Skip to main content
Erschienen in: Respiratory Research 1/2020

Open Access 01.12.2020 | Study protocol

Acute respiratory distress syndrome subphenotypes and therapy responsive traits among preclinical models: protocol for a systematic review and meta-analysis

verfasst von: Adrien Carla, Bruno Pereira, Hanifa Boukail, Jules Audard, Nathalie Pinol-Domenech, Manuela De Carvalho, Raiko Blondonnet, Ruoyang Zhai, Dominique Morand, Céline Lambert, Vincent Sapin, Lorraine B. Ware, Carolyn S. Calfee, Julie A. Bastarache, John G. Laffey, Nicole P. Juffermans, Lieuwe D. Bos, Antonio Artigas, Patricia R. M. Rocco, Michael A. Matthay, Daniel F. McAuley, Jean-Michel Constantin, Matthieu Jabaudon, for the ESICM Translational Biology Group of the Acute Respiratory Failure section

Erschienen in: Respiratory Research | Ausgabe 1/2020

Abstract

Background

Subphenotypes were recently reported within clinical acute respiratory distress syndrome (ARDS), with distinct outcomes and therapeutic responses. Experimental models have long been used to mimic features of ARDS pathophysiology, but the presence of distinct subphenotypes among preclinical ARDS remains unknown. This review will investigate whether: 1) subphenotypes can be identified among preclinical ARDS models; 2) such subphenotypes can identify some responsive traits.

Methods

We will include comparative preclinical (in vivo and ex vivo) ARDS studies published between 2009 and 2019 in which pre-specified therapies were assessed (interleukin (IL)-10, IL-2, stem cells, beta-agonists, corticosteroids, fibroblast growth factors, modulators of the receptor for advanced glycation end-products pathway, anticoagulants, and halogenated agents) and outcomes compared to a control condition. The primary outcome will be a composite of the four key features of preclinical ARDS as per the American Thoracic Society consensus conference (histologic evidence of lung injury, altered alveolar-capillary barrier, lung inflammatory response, and physiological dysfunction). Secondary outcomes will include the single components of the primary composite outcome, net alveolar fluid clearance, and death. MEDLINE, Embase, and Cochrane databases will be searched electronically and data from eligible studies will be extracted, pooled, and analyzed using random-effects models. Individual study reporting will be assessed according to the Animal Research: Reporting of In Vivo Experiments guidelines. Meta-regressions will be performed to identify subphenotypes prior to comparing outcomes across subphenotypes and treatment effects.

Discussion

This study will inform on the presence and underlying pathophysiological features of subphenotypes among preclinical models of ARDS and should help to determine whether sufficient evidence exists to perform preclinical trials of subphenotype-targeted therapies, prior to potential clinical translation.

Systematic review registration

PROSPERO (ID: CRD42019157236).
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12931-020-01337-9.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ALVEOLI
Assessment of low tidal volume and elevated end-expiratory volume to obviate Lung injury
ARDS
Acute respiratory distress syndrome
ARMA
Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome
ARRIVE
Animal research: reporting of in vivo experiments
ATS
American thoracic society
ESICM
European society of intensive care medicine
FACTT
Fluids and catheters treatment trial
FiO2
Fraction of inspired oxygen
HARP-2
Hydroxymethylglutaryl-CoA reductase inhibition with simvastatin in acute lung injury to reduce pulmonary dysfunction–2
ICU
Intensive care unit
IL
Interleukin
PaO2
Partial pressure of arterial oxygen
PEEP
Positive end-expiratory pressure
PRESS
Peer review of electronic search strategies
PRISMA
Preferred reporting items for systematic reviews and meta-analyses
PRISMA-P
Preferred reporting items for systematic reviews and meta-analyses protocols
REDCAP
Research electronic data capture
RAGE
Receptor for advanced glycation end-products
RCT
Randomized clinical trial
ROBINS-I
Risk of bias in nonrandomized studies - of interventions
RR
Relative risk
SMD
Standardized mean differences
sTNFr-1
Soluble tumor necrosis factor receptor 1

Background

The acute respiratory distress syndrome (ARDS) is a life-threatening clinical syndrome of rapid-onset pulmonary failure characterized by a dysregulated inflammatory response that results in severe respiratory failure and the need for mechanical ventilation [1]. Although the prevalence of ARDS is low (1–5/10,000 inhabitants), its incidence is high in critically ill patients, as the syndrome is present in approximately 10% of patients upon admission to the intensive care unit (ICU) and in 24% of ICU patients under mechanical ventilation support [2]. Despite decades of research, mortality due to ARDS remains high, with significant long-term comorbidity and reduced quality of life in survivors [2]. Various pharmacotherapeutic agents have failed to improve ARDS outcomes and current treatment strategy is largely supportive, based on optimized ventilatory settings [1]. A key reason for the lack of specific pharmacological therapy is likely due to the heterogeneity within ARDS, which is diagnosed by a constellation of signs as defined by the Berlin criteria [3].
The clinical and biological heterogeneity within syndromes such as sepsis ARDS makes it essential to identify more homogeneous subphenotypes when investigating potential therapies [4, 5]. There has been recent recognition of distinct subphenotypes (on the basis of clinical/biochemical variables, natural history, disease manifestation, and/or treatment response without any implication about mechanism) and endotypes (defined by a distinct functional/pathobiological mechanism) within patients with ARDS [4, 6, 7]. Therefore, while benefit has been demonstrated in numerous preclinical studies for many candidate therapies, most have failed to translate to improved patient outcomes in randomized clinical trials (RCTs), suggesting that the appropriate subset of patients to target with the novel therapies may not have been correctly identified [8].
The retrospective analysis of data from two large multicenter RCTs (the “ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome [ARMA] and “assessment of low tidal volume and elevated end-expiratory volume to obviate lung injury” [ALVEOLI] trials) [9] has first identified at least two ARDS subphenotypes within clinical ARDS which were differentiated by the presence of shock, metabolic acidosis and a higher inflammatory status (interleukin (IL)-6 and soluble tumor necrosis factor receptor (sTNFr)-1). These two phenotypes are sometimes referred to as “endotypes” because they seem defined by distinct degrees of inflammation, but their underlying mechanisms remain largely unknown. However, these subphenotypes were stable over the first 3 days of enrolment in both RCTs, suggesting that they are linked to true biological processes. The hyper-inflammatory subphenotype demonstrated significantly worse outcomes when compared to the hypo-inflammatory subphenotype, with higher mortality and lesser ventilator-free and organ failure-free days. The analysis of plasma biomarkers in patients with ARDS previously enrolled in the MARS prospective cohort also identified two biological subphenotypes, which were very similar to those found earlier and were again associated with distinct mortality rates [10]. In the ALVEOLI trial, where a lower positive end-expiratory pressure (PEEP) strategy was compared to a higher PEEP strategy, the two subphenotypes demonstrated a distinct response in terms of major outcomes, suggesting the potential for using this subphenotypic classification to identify therapy responsive traits. This hypothesis was further confirmed through secondary analysis of the fluids and catheters treatment trial (FACTT) in which the two inflammatory subphenotypes had distinct effects of the randomly assigned fluid management strategy on 90-day mortality [11]. The potential for the inflammation-based subphenotypic classification was again demonstrated in a secondary analysis of the hydroxymethylglutaryl-CoA reductase inhibition with simvastatin in acute lung injury to reduce pulmonary dysfunction–2 (HARP-2) trial, a multicenter trial investigating the potential of simvastatin as an anti-inflammatory therapy for ARDS. Subphenotyping based on the hypo- versus hyper-inflammatory profile identified a therapy response trait as evidenced by the improved survival of patients classified as hyper-inflammatory and randomized to simvastatin [12].
Translation of basic research findings to clinical practice remains daunting because of the heterogeneity and complexity of ARDS [13]. A substantial number of preclinical models of experimental lung injury have been used to investigate mechanisms of lung injury and test novel therapies [1416]. Although none of these experimental models fully reproduces all features of human lung injury, most of them are based on the reproduction, in animals, of known risk factors for ARDS, such as pulmonary or extrapulmonary sepsis, lipid embolism such as when secondary to bone fracture, and acid aspiration, among other clinical risks [14, 15, 17]. Recently, more translational models of acute lung injury have been used, such as the ex vivo human lung preparation [1821]. Although the nature of the insult leading to acute lung injury in preclinical models grossly influences one or more features of human ARDS, the presence of subphenotypes among experimental models of ARDS has never been systematically assessed to date.

Study question

We hypothesize that ARDS subphenotypes may exist among preclinical ARDS studies and that they may have distinct treatment response to select therapies.
To test this hypothesis, we designed a systematic review and meta-analysis of controlled preclinical trials in experimental, − in vivo (animal) and ex vivo (human lung preparation) -, models of ARDS. The primary goal of the review is to compare the key features of preclinical ARDS, as defined by the American Thoracic Society (ATS) consensus conference (histologic evidence of lung injury, altered alveolar-capillary barrier, lung inflammatory response, and physiological dysfunction) [15], in lung-injured experimental groups treated or not with any of some prespecified candidate therapies.

Methods

Protocol and registration

The systematic review and meta-analysis protocol originated from discussions between our team of translational research scientists (AC, JA, RB, DM, VS, JMC, MJ), information system specialists (NPD, MDC), a statistical and methodologist (BP), and the European Society of Intensive Care Medicine (ESICM) Translational Biology Group of the Acute Respiratory Failure section. The protocol was registered on PROSPERO (ID: CRD42019157236) in December 2019.

Study eligibility criteria

We will include controlled comparative studies (randomized and non-randomized) of preclinical acute lung injury/ARDS published from 2009 to 2019 that evaluate the efficacy and safety outcomes of a number of predefined therapies (stem cells, fibroblast growth factors, IL-2, IL-10, beta-agonists, corticosteroids, modulators of the receptor for advanced glycation end-products (RAGE) pathway, anticoagulants, and halogenated agents). In order to collect sufficient data to test our hypotheses, it was decided a priori to include only studies in which at least two features, out of the four key features of preclinical ARDS proposed by the 2011 Official ATS Workshop Report [15], are assessed.

Preclinical model eligibility criteria

We will include preclinical in vivo animal and ex vivo human models of experimentally induced acute lung injury that mimic at least some aspects of the pathophysiology of humans with ARDS according to the ATS consensus criteria [15]. The inclusion of a wide range of acute lung injury models should enhance the generalizability of our study findings. Acute lung injury in experimental models may be induced by several methods (Table 1). These include intravenous, intratracheal or intraperitoneal administration of bacteria or endotoxin, induction of injury by the ventilator (ventilator-induced lung injury), chemical agents (oleic acid, hydrochloric acid), trauma, shock (for example hemorrhagic or induced by sepsis), or remote organ injury (for example pancreatitis, ischemia-reperfusion). We will exclude neonatal animal models of acute lung injury because the mechanisms of disease, and efficacy of treatments, are likely to be different in this specific setting.
Table 1
Preclinical models of acute respiratory distress syndrome included in the review
Oleic acid
Lipopolysaccharides
Acid aspiration
Hyperoxia
Saline lavage
Pulmonary ischemia/reperfusion
Nonpulmonary ischemia/reperfusion
Intravenous bacteria
Intrapulmonary bacteria
Peritonitis
Cecal ligation and puncture
Trauma/Shock
Mechanical ventilation (ventilator-induced lung injury)
Chemical or chemotherapeutic injury (such as detergents)
Pancreatitis
Burns

Interventions

Preclinical intervention groups will include animals and ex vivo human lungs from studies that examine the therapeutic use of stem cells (and their derived microvesicles), fibroblast growth factors, IL-2, IL-10, beta-agonists, corticosteroids, RAGE modulators, anticoagulants, and halogenated agents such as isoflurane, sevoflurane, and desflurane (Table 2). These interventions have been selected a priori because they are the main areas of interest of our research groups [2230]. In addition to studies of therapies that are administered after lung injury is modeled, experiments using pretreatment will also be included since they are clinically relevant for the prevention of ARDS. Moreover, studies using a co-treatment or multiple injurious hits will be included.
Table 2
Predefined list of select therapies included in the meta-analysis
Class
Example of therapies
Interleukins
IL-10, IL-2
Cell therapy
MSCs, MSC-derived microvesicles
Adrenergic beta-agonists
Salbutamol, albuterol, sultanol, proventil
Growing factors
Fibroblast growing factors, keratinocyte growth factor, endothelial growth factor
RAGE modulators
Anti-RAGE antibody, recombinant soluble RAGE, gene knockout, endogenous secretory RAGE
Corticosteroids
Glucocorticoids, mineralocorticoids, 3-oxo steroid, 17-ketosteroids, domolene, cortifan, epicortisol, komed-hc, heb-cort, prednisone, encortone, panasol, deltacortisone, decortisyl, decortin, predeltin, orasone, prednidib, prednisone, dacortin, cortancyl, solupred, deltasone, pronisone, sterapred, encorton, panafcort, delta-dome, dehydrocortisone, prednisonum, lisacort, ultracorten, winpred, metacortandracin, meticorten, prednisolone, predate, prednisolone, di-adreson f, delta-1-hydrocortisone, prednisolonum, sterane, meti derm, metacortandralone, predonine
Anticoagulant agents
Heparin, antithrombin, plasminogen activator, fibrinolytic agents, tissue factor pathway inhibitor, lipoprotein-associated coagulation inhibitor, thrombin-thrombomodulin complex, thrombin receptor, activated protein C, drotrecogin alfa activated
Halogenated agents
Sevoflurane, desflurane, isoflurane
IL interleukin; MSCs mesenchymal stem (stromal) cells; RAGE receptor for advanced glycation end-products

Comparison

The preclinical comparison group will include data from studies that have had experimental acute lung injury induced but have not been administered any of the prespecified therapies listed in the previous paragraph. This will allow to perform effect size calculations in our meta-analysis to examine how efficacious and safe these therapies are in preclinical ARDS in general, and in preclinical ARDS subphenotypes in particular. In addition, we will use other control groups, such as healthy or sham-injured controls, to examine the severity of preclinical ARDS.

Preclinical primary endpoint

The primary endpoint is a composite of the four key features that define preclinical ARDS, with specific measurements defined by the consensus criteria proposed by the ATS [15].
These include histological evidence of tissue injury (usually based on the lung injury score), alteration of the alveolar-capillary barrier (for example increased concentration of high molecular weight proteins in bronchoalveolar fluid), measures of the inflammatory response in the lung (for example pulmonary leukocytes or neutrophils, cytokines), and measures of physiological dysfunction such as the alveolar-arterial gradient of oxygen concentration and the partial pressure of arterial oxygen (PaO2) to fraction of inspired oxygen (FiO2) ratio. These quantitative features and measurements are summarized in Table 3. We will focus on the “very relevant” measurements defined by the consensus criteria [15].
Table 3
Acute lung injury features and measurements
Features
Relevant measurements
Histological evidence of lung injury
Accumulation of neutrophils in the alveolar/interstitial space
Formation of hyaline membranes
Proteinaceous debris in alveolar space
Proteinaceous debris in alveolar space
Thickening of the alveolar wall
Injury by a standardized histology score
Evidence of hemorrhage
Areas of atelectasis
Gross macroscopic changes such as discoloration of the lungs
Alteration of the alveolar capillary barrier
Increased extravascular lung water content
Accumulation of protein/tracer in airspaces/extravascular space
Total BAL protein concentration
BAL concentration of high molecular weight proteins
Vascular filtration coefficient
Translocation of a protein from the airspaces into plasma
Increased lung lymph flow, lymph protein concentration
Inflammation
BAL total neutrophil counts
Lung MPO activity
Concentrations of cytokines and chemokines in lung tissue, in BAL fluid, and/or in plasma
Physiological dysfunction
Hypoxemia
Increased A-a oxygen difference
BAL bronchoalveolar lavage; MPO myeloperoxidase
Because the assessment of our composite endpoint must reflect that the timing of experimental induction of ARDS may vary across studies, the primary endpoint will be analyzed in categories of time from the induction of ARDS of less than 6 h, between 6 and 24 h, and greater than 24 h. These time points for measurement were selected since the development of inflammation and acute lung injury occurs over time and contributes to death and morbidity in this population [2].

Preclinical secondary endpoints

Secondary endpoints will include death, net alveolar fluid clearance, and each individual component of the primary composite endpoint. The same categories of time will be used for primary and secondary endpoints. Where reported, the occurrence of adverse events will also be documented for each included study. For animal models that use infectious induction of acute lung injury, we will describe pathogen clearance (for example number of bacterial colony-forming units).

Information sources

Search strategies and equation of searching have been developed and tested with the help of two librarians using the Peer Review of Electronic Search Strategies (PRESS) template (see Additional file 1 for the representative search strategy) [31]. We will search on MEDLINE, Embase, and Cochrane databases. Search strategies will use a combination of controlled vocabulary (for example corticoids, mesenchymal stem cells, acute lung injury, respiratory distress syndrome) and keywords (for example mesenchymal stromal cells, acute lung injury, acute/adult respiratory distress syndrome). Vocabulary and syntax will be adjusted across the databases. Two recently published animal filters [32, 33], validated for PubMed/MEDLINE and Embase and amended slightly, will be used to increase relevancy. These filters will be adjusted for use in the other databases where a validated filter is unavailable. Language will be restricted to English studies and no studies published before 2009 will be included. The bibliographies of included studies and relevant reviews will also be hand searched for further preclinical studies.

Study selection

The titles and abstracts of search results will be screened independently by two investigators (AC, MJ). The full text of all potentially eligible studies will be retrieved and reviewed for eligibility. Disagreements between reviewers will be resolved by consensus or by a third member of the systematic review team (JA). Reasons for exclusion of potentially eligible studies will be recorded to enable a transparent selection process and to be in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines developed for proper reporting of clinical systematic reviews [34]. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses Protocols (PRISMA-P) checklist for this review protocol is available as Additional file 2.

Data collection and process and data items

Data will be extracted by four members (AC, JA, RZ, MJ) of the research team and collected into standardized, pilot-tested forms using REDCap electronic data capture tools hosted at CHU Clermont-Ferrand [35]. REDCap (Research Electronic Data Capture) is a secure, web-based application designed to support data capture for research studies, providing: (1) an intuitive interface for validated data entry; (2) audit trails for tracking data manipulation and export procedures; (3) automated export procedures for seamless data downloads to common statistical packages; and (4) procedures for importing data from external sources. Specific data elements are listed in Table 4.
Table 4
Data collection elements
Categories
Main items
Study characteristics
DOI, Num ID (specific number ID for the purpose of the meta-analysis), title, author, year, country and journal of publication, country of study, mono/multicentric
Study population (experimental model)
Animal species or ex vivo model, age, gender, strain, weight, and comorbidity
Type of acute lung injury model
As defined in Table 1, with timing
Intervention
Type (see Table 2), number, timing, mode of administration, dose
Co-intervention
Resuscitation fluids, antibiotics, mechanical ventilation parameters, mode of anesthesia, drugs, etc.
Preclinical endpoints
As defined in Table 3
Risk of bias assessments (Cochrane risk of bias tool)
Randomization, allocation, blinding and completeness of follow-up
Quality of reporting of individual preclinical studies
ARRIVE (Animal Research: Reporting of In Vivo Experiments) guidelines elements
DOI digital object identifier

Assessment of risk of bias

The risk of bias will be evaluated by two reviewers (AC, MJ) for each included preclinical study. Since no validated and standardized risk of bias checklist exists for preclinical studies, we will describe the biases of the included studies using the Cochrane risk of bias assessment [36] and the Risk Of Bias In Nonrandomized Studies - of Interventions (ROBINS-I) [37] tools. Items in this tool include assessments for concealment of allocation, random sequence generation, blinding of personnel and the endpoint measurements, and completeness of endpoint reporting. Each bias criterion will be assigned a value of low, high, or unclear risk of bias for each included study [36].

Assessment of construct validity and external validity

We will also record features that will facilitate judgements of construct validity and external validity [38]. Construct validity in preclinical research concerns the extent to which an experimental system accurately models a clinical entity. These will include: age, gender, weight, species, and strain of animal; presence of comorbidities; type of acute lung injury model; timing, dose and mode of administration of treatment; and use of co-interventions (for example fluid resuscitation, type of anesthesia, use of antibiotics for infectious acute lung injury models) (Table 5). External validity in preclinical research concerns the extent to which cause and effect relationships holdup under varied conditions [39], such as the use of a multicenter preclinical design.
Table 5
Elements of construct validity and external validity
Category
Items
Animal species and strain (for mice)
Human lung preparation, mouse, rat, sheep, pig, rabbit, dog or others
Animal age
Young, middle-aged, and mature, depending on species
Animal gender
Male, female, both
Comorbidity
Yes or no, type
Model of acute lung injury
As defined in Table 1
Severity of lung injury
Lung injury score
Intervention type
The drug, mode of administration and dose
Timing of intervention regarding to acute lung injury induction
Before lung injury, 0 to 1, 1 to 6, and > 6 h after lung injury
Type of control
Injured, non-injured
Use of co-interventions
Resuscitation fluids, use of antibiotics, type of anesthesia, mechanical ventilation strategy
Number of study centers
Single versus multi-center

Description of reporting

We will describe the quality of reporting of the included preclinical studies using the elements of the Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines. The ARRIVE guidelines were developed to enhance the transparent and comprehensive reporting of research methods and results for in vivo animal experiments [40].

Ethics and dissemination

This study does not require ethical approval. Data collected through this systematic review will be managed by the ESICM Translational Biology Group and made available upon reasonable request. The results of this systematic review will be presented at relevant conferences and published in a peer-reviewed journal.

Data analysis

Statistical analyses will be performed using Stata (version 15; StataCorp, College Station, US), Comprehensive Meta-Analysis (version 2; Biostat, Englewood, NJ), and Mplus [41] softwares. For descriptive analyses, data will be presented as mean and standard deviation or median and interquartile range, according to statistical distribution. To address the non-independence of data due to study effect, random-effects models will be performed rather than fixed-effect as certain experimental parameters can have wide variation. The dichotomous endpoints from each included study will be pooled and described as relative risks (RR) and 95% confidence intervals incorporating a random effects modeling approach with the use of forest plots for presentation of the data [42]. Continuous endpoints will be pooled using the ratio of weighted means method with inverse variance random effects modeling and Hedges’s standardized mean differences (SMD) [43]. When necessary, the RRs reported in individual studies will be converted into SMD [44]. The statistical heterogeneity of included preclinical studies will be measured using the I2 [45], which will be used to measure heterogeneity with 25, 50 and 75% indicating low, moderate and high heterogeneity, respectively. Egger’s test and funnel plots will be used to assess publication bias. In the absence of bias, studies will be distributed evenly around the mean effect size because of random sampling error.
Then, meta-regression will be conducted, with covariates determined according to the aforementioned analyses and to clinical and/or biological relevance, and methodological issues such as the categories of time from ARDS induction. Particular attention will be paid to the study of multicollinearity and interactions between covariates 1) studying the relationships between the covariates and 2) evaluating the impact of adding or removing variables to or from the multivariable model. Whenever possible in appropriate data, latent class analyses will be carried out to identify subphenotypes within experimental models of ARDS, associated with distinct outcomes and differential responses to therapy. Statistical analyses will be conducted including all controlled comparative studies, with the type of study considered as stratification variable (randomized and non-randomized).
When the number of studies and sample size will permit, analyses will be performed by study type. Several subgroup analyses to examine preclinical heterogeneity will be conducted on the primary endpoint. These analyses will include “clinical”/biological features (such as the type of experimental model; animal age, gender, weight, species, and strain; presence of comorbidities; cause of experimental ARDS; severity of the ARDS model; type of tested therapies (Table 2); type of controls; type of anesthesia; use of co-interventions, antibiotics, and mechanical ventilation settings; multiple-hit versus single-hit model of ARDS) and methodological features (such as single versus multi-center study, presence of an a priori sample size calculation). These subgroup analyses will be exploratory and the results will be interpreted with caution. In addition, subphenotypes that may be identified among preclinical studies will be compared to those already described in Humans (and those that could be identified in the future) using the same set of variables than in patients such as plasma levels of some proinflammatory cytokines, when applicable [6, 7].
Sensitivity analyses will be conducted to assess how including and excluding studies influences the results. More precisely, sensitivity analyses will be performed to measure the impact of high heterogeneity or a methodological quality estimated too low, and risk of bias assessments. In addition, sensitivity analyses will be performed, when possible, to assess the effects of the nature, sequence, and timing of all the injurious hits that are used in studies based on multiple-hit models of experimental ARDS.

Discussion

The results of this systematic review and meta-analysis will inform translational and clinical scientists, researchers, and clinicians internationally regarding the existing preclinical evidence for subphenotypes among preclinical, in vivo or ex vivo models of ARDS. Such data will be crucial to confirm the potential of personalized ARDS medicine [6, 8, 46], i.e. the promise to deliver the right -targeted- treatment to the right subject. These data will also inform on the potential value of testing candidate therapies (including therapies that have already been tested in previous studies) that target distinct functional and/or pathobiological processes (such as lung epithelial injury and alveolar inflammation [9, 47]) in select experimental models that might best mimic ARDS subphenotypes described in patients to date [6]. For example, although it seems plausible that tracheal instillation with hydrochloric acid causes prominent injury to the alveolar epithelium and live bacteria (such as Streptococcus pneumoniae) causes major alveolar inflammation profiles, it remains unknown whether these models might have reliable value to represent clinical subphenotypes of higher inflammation and of higher degree of lung epithelial injury, respectively [6].
Our review is timely since there is an increasing amount of research dedicated to the identification of ARDS subphenotypes, their underlying mechanisms, and their potential responsive treats [4, 7, 912, 4852]. The results from this study should therefore be useful in order to generate hypotheses on whether some therapies (see Table 2) might differentially benefit to some preclinical subphenotypes, as compared to others. In case of positive results, this might open new perspectives on preclinical precision therapies, with the ultimate goal of future clinical translation. However, the predefined, limited number of therapies investigated through our review (see Table 2) is a limitation that should be acknowledged; this limited number reflects both our specific research topics of interest and a balance between scientific validity and feasibility. Our results will inform on whether the nature of the lung injurious hit used in published experimental models of ARDS could identify subphenotypes with distinct degrees of severity and treatment effects. Yet, we might not be able to distinguish such subphenotypes using individual subject data analysis in a given experimental model, such as recently reported in a secondary analysis of a murine cecal ligation and puncture sepsis model, with one subphenotype characterized by faster deterioration and more severe inflammation than the other and distinct responses to some interventions (immediate versus delayed antibiotics and fluids) [53].
In a broader perspective, we also hope this review will increase our knowledge on, and identify challenges and barriers related to, the conduct of preclinical ARDS studies. Ultimately, it will also inform and enrich future preclinical and clinical precision, subphenotype-based, research that should aid the translation of novel therapies for ARDS.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12931-020-01337-9.

Acknowledgements

Not applicable

Patient and public involvement (PPI) statement

This research was done without patient involvement. Patients were not invited to comment on the study design and were not consulted to develop patient relevant outcomes or interpret the results. Patients were not invited to contribute to the writing or editing of this document for readability or accuracy.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Thompson BT, Chambers RC, Liu KD. Acute respiratory distress syndrome. N Engl J Med. 2017;377:562–72.PubMedCrossRef Thompson BT, Chambers RC, Liu KD. Acute respiratory distress syndrome. N Engl J Med. 2017;377:562–72.PubMedCrossRef
2.
Zurück zum Zitat Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, 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.PubMedCrossRef Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, 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.PubMedCrossRef
3.
Zurück zum Zitat Definition Task Force ARDS, Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND. Caldwell E, et al. acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012;307:2526–33. Definition Task Force ARDS, Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND. Caldwell E, et al. acute respiratory distress syndrome: the Berlin Definition. JAMA. 2012;307:2526–33.
4.
Zurück zum Zitat Prescott HC, Calfee CS, Thompson BT, Angus DC, Liu VX. Toward smarter lumping and smarter splitting: rethinking strategies for Sepsis and acute respiratory distress syndrome clinical trial design. Am J Respir Crit Care Med. 2016;194:147–55.PubMedPubMedCentralCrossRef Prescott HC, Calfee CS, Thompson BT, Angus DC, Liu VX. Toward smarter lumping and smarter splitting: rethinking strategies for Sepsis and acute respiratory distress syndrome clinical trial design. Am J Respir Crit Care Med. 2016;194:147–55.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Seymour CW, Kennedy JN, Wang S, Chang C-CH, Elliott CF, Xu Z, et al. Derivation, Validation, and Potential Treatment Implications of Novel Clinical Phenotypes for Sepsis. JAMA. 2019;321(20):2003–17.PubMedPubMedCentralCrossRef Seymour CW, Kennedy JN, Wang S, Chang C-CH, Elliott CF, Xu Z, et al. Derivation, Validation, and Potential Treatment Implications of Novel Clinical Phenotypes for Sepsis. JAMA. 2019;321(20):2003–17.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Jabaudon M, Blondonnet R, Audard J, Fournet M, Godet T, Sapin V, et al. Recent directions in personalised acute respiratory distress syndrome medicine. Anaesth Crit Care Pain Med. 2018;37:251–8.PubMedCrossRef Jabaudon M, Blondonnet R, Audard J, Fournet M, Godet T, Sapin V, et al. Recent directions in personalised acute respiratory distress syndrome medicine. Anaesth Crit Care Pain Med. 2018;37:251–8.PubMedCrossRef
7.
8.
Zurück zum Zitat Beitler JR, Goligher EC, Schmidt M, Spieth PM, Zanella A, Martin-Loeches I, et al. Personalized medicine for ARDS: the 2035 research agenda. Intensive Care Med. 2016;42:756–67.PubMedPubMedCentralCrossRef Beitler JR, Goligher EC, Schmidt M, Spieth PM, Zanella A, Martin-Loeches I, et al. Personalized medicine for ARDS: the 2035 research agenda. Intensive Care Med. 2016;42:756–67.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Calfee CS, Delucchi K, Parsons PE, Thompson BT, Ware LB, Matthay MA, et al. Subphenotypes in acute respiratory distress syndrome: latent class analysis of data from two randomised controlled trials. Lancet Respir Med. 2014;2:611–20.PubMedPubMedCentralCrossRef Calfee CS, Delucchi K, Parsons PE, Thompson BT, Ware LB, Matthay MA, et al. Subphenotypes in acute respiratory distress syndrome: latent class analysis of data from two randomised controlled trials. Lancet Respir Med. 2014;2:611–20.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Bos LD, Schouten LR, van Vught LA, Wiewel MA, Ong DSY, Cremer O, et al. Identification and validation of distinct biological phenotypes in patients with acute respiratory distress syndrome by cluster analysis. Thorax. 2017;72:876–83.PubMedCrossRef Bos LD, Schouten LR, van Vught LA, Wiewel MA, Ong DSY, Cremer O, et al. Identification and validation of distinct biological phenotypes in patients with acute respiratory distress syndrome by cluster analysis. Thorax. 2017;72:876–83.PubMedCrossRef
11.
Zurück zum Zitat Famous KR, Delucchi K, Ware LB, Kangelaris KN, Liu KD, Thompson BT, et al. Acute respiratory distress syndrome subphenotypes respond differently to randomized fluid management strategy. Am J Respir Crit Care Med. 2017;195:331–8.PubMedPubMedCentralCrossRef Famous KR, Delucchi K, Ware LB, Kangelaris KN, Liu KD, Thompson BT, et al. Acute respiratory distress syndrome subphenotypes respond differently to randomized fluid management strategy. Am J Respir Crit Care Med. 2017;195:331–8.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Calfee CS, Delucchi KL, Sinha P, Matthay MA, Hackett J, Shankar-Hari M, et al. Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised controlled trial. Lancet Respir Med. 2018;6:691–8.PubMedPubMedCentralCrossRef Calfee CS, Delucchi KL, Sinha P, Matthay MA, Hackett J, Shankar-Hari M, et al. Acute respiratory distress syndrome subphenotypes and differential response to simvastatin: secondary analysis of a randomised controlled trial. Lancet Respir Med. 2018;6:691–8.PubMedPubMedCentralCrossRef
13.
15.
Zurück zum Zitat Matute-Bello G, Downey G, Moore BB, Groshong SD, Matthay MA, Slutsky AS, et al. An official American Thoracic Society workshop report: features and measurements of experimental acute lung injury in animals. Am J Respir Cell Mol Biol. 2011;44:725–38.PubMedCrossRefPubMedCentral Matute-Bello G, Downey G, Moore BB, Groshong SD, Matthay MA, Slutsky AS, et al. An official American Thoracic Society workshop report: features and measurements of experimental acute lung injury in animals. Am J Respir Cell Mol Biol. 2011;44:725–38.PubMedCrossRefPubMedCentral
16.
Zurück zum Zitat Laffey JG, Kavanagh BP. Fifty years of research in ARDS. Insight into acute respiratory distress syndrome. From models to patients. Am J Respir Crit Care Med. 2017;196:18–28.PubMedCrossRef Laffey JG, Kavanagh BP. Fifty years of research in ARDS. Insight into acute respiratory distress syndrome. From models to patients. Am J Respir Crit Care Med. 2017;196:18–28.PubMedCrossRef
17.
Zurück zum Zitat Rocco PRM, Nieman GF. ARDS: what experimental models have taught us. Intensive Care Med. 2016;42:806–10.PubMedCrossRef Rocco PRM, Nieman GF. ARDS: what experimental models have taught us. Intensive Care Med. 2016;42:806–10.PubMedCrossRef
18.
Zurück zum Zitat McAuley CDF, Hamid GF, Laffey UI, Abbott JG. McKenna J, et al. clinical grade allogeneic human mesenchymal stem cells restore alveolar fluid clearance in human lungs rejected for transplantation. Am J Physiol Lung Cell Mol Physiol. 2014;306(9):L809–15.PubMedPubMedCentralCrossRef McAuley CDF, Hamid GF, Laffey UI, Abbott JG. McKenna J, et al. clinical grade allogeneic human mesenchymal stem cells restore alveolar fluid clearance in human lungs rejected for transplantation. Am J Physiol Lung Cell Mol Physiol. 2014;306(9):L809–15.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Park J, Kim S, Lim H, Liu A, Hu S, Lee J, et al. Therapeutic effects of human mesenchymal stem cell microvesicles in an ex vivo perfused human lung injured with severe E coli pneumonia. Thorax. 2019;74:43–50.PubMedCrossRef Park J, Kim S, Lim H, Liu A, Hu S, Lee J, et al. Therapeutic effects of human mesenchymal stem cell microvesicles in an ex vivo perfused human lung injured with severe E coli pneumonia. Thorax. 2019;74:43–50.PubMedCrossRef
20.
Zurück zum Zitat Lee JW, Fang X, Gupta N, Serikov V, Matthay MA. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc Natl Acad Sci U S A. 2009;106:16357–62.PubMedPubMedCentralCrossRef Lee JW, Fang X, Gupta N, Serikov V, Matthay MA. Allogeneic human mesenchymal stem cells for treatment of E. coli endotoxin-induced acute lung injury in the ex vivo perfused human lung. Proc Natl Acad Sci U S A. 2009;106:16357–62.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Ross JT, Nesseler N, Lee J-W, Ware LB, Matthay MA. The ex vivo human lung: research value for translational science. JCI Insight. 2019;4(11). Ross JT, Nesseler N, Lee J-W, Ware LB, Matthay MA. The ex vivo human lung: research value for translational science. JCI Insight. 2019;4(11).
22.
Zurück zum Zitat Matthay MA, Calfee CS, Zhuo H, Thompson BT, Wilson JG, Levitt JE, et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised phase 2a safety trial. Lancet Respir Med. 2019;7:154–62.PubMedCrossRef Matthay MA, Calfee CS, Zhuo H, Thompson BT, Wilson JG, Levitt JE, et al. Treatment with allogeneic mesenchymal stromal cells for moderate to severe acute respiratory distress syndrome (START study): a randomised phase 2a safety trial. Lancet Respir Med. 2019;7:154–62.PubMedCrossRef
23.
Zurück zum Zitat Matthay MA, Goolaerts A, Howard JP, Lee JW. Mesenchymal stem cells for acute lung injury: preclinical evidence. Crit Care Med. 2010;38:S569–73.PubMedPubMedCentralCrossRef Matthay MA, Goolaerts A, Howard JP, Lee JW. Mesenchymal stem cells for acute lung injury: preclinical evidence. Crit Care Med. 2010;38:S569–73.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Ware LB, Landeck M, Koyama T, Zhao Z, Singer J, Kern R, et al. A randomized trial of the effects of nebulized albuterol on pulmonary edema in brain-dead organ donors. Am J Transplant. 2014;14:621–8.PubMedPubMedCentralCrossRef Ware LB, Landeck M, Koyama T, Zhao Z, Singer J, Kern R, et al. A randomized trial of the effects of nebulized albuterol on pulmonary edema in brain-dead organ donors. Am J Transplant. 2014;14:621–8.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network, Matthay MA, Brower RG, Carson S, Douglas IS, Eisner M, 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;184:561–8.CrossRef National Heart, Lung, and Blood Institute Acute Respiratory Distress Syndrome (ARDS) Clinical Trials Network, Matthay MA, Brower RG, Carson S, Douglas IS, Eisner M, 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;184:561–8.CrossRef
26.
Zurück zum Zitat Blondonnet R, Audard J, Belville C, Clairefond G, Lutz J, Bouvier D, et al. RAGE inhibition reduces acute lung injury in mice. Sci Rep. 2017;7:7208.PubMedPubMedCentralCrossRef Blondonnet R, Audard J, Belville C, Clairefond G, Lutz J, Bouvier D, et al. RAGE inhibition reduces acute lung injury in mice. Sci Rep. 2017;7:7208.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Audard J, Godet T, Blondonnet R, Joffredo J-B, Paquette B, Belville C, et al. Inhibition of the receptor for advanced Glycation end-products in acute respiratory distress syndrome: a randomised laboratory trial in piglets. Sci Rep. 2019;9:9227.PubMedPubMedCentralCrossRef Audard J, Godet T, Blondonnet R, Joffredo J-B, Paquette B, Belville C, et al. Inhibition of the receptor for advanced Glycation end-products in acute respiratory distress syndrome: a randomised laboratory trial in piglets. Sci Rep. 2019;9:9227.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Camprubí-Rimblas M, Tantinyà N, Bringué J, Guillamat-Prats R, Artigas A. Anticoagulant therapy in acute respiratory distress syndrome. Ann Transl Med. 2018;6:36.PubMedPubMedCentralCrossRef Camprubí-Rimblas M, Tantinyà N, Bringué J, Guillamat-Prats R, Artigas A. Anticoagulant therapy in acute respiratory distress syndrome. Ann Transl Med. 2018;6:36.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Jabaudon M, Boucher P, Imhoff E, Chabanne R, Faure J-S, Roszyk L, et al. Sevoflurane for sedation in acute respiratory distress syndrome. A randomized controlled pilot study. Am J Respir Crit Care Med. 2017;195:792–800.PubMedCrossRef Jabaudon M, Boucher P, Imhoff E, Chabanne R, Faure J-S, Roszyk L, et al. Sevoflurane for sedation in acute respiratory distress syndrome. A randomized controlled pilot study. Am J Respir Crit Care Med. 2017;195:792–800.PubMedCrossRef
31.
Zurück zum Zitat McGowan J, Sampson M, Salzwedel DM, Cogo E, Foerster V, Lefebvre C. PRESS peer review of electronic search strategies: 2015 guideline statement. J Clin Epidemiol. 2016;75:40–6.PubMedCrossRef McGowan J, Sampson M, Salzwedel DM, Cogo E, Foerster V, Lefebvre C. PRESS peer review of electronic search strategies: 2015 guideline statement. J Clin Epidemiol. 2016;75:40–6.PubMedCrossRef
32.
Zurück zum Zitat Hooijmans CR, Tillema A, Leenaars M, Ritskes-Hoitinga M. Enhancing search efficiency by means of a search filter for finding all studies on animal experimentation in PubMed. Lab Anim. 2010;44:170–5.PubMedPubMedCentralCrossRef Hooijmans CR, Tillema A, Leenaars M, Ritskes-Hoitinga M. Enhancing search efficiency by means of a search filter for finding all studies on animal experimentation in PubMed. Lab Anim. 2010;44:170–5.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat de Vries RBM, Hooijmans CR, Tillema A, Leenaars M, Ritskes-Hoitinga M. A search filter for increasing the retrieval of animal studies in Embase. Lab Anim. 2011;45:268–70.PubMedPubMedCentralCrossRef de Vries RBM, Hooijmans CR, Tillema A, Leenaars M, Ritskes-Hoitinga M. A search filter for increasing the retrieval of animal studies in Embase. Lab Anim. 2011;45:268–70.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JPA, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. J Clin Epidemiol. 2009;62:e1–34.CrossRefPubMed Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JPA, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. J Clin Epidemiol. 2009;62:e1–34.CrossRefPubMed
35.
Zurück zum Zitat Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)--a metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377–81.PubMedCrossRef Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)--a metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377–81.PubMedCrossRef
36.
Zurück zum Zitat Higgins JPT, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011;343:d5928.PubMedPubMedCentralCrossRef Higgins JPT, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011;343:d5928.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Sterne JA, Hernán MA, Reeves BC, Savović J, Berkman ND, Viswanathan M, et al. ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ. 2016;355:i4919.PubMedPubMedCentralCrossRef Sterne JA, Hernán MA, Reeves BC, Savović J, Berkman ND, Viswanathan M, et al. ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ. 2016;355:i4919.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Henderson VC, Kimmelman J, Fergusson D, Grimshaw JM, Hackam DG. Threats to validity in the design and conduct of preclinical efficacy studies: a systematic review of guidelines for in vivo animal experiments. PLoS Med. 2013;10:e1001489.PubMedPubMedCentralCrossRef Henderson VC, Kimmelman J, Fergusson D, Grimshaw JM, Hackam DG. Threats to validity in the design and conduct of preclinical efficacy studies: a systematic review of guidelines for in vivo animal experiments. PLoS Med. 2013;10:e1001489.PubMedPubMedCentralCrossRef
39.
40.
Zurück zum Zitat Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol. 2010;8:e1000412.PubMedPubMedCentralCrossRef Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol. 2010;8:e1000412.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Muthen LK, Muthen B. Mplus Version 8 User’s Guide. Muthen & Muthen; 2017. Muthen LK, Muthen B. Mplus Version 8 User’s Guide. Muthen & Muthen; 2017.
43.
Zurück zum Zitat Friedrich JO, Adhikari NKJ, Beyene J. The ratio of means method as an alternative to mean differences for analyzing continuous outcome variables in meta-analysis: a simulation study. BMC Med Res Methodol. 2008;8:32.PubMedPubMedCentralCrossRef Friedrich JO, Adhikari NKJ, Beyene J. The ratio of means method as an alternative to mean differences for analyzing continuous outcome variables in meta-analysis: a simulation study. BMC Med Res Methodol. 2008;8:32.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Chinn S. A simple method for converting an odds ratio to effect size for use in meta-analysis. Stat Med. 2000;19:3127–31.PubMedCrossRef Chinn S. A simple method for converting an odds ratio to effect size for use in meta-analysis. Stat Med. 2000;19:3127–31.PubMedCrossRef
45.
Zurück zum Zitat Higgins JPT, Thompson SG. Quantifying heterogeneity in a meta-analysis. Stat Med. 2002;21:1539–58.PubMedCrossRef Higgins JPT, Thompson SG. Quantifying heterogeneity in a meta-analysis. Stat Med. 2002;21:1539–58.PubMedCrossRef
46.
Zurück zum Zitat Meyer NJ, Calfee CS. Novel translational approaches to the search for precision therapies for acute respiratory distress syndrome. Lancet Respir Med. 2017;5:512–23.PubMedPubMedCentralCrossRef Meyer NJ, Calfee CS. Novel translational approaches to the search for precision therapies for acute respiratory distress syndrome. Lancet Respir Med. 2017;5:512–23.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Jabaudon M, Blondonnet R, Pereira B, Cartin-Ceba R, Lichtenstern C, Mauri T, et al. Plasma sRAGE is independently associated with increased mortality in ARDS: a meta-analysis of individual patient data. Intensive Care Med. 2018;44:1388–99.PubMedPubMedCentralCrossRef Jabaudon M, Blondonnet R, Pereira B, Cartin-Ceba R, Lichtenstern C, Mauri T, et al. Plasma sRAGE is independently associated with increased mortality in ARDS: a meta-analysis of individual patient data. Intensive Care Med. 2018;44:1388–99.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Sinha P, Delucchi KL, Thompson BT, McAuley DF, Matthay MA, Calfee CS, et al. Latent class analysis of ARDS subphenotypes: a secondary analysis of the statins for acutely injured lungs from sepsis (SAILS) study. Intensive Care Med. 2018;44:1859–69.PubMedPubMedCentralCrossRef Sinha P, Delucchi KL, Thompson BT, McAuley DF, Matthay MA, Calfee CS, et al. Latent class analysis of ARDS subphenotypes: a secondary analysis of the statins for acutely injured lungs from sepsis (SAILS) study. Intensive Care Med. 2018;44:1859–69.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat 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.PubMedCrossRef 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.PubMedCrossRef
50.
Zurück zum Zitat Kitsios GD, Yang L, Manatakis DV, Nouraie M, Evankovich J, Bain W, et al. Host-response subphenotypes offer prognostic enrichment in patients with or at risk for acute respiratory distress syndrome. Crit Care Med. 2019;47:1724–34.PubMedCrossRefPubMedCentral Kitsios GD, Yang L, Manatakis DV, Nouraie M, Evankovich J, Bain W, et al. Host-response subphenotypes offer prognostic enrichment in patients with or at risk for acute respiratory distress syndrome. Crit Care Med. 2019;47:1724–34.PubMedCrossRefPubMedCentral
51.
Zurück zum Zitat Viswan A, Ghosh P, Gupta D, Azim A, Sinha N. Distinct metabolic Endotype mirroring acute respiratory distress syndrome (ARDS) subphenotype and its heterogeneous biology. Sci Rep. 2019;9:2108.PubMedPubMedCentralCrossRef Viswan A, Ghosh P, Gupta D, Azim A, Sinha N. Distinct metabolic Endotype mirroring acute respiratory distress syndrome (ARDS) subphenotype and its heterogeneous biology. Sci Rep. 2019;9:2108.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Sarma A, Calfee CS, Ware LB. Biomarkers and precision medicine: state of the art. Crit Care Clin. 2020;36:155–65.PubMedCrossRef Sarma A, Calfee CS, Ware LB. Biomarkers and precision medicine: state of the art. Crit Care Clin. 2020;36:155–65.PubMedCrossRef
53.
Zurück zum Zitat Seymour CW, Kerti SJ, Lewis AJ, Kennedy J, Brant E, Griepentrog JE, et al. Murine sepsis phenotypes and differential treatment effects in a randomized trial of prompt antibiotics and fluids. Crit Care. 2019;23:384.PubMedPubMedCentralCrossRef Seymour CW, Kerti SJ, Lewis AJ, Kennedy J, Brant E, Griepentrog JE, et al. Murine sepsis phenotypes and differential treatment effects in a randomized trial of prompt antibiotics and fluids. Crit Care. 2019;23:384.PubMedPubMedCentralCrossRef
Metadaten
Titel
Acute respiratory distress syndrome subphenotypes and therapy responsive traits among preclinical models: protocol for a systematic review and meta-analysis
verfasst von
Adrien Carla
Bruno Pereira
Hanifa Boukail
Jules Audard
Nathalie Pinol-Domenech
Manuela De Carvalho
Raiko Blondonnet
Ruoyang Zhai
Dominique Morand
Céline Lambert
Vincent Sapin
Lorraine B. Ware
Carolyn S. Calfee
Julie A. Bastarache
John G. Laffey
Nicole P. Juffermans
Lieuwe D. Bos
Antonio Artigas
Patricia R. M. Rocco
Michael A. Matthay
Daniel F. McAuley
Jean-Michel Constantin
Matthieu Jabaudon
for the ESICM Translational Biology Group of the Acute Respiratory Failure section
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2020
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-020-01337-9

Weitere Artikel der Ausgabe 1/2020

Respiratory Research 1/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.