Advancing precision medicine for acute respiratory distress syndrome

Jeremy R Beitler, B Taylor Thompson, Rebecca M Baron, Julie A Bastarache, Loren C Denlinger, Laura Esserman, Michelle N Gong, Lisa M LaVange, Roger J Lewis, John C Marshall, Thomas R Martin, Daniel F McAuley, Nuala J Meyer, Marc Moss, Lora A Reineck, Eileen Rubin, Eric P Schmidt, Theodore J Standiford, Lorraine B Ware, Hector R Wong, Neil R Aggarwal, Carolyn S Calfee, Jeremy R Beitler, B Taylor Thompson, Rebecca M Baron, Julie A Bastarache, Loren C Denlinger, Laura Esserman, Michelle N Gong, Lisa M LaVange, Roger J Lewis, John C Marshall, Thomas R Martin, Daniel F McAuley, Nuala J Meyer, Marc Moss, Lora A Reineck, Eileen Rubin, Eric P Schmidt, Theodore J Standiford, Lorraine B Ware, Hector R Wong, Neil R Aggarwal, Carolyn S Calfee

Abstract

Acute respiratory distress syndrome (ARDS) is a heterogeneous clinical syndrome. Understanding of the complex pathways involved in lung injury pathogenesis, resolution, and repair has grown considerably in recent decades. Nevertheless, to date, only therapies targeting ventilation-induced lung injury have consistently proven beneficial, and despite these gains, ARDS morbidity and mortality remain high. Many candidate therapies with promise in preclinical studies have been ineffective in human trials, probably at least in part due to clinical and biological heterogeneity that modifies treatment responsiveness in human ARDS. A precision medicine approach to ARDS seeks to better account for this heterogeneity by matching therapies to subgroups of patients that are anticipated to be most likely to benefit, which initially might be identified in part by assessing for heterogeneity of treatment effect in clinical trials. In October 2019, the US National Heart, Lung, and Blood Institute convened a workshop of multidisciplinary experts to explore research opportunities and challenges for accelerating precision medicine in ARDS. Topics of discussion included the rationale and challenges for a precision medicine approach in ARDS, the roles of preclinical ARDS models in precision medicine, essential features of cohort studies to advance precision medicine, and novel approaches to clinical trials to support development and validation of a precision medicine strategy. In this Position Paper, we summarise workshop discussions, recommendations, and unresolved questions for advancing precision medicine in ARDS. Although the workshop took place before the COVID-19 pandemic began, the pandemic has highlighted the urgent need for precision therapies for ARDS as the global scientific community grapples with many of the key concepts, innovations, and challenges discussed at this workshop.

Conflict of interest statement

Declaration of interests JRB reports grants from the US National Institutes of Health (NIH) and Quantum Leap Healthcare Collaborative and personal fees from Sedana Medical and Hamilton Medical, outside of the submitted work. BTT reports grants from NIH and personal fees from Bayer, Novartis, and Thetis, outside of the submitted work. RMB reports grants from NIH and personal fees from Merck and Genentech, outside of the submitted work. JAB reports grants from NIH, outside of the submitted work. LCD reports grants from NIH, grants and personal fees from AstraZeneca, and personal fees from Sanofi-Regeneron, outside of the submitted work. LE reports being an uncompensated board member of Quantum Leap Healthcare Collaborative. MNG reports grants from NIH and the Agency for Healthcare Research and Quality, outside of the submitted work. RJL is the senior medical scientist at Berry Consultants, a statistical consulting firm that specialises in innovative clinical trial design, including Bayesian adaptive and platform trial designs. JCM is chair of the International Forum for Acute Care Trialists, and reports personal fees from AM Pharma, Gilead Pharmaceuticals, and the Society of Critical Care Medicine, outside of the submitted work. TRM reports personal fees from Novartis Pharmaceuticals, Translate Bio, NIH, the Gates Foundation, and TVM Capital Life Science, outside of the submitted work. DFM reports personal fees for consultancy from GlaxoSmithKline, Boehringer Ingelheim, Bayer, and Novartis, and he holds a patent issued to his institution for a treatment for acute respiratory distress syndrome (ARDS), outside of the submitted work. NJM reports grants from NIH, Athersys, the Marcus Foundation, BioMarck, and Quantum Leap Healthcare Collaborative, outside of the submitted work. MM reports grants from NIH, outside of the submitted work. ER is the president of the ARDS Foundation. EPS reports grants from NIH and equity interest in IHP Therapeutics, outside of the submitted work. LBW reports personal fees from Merck, Bayer, Boehringer Ingelheim, CSL Behring, Quark, Foresee Pharmaceuticals, and Citius, and grants from CSL Behring and Genentech, outside of the submitted work. HRW reports grants from NIH and holds a patent for sepsis biomarkers for prognostic and predictive enrichment. CSC reports grants from NIH and Quantum Leap Healthcare Collaborative, grants and personal fees from Genentech and Bayer, and personal fees from Quark Pharmaceuticals, Prometric, Gen1e Life Sciences, and Vasomune, outside of the submitted work. The other authors declared no conflicts of interest.

Copyright © 2022 Elsevier Ltd. All rights reserved.

Figures

Figure
Figure
Proposed research schema to advance precision medicine pharmacotherapy in acute respiratory distress syndrome *Phase 2 and 3 trials might use a platform design if multiple candidate therapies are deemed ready for testing and amenable to a platform. Other trial designs might also be considered. The number of simultaneous arms need not be constant and might depend on resources, enrolment rate, and number and priority of candidate treatments, among other factors. †Biomarker signatures are denoted by coloured arrows.

References

    1. Bellani G, Laffey JG, Pham T, et al. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA. 2016;315:788–800.
    1. Brower RG, Matthay MA, Morris A, Schoenfeld D, Thompson BT, Wheeler A. 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.
    1. Ranieri VM, Rubenfeld GD, Thompson BT, et al. Acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012;307:2526–2533.
    1. Herridge MS, Tansey CM, Matté A, et al. Functional disability 5 years after acute respiratory distress syndrome. N Engl J Med. 2011;364:1293–1304.
    1. Hopkins RO, Weaver LK, Collingridge D, Parkinson RB, Chan KJ, Orme JF., Jr Two-year cognitive, emotional, and quality-of-life outcomes in acute respiratory distress syndrome. Am J Respir Crit Care Med. 2005;171:340–347.
    1. Matthay MA, Zemans RL, Zimmerman GA, et al. Acute respiratory distress syndrome. Nat Rev Dis Primers. 2019;5:18.
    1. Collins FS, Varmus H. A new initiative on precision medicine. N Engl J Med. 2015;372:793–795.
    1. Calfee CS, Delucchi KL, Sinha P, et al. Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised controlled trial. Lancet Respir Med. 2018;6:691–698.
    1. Calfee CS, Delucchi K, Parsons PE, Thompson BT, Ware LB, Matthay MA. Subphenotypes in acute respiratory distress syndrome: latent class analysis of data from two randomised controlled trials. Lancet Respir Med. 2014;2:611–620.
    1. Famous KR, Delucchi K, Ware LB, et al. Acute respiratory distress syndrome subphenotypes respond differently to randomized fluid management strategy. Am J Respir Crit Care Med. 2017;195:331–338.
    1. Robert C, Grob JJ, Stroyakovskiy D, et al. Five-year outcomes with dabrafenib plus trametinib in metastatic melanoma. N Engl J Med. 2019;381:626–636.
    1. Rugo HS, Olopade OI, DeMichele A, et al. Adaptive randomization of veliparib–carboplatin treatment in breast cancer. N Engl J Med. 2016;375:23–34.
    1. Jarjour NN, Erzurum SC, Bleecker ER, et al. Severe asthma: lessons learned from the National Heart, Lung, and Blood Institute Severe Asthma Research Program. Am J Respir Crit Care Med. 2012;185:356–362.
    1. Thille AW, Esteban A, Fernández-Segoviano P, et al. Comparison of the Berlin definition for acute respiratory distress syndrome with autopsy. Am J Respir Crit Care Med. 2013;187:761–767.
    1. Villar J, Pérez-Méndez L, Blanco J, et al. A universal definition of ARDS: the PaO2/FiO2 ratio under a standard ventilatory setting— a prospective, multicenter validation study. Intensive Care Med. 2013;39:583–592.
    1. Guérin C, Reignier J, Richard JC, et al. Prone positioning in severe acute respiratory distress syndrome. N Engl J Med. 2013;368:2159–2168.
    1. Beitler JR. Bedside respiratory physiology to detect risk of lung injury in acute respiratory distress syndrome. Curr Opin Crit Care. 2019;25:3–11.
    1. Amato MBP, Meade MO, Slutsky AS, et al. Driving pressure and survival in the acute respiratory distress syndrome. N Engl J Med. 2015;372:747–755.
    1. Beitler JR, Shaefi S, Montesi SB, et al. Prone positioning reduces mortality from acute respiratory distress syndrome in the low tidal volume era: a meta-analysis. Intensive Care Med. 2014;40:332–341.
    1. Cressoni M, Cadringher P, Chiurazzi C, et al. Lung inhomogeneity in patients with acute respiratory distress syndrome. Am J Respir Crit Care Med. 2014;189:149–158.
    1. Constantin JM, Jabaudon M, Lefrant JY, et al. 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.
    1. Mrozek S, Jabaudon M, Jaber S, et al. Elevated plasma levels of sRAGE are associated with nonfocal CT-based lung imaging in patients with ARDS: a prospective multicenter study. Chest. 2016;150:998–1007.
    1. Calfee CS, Janz DR, Bernard GR, et al. Distinct molecular phenotypes of direct vs indirect ARDS in single-center and multicenter studies. Chest. 2015;147:1539–1548.
    1. Thille AW, Esteban A, Fernández-Segoviano P, et al. Chronology of histological lesions in acute respiratory distress syndrome with diffuse alveolar damage: a prospective cohort study of clinical autopsies. Lancet Respir Med. 2013;1:395–401.
    1. Luo L, Shaver CM, Zhao Z, et al. Clinical predictors of hospital mortality differ between direct and indirect ARDS. Chest. 2017;151:755–763.
    1. Gibelin A, Parrot A, Maitre B, et al. Acute respiratory distress syndrome mimickers lacking common risk factors of the Berlin definition. Intensive Care Med. 2016;42:164–172.
    1. Papazian L, Forel JM, Gacouin A, et al. Neuromuscular blockers in early acute respiratory distress syndrome. N Engl J Med. 2010;363:1107–1116.
    1. National Heart, Lung, and Blood Institute ARDS Clinical Trials Network Comparison of two fluid-management strategies in acute lung injury. N Engl J Med. 2006;354:2564–2575.
    1. Matthay MA, Brower RG, Carson S, et al. Randomized, placebo-controlled clinical trial of an aerosolized β2-agonist for treatment of acute lung injury. Am J Respir Crit Care Med. 2011;184:561–568.
    1. Festic E, Carr GE, Cartin-Ceba R, et al. Randomized clinical trial of a combination of an inhaled corticosteroid and beta agonist in patients at risk of developing the acute respiratory distress syndrome. Crit Care Med. 2017;45:798–805.
    1. Moss M, Guidot DM, Wong-Lambertina M, Ten Hoor T, Perez RL, Brown LAS. The effects of chronic alcohol abuse on pulmonary glutathione homeostasis. Am J Respir Crit Care Med. 2000;161:414–419.
    1. Calfee CS, Matthay MA, Kangelaris KN, et al. Cigarette smoke exposure and the acute respiratory distress syndrome. Crit Care Med. 2015;43:1790–1797.
    1. Ware LB, Zhao Z, Koyama T, et al. Long-term ozone exposure increases the risk of developing the acute respiratory distress syndrome. Am J Respir Crit Care Med. 2016;193:1143–1150.
    1. Goh C, Knight JC. Enhanced understanding of the host-pathogen interaction in sepsis: new opportunities for omic approaches. Lancet Respir Med. 2017;5:212–223.
    1. National Heart, Lung, and Blood Institute ARDS Clinical Trials Network Rosuvastatin for sepsis-associated acute respiratory distress syndrome. N Engl J Med. 2014;370:2191–2200.
    1. Meduri GU, Headley S, Kohler G, et al. Persistent elevation of inflammatory cytokines predicts a poor outcome in ARDS. Plasma IL-1 beta and IL-6 levels are consistent and efficient predictors of outcome over time. Chest. 1995;107:1062–1073.
    1. Parsons PE, Eisner MD, Thompson BT, et al. Lower tidal volume ventilation and plasma cytokine markers of inflammation in patients with acute lung injury. Crit Care Med. 2005;33:1–6. discussion 230–32.
    1. Jabaudon M, Blondonnet R, Pereira B, et al. Plasma sRAGE is independently associated with increased mortality in ARDS: a meta-analysis of individual patient data. Intensive Care Med. 2018;44:1388–1399.
    1. Calfee CS, Gallagher D, Abbott J, Thompson BT, Matthay MA. Plasma angiopoietin-2 in clinical acute lung injury: prognostic and pathogenetic significance. Crit Care Med. 2012;40:1731–1737.
    1. Bos LD, Schouten LR, van Vught LA, et al. Identification and validation of distinct biological phenotypes in patients with acute respiratory distress syndrome by cluster analysis. Thorax. 2017;72:876–883.
    1. Bos LDJ, Scicluna BP, Ong DSY, Cremer O, van der Poll T, Schultz MJ. Understanding heterogeneity in biologic phenotypes of acute respiratory distress syndrome by leukocyte expression profiles. Am J Respir Crit Care Med. 2019;200:42–50.
    1. Sinha P, Delucchi KL, McAuley DF, O'Kane CM, Matthay MA, Calfee CS. Development and validation of parsimonious algorithms to classify acute respiratory distress syndrome phenotypes: a secondary analysis of randomised controlled trials. Lancet Respir Med. 2020;8:247–257.
    1. Rautanen A, Mills TC, Gordon AC, et al. Genome-wide association study of survival from sepsis due to pneumonia: an observational cohort study. Lancet Respir Med. 2015;3:53–60.
    1. Everitt AR, Clare S, Pertel T, et al. IFITM3 restricts the morbidity and mortality associated with influenza. Nature. 2012;484:519–523.
    1. Jones TK, Feng R, Kerchberger VE, 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.
    1. Reilly JP, Wang F, Jones TK, et al. Plasma angiopoietin-2 as a potential causal marker in sepsis-associated ARDS development: evidence from Mendelian randomization and mediation analysis. Intensive Care Med. 2018;44:1849–1858.
    1. Guillen-Guio B, Lorenzo-Salazar JM, Ma SF, et al. Sepsis-associated acute respiratory distress syndrome in individuals of European ancestry: a genome-wide association study. Lancet Respir Med. 2020;8:258–266.
    1. Christie JD, Wurfel MM, Feng R, et al. Genome wide association identifies PPFIA1 as a candidate gene for acute lung injury risk following major trauma. PLoS One. 2012;7
    1. Kerchberger VE, Bastarache JA, Shaver CM, et al. Haptoglobin-2 variant increases susceptibility to acute respiratory distress syndrome during sepsis. JCI Insight. 2019;4
    1. Dickson RP, Erb-Downward JR, Huffnagle GB. Towards an ecology of the lung: new conceptual models of pulmonary microbiology and pneumonia pathogenesis. Lancet Respir Med. 2014;2:238–246.
    1. Dickson RP. The microbiome and critical illness. Lancet Respir Med. 2016;4:59–72.
    1. Dickson RP, Singer BH, Newstead MW, et al. Enrichment of the lung microbiome with gut bacteria in sepsis and the acute respiratory distress syndrome. Nat Microbiol. 2016;1
    1. 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.
    1. McAuley D. Clinical evaluation of a point of care (POC) assay to identify phenotypes in the acute respiratory distress syndrome (NCT04009330) March 20, 2020.
    1. Esteban A, Fernández-Segoviano P, Frutos-Vivar F, et al. Comparison of clinical criteria for the acute respiratory distress syndrome with autopsy findings. Ann Intern Med. 2004;141:440–445.
    1. Gajic O, Dabbagh O, Park PK, et al. Early identification of patients at risk of acute lung injury: evaluation of lung injury prediction score in a multicenter cohort study. Am J Respir Crit Care Med. 2011;183:462–470.
    1. Wood KA, Huang D, Angus DC. Improving clinical trial design in acute lung injury. Crit Care Med. 2003;31(suppl):S305–S311.
    1. Agusti A, Bel E, Thomas M, et al. Treatable traits: toward precision medicine of chronic airway diseases. Eur Respir J. 2016;47:410–419.
    1. Wong HR, Cvijanovich NZ, Anas N, et al. Pediatric Sepsis Biomarker Risk Model-II: redefining the pediatric sepsis biomarker risk model with septic shock phenotype. Crit Care Med. 2016;44:2010–2017.
    1. Wong HR, Caldwell JT, Cvijanovich NZ, et al. Prospective clinical testing and experimental validation of the Pediatric Sepsis Biomarker Risk Model. Sci Transl Med. 2019;11
    1. Sinha P, Delucchi KL, Thompson BT, McAuley DF, Matthay MA, Calfee CS. Latent class analysis of ARDS subphenotypes: a secondary analysis of the statins for acutely injured lungs from sepsis (SAILS) study. Intensive Care Med. 2018;44:1859–1869.
    1. Churchill GA, Airey DC, Allayee H, et al. The Collaborative Cross, a community resource for the genetic analysis of complex traits. Nat Genet. 2004;36:1133–1137.
    1. Abu Toamih Atamni H, Nashef A, Iraqi FA. The Collaborative Cross mouse model for dissecting genetic susceptibility to infectious diseases. Mamm Genome. 2018;29:471–487.
    1. Rittirsch D, Huber-Lang MS, Flierl MA, Ward PA. Immunodesign of experimental sepsis by cecal ligation and puncture. Nat Protoc. 2009;4:31–36.
    1. Kim J, Arnaout L, Remick D. Hydrocortisone, ascorbic acid and thiamine (HAT) therapy decreases oxidative stress, improves cardiovascular function and improves survival in murine sepsis. Shock. 2020;53:460–467.
    1. Ahuja N, Andres-Hernando A, Altmann C, et al. Circulating IL-6 mediates lung injury via CXCL1 production after acute kidney injury in mice. Am J Physiol Renal Physiol. 2012;303:F864–F872.
    1. Imai Y, Parodo J, Kajikawa O, et al. Injurious mechanical ventilation and end-organ epithelial cell apoptosis and organ dysfunction in an experimental model of acute respiratory distress syndrome. JAMA. 2003;289:2104–2112.
    1. López-Aguilar J, Villagrá A, Bernabé F, et al. Massive brain injury enhances lung damage in an isolated lung model of ventilator-induced lung injury. Crit Care Med. 2005;33:1077–1083.
    1. González-López A, López-Alonso I, Aguirre A, et al. Mechanical ventilation triggers hippocampal apoptosis by vagal and dopaminergic pathways. Am J Respir Crit Care Med. 2013;188:693–702.
    1. Proudfoot AG, McAuley DF, Griffiths MJD, Hind M. Human models of acute lung injury. Dis Model Mech. 2011;4:145–153.
    1. Frank JA, Briot R, Lee JW, Ishizaka A, Uchida T, Matthay MA. Physiological and biochemical markers of alveolar epithelial barrier dysfunction in perfused human lungs. Am J Physiol Lung Cell Mol Physiol. 2007;293:L52–L59.
    1. Wurfel MM, Park WY, Radella F, et al. Identification of high and low responders to lipopolysaccharide in normal subjects: an unbiased approach to identify modulators of innate immunity. J Immunol. 2005;175:2570–2578.
    1. Leligdowicz A, Conroy AL, Hawkes M, et al. Validation of two multiplex platforms to quantify circulating markers of inflammation and endothelial injury in severe infection. PLoS One. 2017;12
    1. Moore WC, Meyers DA, Wenzel SE, et al. Identification of asthma phenotypes using cluster analysis in the Severe Asthma Research Program. Am J Respir Crit Care Med. 2010;181:315–323.
    1. Peters MC, Kerr S, Dunican EM, et al. Refractory airway type 2 inflammation in a large subgroup of asthmatic patients treated with inhaled corticosteroids. J Allergy Clin Immunol. 2019;143:104. 13.e14.
    1. Matthay MA, Arabi YM, Siegel ER, et al. Phenotypes and personalized medicine in the acute respiratory distress syndrome. Intensive Care Med. 2020;46:2136–2152.
    1. Stanski NL, Wong HR. Prognostic and predictive enrichment in sepsis. Nat Rev Nephrol. 2020;16:20–31.
    1. Berry SM, Connor JT, Lewis RJ. The platform trial: an efficient strategy for evaluating multiple treatments. JAMA. 2015;313:1619–1620.
    1. Barker AD, Sigman CC, Kelloff GJ, Hylton NM, Berry DA, Esserman LJ. I-SPY 2: an adaptive breast cancer trial design in the setting of neoadjuvant chemotherapy. Clin Pharmacol Ther. 2009;86:97–100.
    1. Israel E, Denlinger LC, Bacharier LB, et al. PrecISE: Precision medicine in severe asthma: an adaptive platform trial with biomarker ascertainment. J Allergy Clin Immunol. 2021;147:1594–1601.
    1. Siddiqui S, Denlinger LC, Fowler SJ, et al. Unmet needs in severe asthma subtyping and precision medicine trials: bridging clinical and patient perspectives. Am J Respir Crit Care Med. 2019;199:823–829.
    1. Woodcock J, LaVange LM. Master protocols to study multiple therapies, multiple diseases, or both. N Engl J Med. 2017;377:62–70.
    1. Schmid P, Cortes J, Pusztai L, et al. Pembrolizumab for early triple-negative breast cancer. N Engl J Med. 2020;382:810–821.
    1. Yu KD, Ye FG, He M, et al. Effect of adjuvant paclitaxel and carboplatin on survival in women with triple-negative breast cancer: a phase 3 randomized clinical trial. JAMA Oncol. 2020;6:1390–1396.
    1. Ivanova A, Israel E, LaVange LM, et al. The precision interventions for severe and/or exacerbation-prone asthma (PrecISE) adaptive platform trial: statistical considerations. J Biopharm Stat. 2020;30:1026–1037.
    1. Horby P, Lim WS, Emberson JR, et al. Dexamethasone in hospitalized patients with Covid-19. N Engl J Med. 2021;384:693–704.
    1. Sterne JAC, Murthy S, Diaz JV, et al. Association between administration of systemic corticosteroids and mortality among critically ill patients with COVID-19: a meta-analysis. JAMA. 2020;324:1330–1341.
    1. National Institutes of Health COVID-19 therapeutics prioritized for testing in clinical trials. Nov 9, 2020.
    1. National Heart, Lung, and Blood Institute Collaborating Network of Networks for Evaluating COVID-19 and Therapeutic Strategies (CONNECTS) 2020.
    1. QuantumLeap Healthcare Collaborative I-SPY COVID-19 TRIAL: an adaptive platform trial for critically ill patients (NCT04488081) July 27, 2020.
    1. Pan H, Peto R, Henao-Restrepo AM, et al. Repurposed antiviral drugs for Covid-19 — interim WHO Solidarity Trial results. N Engl J Med. 2021;384:497–511.
    1. Angus DC, Derde L, Al-Beidh F, et al. Effect of hydrocortisone on mortality and organ support in patients with severe COVID-19: the REMAP-CAP COVID-19 corticosteroid domain randomized clinical trial. JAMA. 2020;324:1317–1329.
    1. Angus DC, Berry S, Lewis RJ, et al. The REMAP-CAP (randomized embedded multifactorial adaptive platform for community-acquired pneumonia) study: rationale and design. Ann Am Thorac Soc. 2020;17:879–891.
    1. Mathew D, Giles JR, Baxter AE, et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science. 2020;369
    1. Rosenblatt M. How academia and the pharmaceutical industry can work together: the president's lecture, annual meeting of the American Thoracic Society, San Francisco, California. Ann Am Thorac Soc. 2013;10:31–38.
    1. Viele K, Broglio K, McGlothlin A, Saville BR. Comparison of methods for control allocation in multiple arm studies using response adaptive randomization. Clin Trials. 2020;17:52–60.
    1. Murray DD, Babiker AG, Baker JV, et al. Design and implementation of the multi-arm, multi-stage Therapeutics for Inpatients with COVID-19 (TICO) platform master protocol: an Accelerating COVID-19 Therapeutic Interventions and Vaccines (ACTIV) initiative. medRxiv. 2021 doi: 10.1101/2020.11.08.20227876. published online April 8. (preprint).
    1. Spragg RG, Bernard GR, Checkley W, et al. Beyond mortality: future clinical research in acute lung injury. Am J Respir Crit Care Med. 2010;181:1121–1127.
    1. Auriemma CL, Zhuo H, Delucchi K, et al. Acute respiratory distress syndrome-attributable mortality in critically ill patients with sepsis. Intensive Care Med. 2020;46:1222–1231.
    1. Abrams D, Montesi SB, Moore SKL, et al. Powering bias and clinically important treatment effects in randomized trials of critical illness. Crit Care Med. 2020;48:1710–1719.
    1. Gao Smith F, Perkins GD, Gates S, et al. 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.
    1. Beitler JR, Sarge T, Banner-Goodspeed VM, et al. Effect of titrating positive end-expiratory pressure (PEEP) with an esophageal pressure-guided strategy vs an empirical high PEEP-FiO2 strategy on death and days free from mechanical ventilation among patients with acute respiratory distress syndrome: a randomized clinical trial. JAMA. 2019;321:846–857.
    1. Novack V, Beitler JR, Yitshak-Sade M, et al. Alive and ventilator free: a hierarchical, composite outcome for clinical trials in the acute respiratory distress syndrome. Crit Care Med. 2020;48:158–166.
    1. WHO WHO R&D blueprint novel coronavirus Covid-19 therapeutic trial synopsis. Feb 18, 2020.
    1. Needham DM, Sepulveda KA, Dinglas VD, 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.
    1. Mikkelsen ME, Christie JD, Lanken PN, et al. The adult respiratory distress syndrome cognitive outcomes study: long-term neuropsychological function in survivors of acute lung injury. Am J Respir Crit Care Med. 2012;185:1307–1315.
    1. Needham DM, Dinglas VD, Morris PE, et al. Physical and cognitive performance of patients with acute lung injury 1 year after initial trophic versus full enteral feeding. EDEN trial follow-up. Am J Respir Crit Care Med. 2013;188:567–576.
    1. Esserman LJ, Berry DA, DeMichele A, et al. Pathologic complete response predicts recurrence-free survival more effectively by cancer subset: results from the I-SPY 1 TRIAL--CALGB 150007/150012, ACRIN 6657. J Clin Oncol. 2012;30:3242–3249.
    1. Nosengo N. New tricks for old drugs. Nature. 2016;534:314–316.
    1. Huang R, Zhu H, Shinn P, et al. The NCATS Pharmaceutical Collection: a 10-year update. Drug Discov Today. 2019;24:2341–2349.
    1. Papi A, Brightling C, Pedersen SE, Reddel HK. Asthma. Lancet. 2018;391:783–800.
    1. Stein ML, Collins MH, Villanueva JM, et al. Anti-IL-5 (mepolizumab) therapy for eosinophilic esophagitis. J Allergy Clin Immunol. 2006;118:1312–1319.
    1. Wollenberg A, Howell MD, Guttman-Yassky E, et al. Treatment of atopic dermatitis with tralokinumab, an anti-IL-13 mAb. J Allergy Clin Immunol. 2019;143:135–141.

Source: PubMed

3
S'abonner