-0355OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Torres A, Martin-Loeches I. Invasive pulmonary aspergillosis in ventilator-associated pneumonia: the hidden enemy? Am J Respir Crit Care Med. 2020;202:1071–1073. doi: 10.1164/rccm.202006-2605ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Nyga R, Maizel J, Nseir S, Chouaki T, Milic I, Roger PA, et al. Invasive tracheobronchial aspergillosis in critically ill patients with severe influenza: a clinical trial. Am J Respir Crit Care Med. 2020;202:708–716. doi: 10.1164/rccm.201910-1931OC. [DOI] [PubMed] [Google Scholar]
- 72.van de Veerdonk FL, Wauters J, Verweij PE. Invasive Aspergillus tracheobronchitis emerging as a highly lethal complication of severe influenza. Am J Respir Crit Care Med. 2020;202:646–648. doi: 10.1164/rccm.202005-1883ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Althoff MD, Holguin F, Yang F, Grunwald GK, Moss M, Vandivier RW, et al. Noninvasive ventilation use in critically ill patients with acute asthma exacerbations. Am J Respir Crit Care Med. 2020;202:1520–1530. doi: 10.1164/rccm.201910-2021OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Pearson SD, Patel BK. Noninvasive ventilation for acute asthma: the neglected sibling. Am J Respir Crit Care Med. 2020;202:1491–1493. doi: 10.1164/rccm.202007-2876ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Berenyi F, Steinfort DP, Abdelhamid YA, Bailey MJ, Pilcher DV, Bellomo R, et al. Characteristics and outcomes of critically ill patients with acute exacerbation of chronic obstructive pulmonary disease in Australia and New Zealand. Ann Am Thorac Soc. 2020;17:736–745. doi: 10.1513/AnnalsATS.201911-821OC. [DOI] [PubMed] [Google Scholar]
- 76.Blagev DP. Better with time: reductions in mortality in patients with acute exacerbation of chronic obstructive pulmonary disease. Ann Am Thorac Soc. 2020;17:684–685. doi: 10.1513/AnnalsATS.202002-178ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ruan S-Y, Teng N-C, Huang C-T, Tsai S-L, Wang C-Y, Wu C-P, et al. Dynamic changes in prognosis with elapsed time on ventilators among mechanically ventilated patients. Ann Am Thorac Soc. 2020;17:729–735. doi: 10.1513/AnnalsATS.201908-646OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Zaeh SE, Mehta AB. Prognosis for mechanically ventilated patients: a moving target. Ann Am Thorac Soc. 2020;17:682–683. doi: 10.1513/AnnalsATS.202003-242ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Rak KJ, Ashcraft LE, Kuza CC, Fleck JC, DePaoli LC, Angus DC, et al. Effective care practices in patients receiving prolonged mechanical ventilation: an ethnographic study. Am J Respir Crit Care Med. 2020;201:823–831. doi: 10.1164/rccm.201910-2006OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Rose L. Rethinking delivery of care for patients requiring prolonged mechanical ventilation. Am J Respir Crit Care Med. 2020;201:760–761. doi: 10.1164/rccm.202002-0292ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lindroth H, Khan BA, Carpenter JS, Gao S, Perkins AJ, Khan SH, et al. Delirium severity trajectories and outcomes in ICU patients: defining a dynamic symptom phenotype. Ann Am Thorac Soc. 2020;17:1094–1103. doi: 10.1513/AnnalsATS.201910-764OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Deane AM, Little L, Bellomo R, Chapman MJ, Davies AR, Ferrie S, et al. Outcomes six months after delivering 100% or 70% of enteral calorie requirements during critical illness (target): a randomized controlled trial. Am J Respir Crit Care Med. 2020;201:814–822. doi: 10.1164/rccm.201909-1810OC. [DOI] [PubMed] [Google Scholar]
- 83.Morrissette KM, Stapleton RD. Mounting clarity on enteral feeding in critically ill patients. Am J Respir Crit Care Med. 2020;201:758–760. doi: 10.1164/rccm.202001-0126ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Marik PE, Khangoora V, Rivera R, Hooper MH, Catravas J. Hydrocortisone, vitamin C, and thiamine for the treatment of severe sepsis and septic shock: a retrospective before-after study. Chest. 2017;151:1229–1238. doi: 10.1016/j.chest.2016.11.036. [DOI] [PubMed] [Google Scholar]
- 85.Vail EA, Wunsch H, Pinto R, Bosch NA, Walkey AJ, Lindenauer PK, et al. Use of hydrocortisone, ascorbic acid, and thiamine in adults with septic shock. Am J Respir Crit Care Med. 2020;202:1531–1539. doi: 10.1164/rccm.202005-1829OC. [DOI] [PubMed] [Google Scholar]
- 86.Wald EL, Sanchez-Pinto LN, Smith CM, Moran T, Badke CM, Barhight MF, et al. Hydrocortisone-ascorbic acid-thiamine use associated with lower mortality in pediatric septic shock. Am J Respir Crit Care Med. 2020;201:863–867. doi: 10.1164/rccm.201908-1543LE. [DOI] [PubMed] [Google Scholar]
- 87.Cohen J, Bellomo R, Billot L, Burrell LM, Evans DM, Finfer S, et al. Plasma cortisol, aldosterone, and ascorbic acid concentrations in patients with septic shock do not predict treatment effect of hydrocortisone on mortality: a nested cohort study. Am J Respir Crit Care Med. 2020;202:700–707. doi: 10.1164/rccm.202002-0281OC. [DOI] [PubMed] [Google Scholar]
- 88.Møller MH, Shankar-Hari M, Alhazzani W. Use of hydrocortisone based on plasma biomarkers in patients with septic shock: another one bites the dust? Am J Respir Crit Care Med. 2020;202:644–646. doi: 10.1164/rccm.202005-1984ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Khanna A, English SW, Wang XS, Ham K, Tumlin J, Szerlip H, et al. ATHOS-3 Investigators. Angiotensin II for the treatment of vasodilatory shock. N Engl J Med. 2017;377:419–430. doi: 10.1056/NEJMoa1704154. [DOI] [PubMed] [Google Scholar]
- 90.Bellomo R, Forni LG, Busse LW, McCurdy MT, Ham KR, Boldt DW, et al. Renin and survival in patients given angiotensin II for catecholamine-resistant vasodilatory shock: a clinical trial. Am J Respir Crit Care Med. 2020;202:1253–1261. doi: 10.1164/rccm.201911-2172OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Casey JD, Semler MW. Renin, angiotensin II, and the journey to evidence-based individual treatment effects. Am J Respir Crit Care Med. 2020;202:1209–1211. doi: 10.1164/rccm.202007-2731ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Zampieri FG, Damiani LP, Bakker J, Ospina-Tascón GA, Castro R, Cavalcanti AB, et al. Effects of a resuscitation strategy targeting peripheral perfusion status versus serum lactate levels among patients with septic shock: a Bayesian reanalysis of the ANDROMEDA-SHOCK trial. Am J Respir Crit Care Med. 2020;201:423–429. doi: 10.1164/rccm.201905-0968OC. [DOI] [PubMed] [Google Scholar]
- 93.Yarnell CJ, Granton JT, Tomlinson G. Bayesian analysis in critical care medicine. Am J Respir Crit Care Med. 2020;201:396–398. doi: 10.1164/rccm.201910-2019ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Gershengorn HB, Stelfox HT, Niven DJ, Wunsch H. Association of premorbid blood pressure with vasopressor infusion duration in patients with shock. Am J Respir Crit Care Med. 2020;202:91–99. doi: 10.1164/rccm.201908-1681OC. [DOI] [PubMed] [Google Scholar]
- 95.Russell JA. Personalized blood pressure targets in shock: what if your normal blood pressure is “low”? Am J Respir Crit Care Med. 2020;202:10–12. doi: 10.1164/rccm.202004-1124ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Gershengorn HB. Sometimes it is okay to get personal: individualizing targets in critical care. Am J Respir Crit Care Med. 2020;202:1334–1336. doi: 10.1164/rccm.202007-2728ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Panwar R, Tarvade S, Lanyon N, Saxena M, Bush D, Hardie M, et al. REACT Shock Study Investigators and Research Coordinators. Relative hypotension and adverse kidney-related outcomes among critically ill patients with shock: a multicenter, prospective cohort study. Am J Respir Crit Care Med. 2020;202:1407–1418. doi: 10.1164/rccm.201912-2316OC. [DOI] [PubMed] [Google Scholar]
- 98.Ranzani OT, Monteiro MB, Besen BAMP, Azevedo LCP. Association of sepsis diagnosis at daytime and on weekdays with compliance with the 3-hour sepsis treatment bundles: a multicenter cohort study. Ann Am Thorac Soc. 2020;17:980–987. doi: 10.1513/AnnalsATS.201910-781OC. [DOI] [PubMed] [Google Scholar]
- 99.Zampieri FG, de Oliveira NE, Nassar AP, Jr, de Oliveira Manoel AL, Grion C, Lacerda FH, et al. BRICNet. Bundle of coated devices to reduce nosocomial infections in the intensive care unit: CRITIC pilot randomized controlled trial. Ann Am Thorac Soc. 2020;17:1257–1263. doi: 10.1513/AnnalsATS.202003-206OC. [DOI] [PubMed] [Google Scholar]
- 100.Maley JH, Worsham CM, Landon BE, Stevens JP. Association between palliative care and end-of-life resource use for older adults hospitalized with septic shock. Ann Am Thorac Soc. 2020;17:974–979. doi: 10.1513/AnnalsATS.202001-038OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Cooper AJ, Keller SP, Chan C, Glotzbecker BE, Klompas M, Baron RM, et al. Improvements in sepsis-associated mortality in hospitalized patients with cancer versus those without cancer: a 12-year analysis using clinical data. Ann Am Thorac Soc. 2020;17:466–473. doi: 10.1513/AnnalsATS.201909-655OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Greenberg JA. Improvements in sepsis-associated mortality among patients with cancer: an encouraging finding with uncertain implications. Ann Am Thorac Soc. 2020;17:425–426. doi: 10.1513/AnnalsATS.201912-889ED. [DOI] [PubMed] [Google Scholar]
- 103.Stone CA, Jr, Stollings JL, Lindsell CJ, Dear ML, Buie RB, Rice TW, et al. Risk-stratified management to remove low-risk penicillin allergy labels in the ICU. Am J Respir Crit Care Med. 2020;201:1572–1575. doi: 10.1164/rccm.202001-0089LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Machado FR, Cavalcanti AB, Monteiro MB, Sousa JL, Bossa A, Bafi AT, et al. Instituto Latino-Americano de Sepsis network investigators. Predictive accuracy of the quick sepsis-related organ failure assessment score in Brazil: a prospective multicenter study. Am J Respir Crit Care Med. 2020;201:789–798. doi: 10.1164/rccm.201905-0917OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.DeMerle KM, Rudd KE. The quick sepsis-related organ failure assessment score in Brazil: insights for future use in diverse settings. Am J Respir Crit Care Med. 2020;201:755–756. doi: 10.1164/rccm.202001-0058ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Wang RJ, Katha G, Phiri M, Delbridge P, Gordon SB, Calfee CS, et al. Sonographic B-lines, fluid resuscitation, and hypoxemia in Malawian patients with suspected sepsis. Am J Respir Crit Care Med. 2020;202:463–466. doi: 10.1164/rccm.202003-0640LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Stolk RF, van der Pasch E, Naumann F, Schouwstra J, Bressers S, van Herwaarden AE, et al. Norepinephrine dysregulates the immune response and compromises host defense during sepsis. Am J Respir Crit Care Med. 2020;202:830–842. doi: 10.1164/rccm.202002-0339OC. [DOI] [PubMed] [Google Scholar]
- 108.Uhel F, van der Poll T. Norepinephrine in septic shock: a mixed blessing. Am J Respir Crit Care Med. 2020;202:788–789. doi: 10.1164/rccm.202006-2301ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Carlton EF, McHugh WM, McDonough K, Sturza J, Desch K, Cornell TT. Markers of endothelial dysfunction and cytokines in high-risk pediatric patients with severe sepsis. Am J Respir Crit Care Med. 2020;201:380–384. doi: 10.1164/rccm.201904-0910LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Bustamante AC, Opron K, Ehlenbach WJ, Larson EB, Crane PK, Keene CD, et al. Transcriptomic profiles of sepsis in the human brain. Am J Respir Crit Care Med. 2020;201:861–863. doi: 10.1164/rccm.201909-1713LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Bain W, Li H, van der Geest R, Moore SR, Olonisakin TF, Ahn B, et al. Increased alternative complement pathway function and improved survival during critical illness. Am J Respir Crit Care Med. 2020;202:230–240. doi: 10.1164/rccm.201910-2083OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Bosmann M. Complement activation during critical illness: current findings and an outlook in the era of COVID-19. Am J Respir Crit Care Med. 2020;202:163–165. doi: 10.1164/rccm.202005-1926ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Stanski NL, Stenson EK, Cvijanovich NZ, Weiss SL, Fitzgerald JC, Bigham MT, et al. PERSEVERE biomarkers predict severe acute kidney injury and renal recovery in pediatric septic shock. Am J Respir Crit Care Med. 2020;201:848–855. doi: 10.1164/rccm.201911-2187OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Gien J, Soranno D. Identifying the patient at risk for acute kidney injury: pediatric sepsis biomarker risk model study. Am J Respir Crit Care Med. 2020;201:764–766. doi: 10.1164/rccm.202001-0216ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Dépret F, Hollinger A, Cariou A, Deye N, Vieillard-Baron A, Fournier MC, et al. Incidence and outcome of subclinical acute kidney injury using penKid in critically ill patients. Am J Respir Crit Care Med. 2020;202:822–829. doi: 10.1164/rccm.201910-1950OC. [DOI] [PubMed] [Google Scholar]
- 116.Koyner JL. Subclinical acute kidney injury is acute kidney injury and should not be ignored. Am J Respir Crit Care Med. 2020;202:786–787. doi: 10.1164/rccm.202006-2239ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Bhatraju PK, Wurfel MM, Himmelfarb J. Trajectory of kidney function: the canary in sepsis. Am J Respir Crit Care Med. 2020;202:1211–1212. doi: 10.1164/rccm.202007-2627ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Fiorentino M, Xu Z, Smith A, Singbartl K, Palevsky PM, Chawla LS, et al. ProCESS and ProGReSS-AKI Investigators. Serial measurement of cell-cycle arrest biomarkers [TIMP-2]•[IGFBP7] and risk for progression to death, dialysis or severe acute kidney injury in patients with septic shock. Am J Respir Crit Care Med. 2020;202:1262–1270. doi: 10.1164/rccm.201906-1197OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Renken IJE, Vilander LM, Kaunisto MA, Vaara ST, Snieder H, Keus F, et al. No association between genetic loci near IRF2 and TBX1 and acute kidney injury in the critically ill. Am J Respir Crit Care Med. 2020;201:109–111. doi: 10.1164/rccm.201903-0633LE. [DOI] [PubMed] [Google Scholar]
- 120.Zanella A, Caironi P, Castagna L, Rezoagli E, Salerno D, Scotti E, et al. Extracorporeal chloride removal by electrodialysis: a novel approach to correct acidemia. Am J Respir Crit Care Med. 2020;201:799–813. doi: 10.1164/rccm.201903-0538OC. [DOI] [PubMed] [Google Scholar]
- 121.Cove M, Kellum JA. The end of the bicarbonate era? A therapeutic application of the Stewart approach. Am J Respir Crit Care Med. 2020;201:757–758. doi: 10.1164/rccm.201910-2003ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Weissman GE, Kerlin MP, Yuan Y, Kohn R, Anesi GL, Groeneveld PW, et al. Potentially preventable intensive care unit admissions in the United States, 2006-2015. Ann Am Thorac Soc. 2020;17:81–88. doi: 10.1513/AnnalsATS.201905-366OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Admon AJ. Reducing the effect of critical illness by continuing to think beyond the intensive care unit. Ann Am Thorac Soc. 2020;17:33–35. doi: 10.1513/AnnalsATS.201910-753ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Anesi GL, Chowdhury M, Small DS, Delgado MK, Kohn R, Bayes B, et al. Association of a novel index of hospital capacity strain with admission to intensive care units. Ann Am Thorac Soc. 2020;17:1440–1447. doi: 10.1513/AnnalsATS.202003-228OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Forster GM, Bihari S, Tiruvoipati R, Bailey M, Pilcher D. The association between discharge delay from intensive care and patient outcomes. Am J Respir Crit Care Med. 2020;202:1399–1406. doi: 10.1164/rccm.201912-2418OC. [DOI] [PubMed] [Google Scholar]
- 126.Maley JH, Anesi GL. Watchful waiting in the ICU? Considerations for the allocation of ICU resources. Am J Respir Crit Care Med. 2020;202:1332–1333. doi: 10.1164/rccm.202007-2873ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Rojas JC, Lyons PG, Jiang T, Kilaru M, McCauley L, Picart J, et al. Accuracy of clinicians’ ability to predict the need for intensive care unit readmission. Ann Am Thorac Soc. 2020;17:847–853. doi: 10.1513/AnnalsATS.201911-828OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Harlan EA, Valley TS. Navigating discharges from intensive care unit to ward. Ann Am Thorac Soc. 2020;17:807–808. doi: 10.1513/AnnalsATS.202003-227ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Anesi GL, Chelluri J, Qasim ZA, Chowdhury M, Kohn R, Weissman GE, et al. Association of an emergency department-embedded critical care unit with hospital outcomes and intensive care unit use. Ann Am Thorac Soc. 2020;17:1599–1609. doi: 10.1513/AnnalsATS.201912-912OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Lyons PG, Fernando SM. On baseball, counterfactuals, and measuring care delivery performance at the emergency department-intensive care unit interface. Ann Am Thorac Soc. 2020;17:1532–1534. doi: 10.1513/AnnalsATS.202008-951ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Barbash IJ. Connecting the docs: telemedicine support during in-hospital cardiac arrest resuscitation. Ann Am Thorac Soc. 2020;17:278–279. doi: 10.1513/AnnalsATS.201912-884ED. [DOI] [PubMed] [Google Scholar]
- 132.Kelly Costa D, Liu H, Boltey EM, Yakusheva O. The structure of critical care nursing teams and patient outcomes: a network analysis. Am J Respir Crit Care Med. 2020;201:483–485. doi: 10.1164/rccm.201903-0543LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Ito-Masui A, Kawamoto E, Nagai Y, Takagi Y, Ito M, Mizutani N, et al. Feasibility of measuring face-to-face interactions among ICU healthcare professionals using wearable sociometric badges. Am J Respir Crit Care Med. 2020;201:245–247. doi: 10.1164/rccm.201904-0779LE. [DOI] [PubMed] [Google Scholar]
- 134.Sterba KR, Johnson EE, Nadig N, Simpson AN, Simpson KN, Goodwin AJ, et al. Determinants of evidence-based practice uptake in rural intensive care units: a mixed methods study. Ann Am Thorac Soc. 2020;17:1104–1116. doi: 10.1513/AnnalsATS.202002-170OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Peltan ID, Poll JB, Guidry D, Brown SM, Beninati W. Acceptability and perceived utility of telemedical consultation during cardiac arrest resuscitation: a multicenter survey. Ann Am Thorac Soc. 2020;17:321–328. doi: 10.1513/AnnalsATS.201906-485OC. [DOI] [PubMed] [Google Scholar]
- 136.Rinne ST, Swamy L, Anderson E, Shah T, Dixon AE, Kaminski N, et al. Perspectives on burnout from pulmonary, critical care, and sleep medicine division directors. Am J Respir Crit Care Med. 2020;201:111–114. doi: 10.1164/rccm.201906-1262LE. [DOI] [PubMed] [Google Scholar]
- 137.Danziger J, Ángel Armengol de la Hoz M, Li W, Komorowski M, Deliberato RO, Rush BNM, et al. Temporal trends in critical care outcomes in U.S. minority-serving hospitals. Am J Respir Crit Care Med. 2020;201:681–687. doi: 10.1164/rccm.201903-0623OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Nayfeh A, Fowler RA. Understanding patient- and hospital-level factors leading to differences, and disparities, in critical care. Am J Respir Crit Care Med. 2020;201:642–644. doi: 10.1164/rccm.202001-0116ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Scheunemann LP, Leland NE, Perera S, Skidmore ER, Reynolds CF, Pandharipande PP, et al. Sex disparities and functional outcomes after a critical illness. Am J Respir Crit Care Med. 2020;201:869–872. doi: 10.1164/rccm.201902-0328LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Brown ART, Jindani I, Melancon J, Erfe R, Westin J, Feng L, et al. ICU resource use in critically III patients following chimeric antigen receptor T-cell therapy. Am J Respir Crit Care Med. 2020;202:1184–1187. doi: 10.1164/rccm.202002-0286LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Scheunemann LP, White JS, Prinjha S, Hamm ME, Girard TD, Skidmore ER, et al. Post-intensive care unit care: a qualitative analysis of patient priorities and implications for redesign. Ann Am Thorac Soc. 2020;17:221–228. doi: 10.1513/AnnalsATS.201904-332OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Maiden MJ, Cameron PA, Rosenfeld JV, Cooper DJ, McLellan S, Gabbe BJ. Long-term outcomes after severe traumatic brain injury in older adults: a registry-based cohort study. Am J Respir Crit Care Med. 2020;201:167–177. doi: 10.1164/rccm.201903-0673OC. [DOI] [PubMed] [Google Scholar]
- 143.McCredie VA, Turgeon AF. Shades of gray matter in severe traumatic brain injury. Am J Respir Crit Care Med. 2020;201:128–129. doi: 10.1164/rccm.201911-2223ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Lee JT, Brummel NE. Passing the baton: improving postsepsis care through better transitions. Ann Am Thorac Soc. 2020;17:35–37. doi: 10.1513/AnnalsATS.201910-779ED. [DOI] [PubMed] [Google Scholar]
- 145.Prescott HC, Angus DC. Enhancing recovery from sepsis: a review. JAMA. 2018;319:62–75. doi: 10.1001/jama.2017.17687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Taylor SP, Chou S-H, Sierra MF, Shuman TP, McWilliams AD, Taylor BT, et al. Association between adherence to recommended care and outcomes for adult survivors of sepsis. Ann Am Thorac Soc. 2020;17:89–97. doi: 10.1513/AnnalsATS.201907-514OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Skrobik Y. Insights into critical care and post-ICU opiate administration. Am J Respir Crit Care Med. 2020;202:484–486. doi: 10.1164/rccm.202005-1815ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Wunsch H, Hill AD, Fu L, Fowler RA, Wang HT, Gomes T, et al. New opioid use after invasive mechanical ventilation and hospital discharge. Am J Respir Crit Care Med. 2020;202:568–575. doi: 10.1164/rccm.201912-2503OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Amar-Dolan LG, Horn MH, O’Connell B, Parsons SK, Roussin CJ, Weinstock PH, et al. “This is how hard it is”: family experience of hospital-to-home transition with a tracheostomy. Ann Am Thorac Soc. 2020;17:860–868. doi: 10.1513/AnnalsATS.201910-780OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Baker CD. Take me home to the place I belong: discharging the tracheostomy-dependent child. Ann Am Thorac Soc. 2020;17:809–810. doi: 10.1513/AnnalsATS.202004-349ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Greenberg JA, Quinn TV, Gerhart J, Shah RC. Surrogates and physician preferences regarding the continuation of mechanical ventilation among critically ill adults. Ann Am Thorac Soc. 2020;17:1448–1454. doi: 10.1513/AnnalsATS.202003-239OC. [DOI] [PubMed] [Google Scholar]
- 152.Neville TH, Tarn DM, Pavlish CL, Wenger NS. The community perspective on potentially inappropriate treatment. Ann Am Thorac Soc. 2020;17:854–859. doi: 10.1513/AnnalsATS.201912-890OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Rolnick JA, Ersek M, Wachterman MW, Halpern SD. The quality of end-of-life care among ICU versus ward decedents. Am J Respir Crit Care Med. 2020;201:832–839. doi: 10.1164/rccm.201907-1423OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Wunsch H. When intensive care becomes good end-of-life care. Am J Respir Crit Care Med. 2020;201:762. doi: 10.1164/rccm.202001-0076ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Vranas KC, Ouyang D, Lin AL, Slatore CG, Sullivan DR, Kerlin MP, et al. Gender differences in authorship of critical care literature. Am J Respir Crit Care Med. 2020;201:840–847. doi: 10.1164/rccm.201910-1957OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Antkowiak MC, Parsons PE, Stapleton RD. Slow progress toward gender equality in critical care medicine. Am J Respir Crit Care Med. 2020;201:763–764. doi: 10.1164/rccm.202002-0334ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Burns KEA, Yan SD, Thorpe K, Bhimani Z, Marshall JC. Three decades of collaboration in critical care research. Am J Respir Crit Care Med. 2020;201:872–875. doi: 10.1164/rccm.202001-0098LE. [DOI] [PubMed] [Google Scholar]
- 158.Baron RM, Christman JW, Liu KD, Matthay MA, Self WH, McVerry BJ, et al. Monitoring research blood sampling in critically ill patients: avoiding iatrogenic anemia. Am J Respir Crit Care Med. 2020;202:885–887. doi: 10.1164/rccm.202001-0003LE. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.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]
- 160.Krutsinger DC, O’Leary KL, Ellenberg SS, Cotner CE, Halpern SD, Courtright KR. A randomized controlled trial of behavioral nudges to improve enrollment in critical care trials. Ann Am Thorac Soc. 2020;17:1117–1125. doi: 10.1513/AnnalsATS.202003-194OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Garland A, Marrie RA, Wunsch H, Yogendran M, Chateau D. Accuracy of administrative hospital data to identify use of life support modalities: a Canadian study. Ann Am Thorac Soc. 2020;17:229–235. doi: 10.1513/AnnalsATS.201902-106OC. [DOI] [PubMed] [Google Scholar]
- 162.Boncyk CS, Jelly CA, Freundlich RE. The blessing and the curse of the administrative database. Ann Am Thorac Soc. 2020;17:174–175. doi: 10.1513/AnnalsATS.201906-430ED. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Kudchadkar SR, Carroll CL. Using social media for rapid information dissemination in a pandemic: #PedsICU and coronavirus disease 2019. Pediatr Crit Care Med. 2020;21:e538–e546. doi: 10.1097/PCC.0000000000002474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Acho M, Lee AC, Lee BW. Ventilators for nonintensivists: basic ventilator parameters. ATS Scholar. 2020;1:194. doi: 10.34197/ats-scholar.2020-0041VO. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Acho M, Lee AC, Lee BW. Ventilators for nonintensivists. Reasonable initial ventilator settings for patients with acute respiratory distress syndrome. ATS Scholar. 2020;1:197–198. doi: 10.34197/ats-scholar.2020-0052VO. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Acho M, Lee AC, Lee BW. Ventilators for nonintensivists: basic modes and breath types. ATS Scholar. 2020;1:195–196. doi: 10.34197/ats-scholar.2020-0044VO. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Acho M, Lee AC, Lee BW. Ventilators for nonintensivists: monitoring initial ventilator settings in patients with acute respiratory distress syndrome. ATS Scholar. 2020;1:331–332. doi: 10.34197/ats-scholar.2020-0057VO. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Hester TB, Cartwright JD, DiGiovine DG, Karlic KJ, Kercheval JB, DiGiovine B, et al. Training and deployment of medical students as respiratory therapist extenders during COVID-19. ATS Scholar. 2020;1:145–151. doi: 10.34197/ats-scholar.2020-0049PS. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Lee JD, Albert TJ, Carmona H, Johnson NJ, Luks AM, Steinbach TC, et al. Just-in-time tools for training non–critical care providers: basics of respiratory failure. ATS Scholar. 2020;1:170–177. doi: 10.34197/ats-scholar.2020-0039IN. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Steinbach TC, Albert TJ, Carmona HD, Johnson NJ, Kritek PA, Lee JD, et al. Just-in-time tools for training non–critical care providers: troubleshooting problems in the ventilated patient. ATS Scholar. 2020;1:178–185. doi: 10.34197/ats-scholar.2020-0038IN. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Petri CR, Brenner LN, Calhoun TF, Chivukula RR, Maley JH, Vinarsky V, et al. The Fast Literature Assessment and Review (FLARE) initiative: a collaborative effort for timely literature appraisal during the coronavirus disease pandemic. ATS Scholar. 2020;1:186–193. doi: 10.34197/ats-scholar.2020-0045IN. [DOI] [PMC free article] [PubMed] [Google Scholar]
