1
|
Wang T, Yin H, Shen G, Cao Y, Qin X, Xu Q, Qi Y, Jiang X, Lu W. Effects of acetaminophen use on mortality of patients with acute respiratory distress syndrome: secondary data mining based on the MIMIC-IV database. BMC Pulm Med 2024; 24:568. [PMID: 39543557 PMCID: PMC11566145 DOI: 10.1186/s12890-024-03379-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Acetaminophen is a commonly used analgesic after surgery, and its impact on prognosis in patients with acute respiratory distress syndrome (ARDS) has not been studied. This study explores the association between the use of acetaminophen and the risk of mortality in patients with ARDS. METHODS In this retrospective cohort study, 3,227 patients with ARDS who had or had not received acetaminophen were obtained from the Medical Information Mart for Intensive Care IV, patients were divided into acetaminophen and non- acetaminophen groups. In-hospital mortality of ARDS patients was considered as primary end point. We used univariate and multivariate Cox regression analyses to assess the relationship of acetaminophen use and in-hospital mortality in patients with ARDS. Subgroup analysis was performed according to age, gender, and severity of ARDS. RESULTS Of the total patients, 2,438 individuals were identified as acetaminophen users. The median duration of follow-up was 10.54 (5.57, 18.82) days. The results showed that the acetaminophen use was associated with a decreased risk of in-hospital mortality [hazard ratio (HR) = 0.67, 95% confidence interval (CI): 0.57-0.78]. Across various subgroups of patients with ARDS based on age, gender, and severity, acetaminophen use exhibited an association with reduced risk of in-hospital mortality. CONCLUSION Acetaminophen use was associated with in-hospital mortality of patients with ARDS. Acetaminophen therapy may represent a promising therapeutic option for ARDS patients and warrants further investigation.
Collapse
Affiliation(s)
- Tong Wang
- Anhui Medical University, Hefei, Anhui Province, 230022, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province, 241000, China
| | - Hongzhen Yin
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province, 241000, China
| | - Guanggui Shen
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province, 241000, China
| | - Yingya Cao
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province, 241000, China
| | - Xuemei Qin
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province, 241000, China
| | - Qiancheng Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province, 241000, China
| | - Yupeng Qi
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province, 241000, China
| | - Xiaogan Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui Province, 241000, China
| | - Weihua Lu
- Anhui Medical University, Hefei, Anhui Province, 230022, China.
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Anhui Province Clinical Research Center for Critical Respiratory Medicine, No.2 West Road of Zheshan, Jinghu District, Wuhu, Anhui Province, 241000, China.
| |
Collapse
|
2
|
Kobayashi H, Amrein K, Mahmoud SH, Lasky-Su JA, Christopher KB. Metabolic phenotypes and vitamin D response in the critically ill: A metabolomic cohort study. Clin Nutr 2024; 43:10-19. [PMID: 39307095 DOI: 10.1016/j.clnu.2024.09.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/07/2024] [Accepted: 09/14/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND & AIMS Although vitamin D deficiency is common in critically ill patients, randomized controlled trials fail to demonstrate benefits of supplementation. We aimed to identify distinct vitamin D3 responsive metabolic phenotypes prior to trial intervention of high-dose vitamin D3 by applying machine learning clustering method to metabolomics data from the Correction of Vitamin D Deficiency in Critically Ill Patients (VITdAL-ICU) trial. METHODS In the randomized, placebo-controlled VITdAL-ICU trial, critically ill adults received placebo or high-dose vitamin D3. To distinguish vitamin D3 responsive metabolic phenotypes prior to intervention, we implemented consensus clustering with partitioning around medoids algorithm to the plasma metabolome data before randomization. Individual metabolite differences were determined utilizing linear mixed-effects regression models stratified for metabolomic phenotypes with false discovery rate adjustment. The association between vitamin D3 supplementation and 180-day mortality was evaluated in each metabolic phenotype, applying multivariable logistic regression analysis. RESULTS In 453 critically ill adults, the study identified 4 distinct metabolic phenotypes (clusters A. N = 134; B. N = 123; C. N = 92; D. N = 104). We found differential metabolic pathway patterns in the four clusters. Specifically, branched chain amino acid catabolic metabolites, long-chain acylcarnitines and diacylglycerol species are significantly increased in a specific metabolic phenotype (cluster D) following high-dose vitamin D3. Further, in cluster D high-dose vitamin D3 supplementation had a significantly lower adjusted odds of 180-day mortality after controlling age, sex, Simplified Acute Physiology Score II, admission diagnosis, and baseline 25-hydroxyvitamin D (OR 0.28 (95%CI, 0.09-0.89); P = 0.03). In metabotype A, B, and C, high-dose vitamin D3 supplementation was not significantly associated with lower 180-day mortality following multivariable adjustment. CONCLUSION In this post-hoc cohort study of the VITdAL-ICU trial, the clustering analysis of plasma metabolome data identified biologically distinct metabolic phenotypes. Among clusters, we found the different associations between high-dose vitamin D3 supplementation and specific metabolite pathways as well as 180-day mortality. Our findings facilitate further research to validate metabolic phenotype-targeted strategies for critical illness treatments.
Collapse
Affiliation(s)
- Hirotada Kobayashi
- Department of Critical Care Medicine, Sunnybrook Health Sciences Center, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, 204 Victoria Street, Toronto, ON M5B 1T8, Canada
| | - Karin Amrein
- Division of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, 8036 Graz, Austria
| | - Sherif H Mahmoud
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, 116 St. and 85 Ave, Edmonton, Alberta T6G 2R3, Canada
| | - Jessica A Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, USA
| | - Kenneth B Christopher
- Channing Division of Network Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, USA; Division of Renal Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, USA.
| |
Collapse
|
3
|
Zhang W, Wu L, Zhang S. Clinical phenotype of ARDS based on K-means cluster analysis: A study from the eICU database. Heliyon 2024; 10:e39198. [PMID: 39469677 PMCID: PMC11513467 DOI: 10.1016/j.heliyon.2024.e39198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
Purpose To explore the characteristics of the clinical phenotype of ARDS based on Machine Learning. Methods This is a study on Machine Learning. Screened cases of acute respiratory distress syndrome (ARDS) in the eICU database collected basic information in the cases and clinical data on the Day 1, Day 3, and Day 7 after the diagnosis of ARDS, respectively. Using the Calinski-Harabasz criterion, Gap Statistic, and Silhouette Coefficient, we determine the optimal clustering number k value. By the K-means cluster analysis to derive clinical phenotype, we analyzed the data collected within the first 24 h. We compared it with the survival of cases under the Berlin standard classification, and also examined the phenotypic conversion within the first 24 h, on day 3, and on day 7 after the diagnosis of ARDS. Results We collected 5054 cases and derived three clinical phenotypes using K-means cluster analysis. Phenotype-I is characterized by fewer abnormal laboratory indicators, higher oxygen partial pressure, oxygenation index, APACHE IV score, systolic and diastolic blood pressure, and lower respiratory rate and heart rate. Phenotype-II is characterized by elevated white blood cell count, blood glucose, creatinine, temperature, heart rate, and respiratory rate. Phenotype-III is characterized by elevated age, partial pressure of carbon dioxide, bicarbonate, GCS score, albumin. The differences in ICU length of stay and in-hospital mortality were significantly different between the three phenotypes (P < 0.05), with phenotype I having the lowest in-hospital mortality (10 %) and phenotype II having the highest (31.8 %). To compare the survival analysis of ARDS patients classified by phenotype and those classified according to Berlin criteria. The results showed that the differences in survival between phenotypes were statistically significant (P < 0.05) under phenotypic classification. Conclusions The clinical classification of ARDS based on K-means clustering analysis is beneficial for further identifying ARDS patients with different characteristics. Compared to the Berlin standard, the new clinical classification of ARDS provides a clearer display of the survival status of different types of patients, which helps to predict patient prognosis.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Critical Care Medicine, Kweichow Moutai Hospital, Renhuai City, Guizhou Province, 564500, China
- Department of Critical Care Medicine, People's Hospital of Leshan, Leshan City, Sichuan Province, 614008, China
| | - Linlin Wu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi City, Guizhou Province, 563000, China
| | - Shucheng Zhang
- Department of Dermatology and Venerology, Qian Foshan Hospital Affiliated to Shandong First Medical University, Jinan City, Shandong Province, 250013, China
| |
Collapse
|
4
|
Chen X, Harhay MO, Tong G, Li F. A BAYESIAN MACHINE LEARNING APPROACH FOR ESTIMATING HETEROGENEOUS SURVIVOR CAUSAL EFFECTS: APPLICATIONS TO A CRITICAL CARE TRIAL. Ann Appl Stat 2024; 18:350-374. [PMID: 38455841 PMCID: PMC10919396 DOI: 10.1214/23-aoas1792] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Assessing heterogeneity in the effects of treatments has become increasingly popular in the field of causal inference and carries important implications for clinical decision-making. While extensive literature exists for studying treatment effect heterogeneity when outcomes are fully observed, there has been limited development in tools for estimating heterogeneous causal effects when patient-centered outcomes are truncated by a terminal event, such as death. Due to mortality occurring during study follow-up, the outcomes of interest are unobservable, undefined, or not fully observed for many participants in which case principal stratification is an appealing framework to draw valid causal conclusions. Motivated by the Acute Respiratory Distress Syndrome Network (ARDSNetwork) ARDS respiratory management (ARMA) trial, we developed a flexible Bayesian machine learning approach to estimate the average causal effect and heterogeneous causal effects among the always-survivors stratum when clinical outcomes are subject to truncation. We adopted Bayesian additive regression trees (BART) to flexibly specify separate mean models for the potential outcomes and latent stratum membership. In the analysis of the ARMA trial, we found that the low tidal volume treatment had an overall benefit for participants sustaining acute lung injuries on the outcome of time to returning home but substantial heterogeneity in treatment effects among the always-survivors, driven most strongly by biologic sex and the alveolar-arterial oxygen gradient at baseline (a physiologic measure of lung function and degree of hypoxemia). These findings illustrate how the proposed methodology could guide the prognostic enrichment of future trials in the field.
Collapse
Affiliation(s)
- Xinyuan Chen
- Department of Mathematics and Statistics, Mississippi State University
| | - Michael O. Harhay
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania
| | - Guangyu Tong
- Department of Biostatistics, Yale School of Public Health
| | - Fan Li
- Department of Biostatistics, Yale School of Public Health
| |
Collapse
|
5
|
Fuentes-Claramonte P, Estradé A, Solanes A, Ramella-Cravaro V, Garcia-Leon MA, de Diego-Adeliño J, Molins C, Fung E, Valentí M, Anmella G, Pomarol-Clotet E, Oliver D, Vieta E, Radua J, Fusar-Poli P. Biomarkers for Psychosis: Are We There Yet? Umbrella Review of 1478 Biomarkers. SCHIZOPHRENIA BULLETIN OPEN 2024; 5:sgae018. [PMID: 39228676 PMCID: PMC11369642 DOI: 10.1093/schizbullopen/sgae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background and Hypothesis This umbrella review aims to comprehensively synthesize the evidence of association between peripheral, electrophysiological, neuroimaging, neuropathological, and other biomarkers and diagnosis of psychotic disorders. Study Design We selected systematic reviews and meta-analyses of observational studies on diagnostic biomarkers for psychotic disorders, published until February 1, 2018. Data extraction was conducted according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines. Evidence of association between biomarkers and psychotic disorders was classified as convincing, highly suggestive, suggestive, weak, or non-significant, using a standardized classification. Quality analyses used the Assessment of Multiple Systematic Reviews (AMSTAR) tool. Study Results The umbrella review included 110 meta-analyses or systematic reviews corresponding to 3892 individual studies, 1478 biomarkers, and 392 210 participants. No factor showed a convincing level of evidence. Highly suggestive evidence was observed for transglutaminase autoantibodies levels (odds ratio [OR] = 7.32; 95% CI: 3.36, 15.94), mismatch negativity in auditory event-related potentials (standardized mean difference [SMD] = 0.73; 95% CI: 0.5, 0.96), P300 component latency (SMD = -0.6; 95% CI: -0.83, -0.38), ventricle-brain ratio (SMD = 0.61; 95% CI: 0.5, 0.71), and minor physical anomalies (SMD = 0.99; 95% CI: 0.64, 1.34). Suggestive evidence was observed for folate, malondialdehyde, brain-derived neurotrophic factor, homocysteine, P50 sensory gating (P50 S2/S1 ratio), frontal N-acetyl-aspartate, and high-frequency heart rate variability. Among the remaining biomarkers, weak evidence was found for 626 and a non-significant association for 833 factors. Conclusions While several biomarkers present highly suggestive or suggestive evidence of association with psychotic disorders, methodological biases, and underpowered studies call for future higher-quality research.
Collapse
Affiliation(s)
- Paola Fuentes-Claramonte
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
- Biomedical Research Networking Centre Consortium on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain
| | - Andrés Estradé
- Department of Psychosis Studies, Early Psychosis: Interventions and Clinical-detection (EPIC) Lab, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, UK
| | - Aleix Solanes
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, Barcelona Autonomous University (UAB), Barcelona, Spain
| | - Valentina Ramella-Cravaro
- Department of Psychosis Studies, Early Psychosis: Interventions and Clinical-detection (EPIC) Lab, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, UK
| | - Maria Angeles Garcia-Leon
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
- Biomedical Research Networking Centre Consortium on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain
| | - Javier de Diego-Adeliño
- Biomedical Research Networking Centre Consortium on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain
- Department of Psychiatry and Forensic Medicine, Barcelona Autonomous University (UAB), Barcelona, Spain
- Sant Pau Mental Health Research Group, Institut de Recerca Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Conrad Molins
- Psychiatric Service, Hospital Universitari Santa Maria, Lleida, Catalonia, Spain
| | - Eric Fung
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
| | - Marc Valentí
- Biomedical Research Networking Centre Consortium on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Barcelona, Spain
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Gerard Anmella
- Biomedical Research Networking Centre Consortium on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Barcelona, Spain
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Edith Pomarol-Clotet
- FIDMAG Germanes Hospitalàries Research Foundation, Barcelona, Spain
- Biomedical Research Networking Centre Consortium on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain
| | - Dominic Oliver
- Department of Psychosis Studies, Early Psychosis: Interventions and Clinical-detection (EPIC) Lab, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, UK
- Department of Psychiatry, University of Oxford, Oxford OX3 7JX, UK
- NIHR Oxford Health Biomedical Research Centre, Oxford OX3 7JX, UK
- OPEN Early Detection Service, Oxford Health NHS Foundation Trust, Oxford OX3 7JX, UK
| | - Eduard Vieta
- Biomedical Research Networking Centre Consortium on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Barcelona, Spain
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Joaquim Radua
- Biomedical Research Networking Centre Consortium on Mental Health (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain
- Department of Psychosis Studies, Early Psychosis: Interventions and Clinical-detection (EPIC) Lab, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, UK
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona (UB), Barcelona, Spain
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet, Stockholm, Sweden
| | - Paolo Fusar-Poli
- Department of Psychosis Studies, Early Psychosis: Interventions and Clinical-detection (EPIC) Lab, Institute of Psychiatry Psychology and Neuroscience, King’s College London, London, UK
- OASIS Service, South London and the Maudsley NHS Foundation Trust, London, UK
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
6
|
Kargarpour Z, Cicko S, Köhler TC, Zech A, Stoshikj S, Bal C, Renner A, Idzko M, El-Gazzar A. Blocking P2Y2 purinergic receptor prevents the development of lipopolysaccharide-induced acute respiratory distress syndrome. Front Immunol 2023; 14:1310098. [PMID: 38179047 PMCID: PMC10765495 DOI: 10.3389/fimmu.2023.1310098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/06/2023] [Indexed: 01/06/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with high morbidity and mortality resulting from a direct or indirect injury of the lung. It is characterized by a rapid alveolar injury, lung inflammation with neutrophil accumulation, elevated permeability of the microvascular-barrier leading to an aggregation of protein-rich fluid in the lungs, followed by impaired oxygenation in the arteries and eventual respiratory failure. Very recently, we have shown an involvement of the Gq-coupled P2Y2 purinergic receptor (P2RY2) in allergic airway inflammation (AAI). In the current study, we aimed to elucidate the contribution of the P2RY2 in lipopolysaccharide (LPS)-induced ARDS mouse model. We found that the expression of P2ry2 in neutrophils, macrophages and lung tissue from animals with LPS-induced ARDS was strongly upregulated at mRNA level. In addition, ATP-neutralization by apyrase in vivo markedly attenuated inflammation and blocking of P2RY2 by non-selective antagonist suramin partially decreased inflammation. This was indicated by a reduction in the number of neutrophils, concentration of proinflammatory cytokines in the BALF, microvascular plasma leakage and reduced features of inflammation in histological analysis of the lung. P2RY2 blocking has also attenuated polymorphonuclear neutrophil (PMN) migration into the interstitium of the lungs in ARDS mouse model. Consistently, treatment of P2ry2 deficient mice with LPS lead to an amelioration of the inflammatory response showed by reduced number of neutrophils and concentrations of proinflammatory cytokines. In attempts to identify the cell type specific role of P2RY2, a series of experiments with conditional P2ry2 knockout animals were performed. We observed that P2ry2 expression in neutrophils, but not in the airway epithelial cells or CD4+ cells, was associated with the inflammatory features caused by ARDS. Altogether, our findings imply for the first time that increased endogenous ATP concentration via activation of P2RY2 is related to the pathogenesis of LPS-induced lung inflammation and may represent a potential therapeutic target for the treatment of ARDS and predictably assess new treatments in ARDS.
Collapse
Affiliation(s)
- Zahra Kargarpour
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Sanja Cicko
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
- Department of Pneumology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Thomas C. Köhler
- Department of Pneumology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Andreas Zech
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
- Department of Pneumology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Slagjana Stoshikj
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Christina Bal
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Andreas Renner
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Marco Idzko
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
- Department of Pneumology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Ahmed El-Gazzar
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
7
|
Iske J, Schroeter A, Knoedler S, Nazari-Shafti TZ, Wert L, Roesel MJ, Hennig F, Niehaus A, Kuehn C, Ius F, Falk V, Schmelzle M, Ruhparwar A, Haverich A, Knosalla C, Tullius SG, Vondran FWR, Wiegmann B. Pushing the boundaries of innovation: the potential of ex vivo organ perfusion from an interdisciplinary point of view. Front Cardiovasc Med 2023; 10:1272945. [PMID: 37900569 PMCID: PMC10602690 DOI: 10.3389/fcvm.2023.1272945] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/22/2023] [Indexed: 10/31/2023] Open
Abstract
Ex vivo machine perfusion (EVMP) is an emerging technique for preserving explanted solid organs with primary application in allogeneic organ transplantation. EVMP has been established as an alternative to the standard of care static-cold preservation, allowing for prolonged preservation and real-time monitoring of organ quality while reducing/preventing ischemia-reperfusion injury. Moreover, it has paved the way to involve expanded criteria donors, e.g., after circulatory death, thus expanding the donor organ pool. Ongoing improvements in EVMP protocols, especially expanding the duration of preservation, paved the way for its broader application, in particular for reconditioning and modification of diseased organs and tumor and infection therapies and regenerative approaches. Moreover, implementing EVMP for in vivo-like preclinical studies improving disease modeling raises significant interest, while providing an ideal interface for bioengineering and genetic manipulation. These approaches can be applied not only in an allogeneic and xenogeneic transplant setting but also in an autologous setting, where patients can be on temporary organ support while the diseased organs are treated ex vivo, followed by reimplantation of the cured organ. This review provides a comprehensive overview of the differences and similarities in abdominal (kidney and liver) and thoracic (lung and heart) EVMP, focusing on the organ-specific components and preservation techniques, specifically on the composition of perfusion solutions and their supplements and perfusion temperatures and flow conditions. Novel treatment opportunities beyond organ transplantation and limitations of abdominal and thoracic EVMP are delineated to identify complementary interdisciplinary approaches for the application and development of this technique.
Collapse
Affiliation(s)
- Jasper Iske
- Department of Cardiothoracic Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Schroeter
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
- Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Samuel Knoedler
- Division of Plastic Surgery, Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Timo Z. Nazari-Shafti
- Department of Cardiothoracic Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leonard Wert
- Department of Cardiothoracic Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Maximilian J. Roesel
- Department of Cardiothoracic Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Felix Hennig
- Department of Cardiothoracic Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Adelheid Niehaus
- Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Christian Kuehn
- Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Fabio Ius
- Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | - Volkmar Falk
- Department of Cardiothoracic Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
- Department of Health Science and Technology, Translational Cardiovascular Technology, ETH Zurich, Zürich, Switzerland
| | - Moritz Schmelzle
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Arjang Ruhparwar
- Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Axel Haverich
- Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Christoph Knosalla
- Department of Cardiothoracic Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Stefan G. Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Florian W. R. Vondran
- Department of General, Visceral and Transplant Surgery, Hannover Medical School, Hannover, Germany
| | - Bettina Wiegmann
- Department for Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| |
Collapse
|
8
|
Jeanmougin T, Cole E, Duceau B, Raux M, James A. Heterogeneity in defining multiple trauma: a systematic review of randomized controlled trials. Crit Care 2023; 27:363. [PMID: 37736733 PMCID: PMC10515068 DOI: 10.1186/s13054-023-04637-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023] Open
Abstract
INTRODUCTION While numerous randomized controlled trials (RCTs) have been conducted in the field of trauma, a substantial portion of them are yielding negative results. One potential contributing factor to this trend could be the lack of agreement regarding the chosen definitions across different trials. The primary objective was to identify the terminology and definitions utilized for the characterization of multiple trauma patients within randomized controlled trials (RCTs). METHODS A systematic review of the literature was performed in MEDLINE, EMBASE and clinicaltrials.gov between January 1, 2002, and July 31, 2022. RCTs or RTCs protocols were eligible if they included multiple trauma patients. The terms employed to characterize patient populations were identified, and the corresponding definitions for these terms were extracted. The subsequent impact on the population recruited was then documented to expose clinical heterogeneity. RESULTS Fifty RCTs were included, and 12 different terms identified. Among these terms, the most frequently used were "multiple trauma" (n = 21, 42%), "severe trauma" (n = 8, 16%), "major trauma" (n = 4, 8%), and trauma with hemorrhagic shock" (n = 4, 8%). Only 62% of RCTs (n = 31) provided a definition for the terms used, resulting a total of 21 different definitions. These definitions primarily relied on the injury severity score (ISS) (n = 15, 30%), displaying an important underlying heterogeneity. The choice of the terms had an impact on the study population, affecting both the ISS and in-hospital mortality. Eleven protocols were included, featuring five different terms, with "severe trauma" being the most frequent, occurring six times (55%). CONCLUSION This systematic review uncovers an important heterogeneity both in the terms and in the definitions employed to recruit trauma patients within RCTs. These findings underscore the imperative of promoting the use of a unique and consistent definition.
Collapse
Affiliation(s)
- Thomas Jeanmougin
- Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anesthesiology and critical care, Pitié-Salpêtrière Hospital, Paris, France
| | - Elaine Cole
- Centre of Trauma Sciences, Blizard Institute, Queen Mary University of London, London, UK
| | - Baptiste Duceau
- Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anesthesiology and critical care, Pitié-Salpêtrière Hospital, Paris, France
| | - Mathieu Raux
- Sorbonne Université, INSERM, UMRS1158 Neurophysiologie Respiratoire Expérimentale et Clinique; AP-HP, Groupe Hospitalier Universitaire APHP-Sorbonne Université, site Pitié-Salpêtrière, Département d'Anesthésie Réanimation, F-75013, Paris, France
| | - Arthur James
- Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anesthesiology and critical care, Pitié-Salpêtrière Hospital, Paris, France.
- Centre of Trauma Sciences, Blizard Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
9
|
Sikora A, Jeong H, Yu M, Chen X, Murray B, Kamaleswaran R. Cluster analysis driven by unsupervised latent feature learning of medications to identify novel pharmacophenotypes of critically ill patients. Sci Rep 2023; 13:15562. [PMID: 37730817 PMCID: PMC10511715 DOI: 10.1038/s41598-023-42657-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/13/2023] [Indexed: 09/22/2023] Open
Abstract
Unsupervised clustering of intensive care unit (ICU) medications may identify unique medication clusters (i.e., pharmacophenotypes) in critically ill adults. We performed an unsupervised analysis with Restricted Boltzmann Machine of 991 medications profiles of patients managed in the ICU to explore pharmacophenotypes that correlated with ICU complications (e.g., mechanical ventilation) and patient-centered outcomes (e.g., length of stay, mortality). Six unique pharmacophenotypes were observed, with unique medication profiles and clinically relevant differences in ICU complications and patient-centered outcomes. While pharmacophenotypes 2 and 4 had no statistically significant difference in ICU length of stay, duration of mechanical ventilation, or duration of vasopressor use, their mortality differed significantly (9.0% vs. 21.9%, p < 0.0001). Pharmacophenotype 4 had a mortality rate of 21.9%, compared with the rest of the pharmacophenotypes ranging from 2.5 to 9%. Phenotyping approaches have shown promise in classifying the heterogenous syndromes of critical illness to predict treatment response and guide clinical decision support systems but have never included comprehensive medication information. This first-ever machine learning approach revealed differences among empirically-derived subgroups of ICU patients that are not typically revealed by traditional classifiers. Identification of pharmacophenotypes may enable enhanced decision making to optimize treatment decisions.
Collapse
Affiliation(s)
- Andrea Sikora
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Augusta, GA, USA.
| | | | - Mengyun Yu
- Department of Statistics, University of Georgia Franklin College of Arts and Sciences, Athens, USA
| | - Xianyan Chen
- Department of Statistics, University of Georgia Franklin College of Arts and Sciences, Athens, USA
| | - Brian Murray
- Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, NC, USA
| | - Rishikesan Kamaleswaran
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
10
|
Zhang YX, Lan MJ, Liang SY, Han CM. [Advances on the application of physical airway clearance techniques in the treatment of inhalation injury]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2023; 39:475-480. [PMID: 37805758 DOI: 10.3760/cma.j.cn501225-20220608-00226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 10/09/2023]
Abstract
Although the treatment of patients with burns combined with inhalation injury has achieved great success, from the perspective of epidemiology, inhalation injury is still the most common cause of death in mass burns. Such patients often suffered burns of large total body surface area, which is difficult to treat, with airway management as one of the core links. Physical airway clearance technique (ACT) acts on a patient's respiratory system by physical means, to discharge secretions and foreign bodies in the airway, achieve airway clearance, and improve gas exchange. In addition, the technique can prevent or alleviate many complications, thereby improving the clinical outcome of patients with inhalation injury. This article reviews the application of physical ACT in the field of inhalation injury, and to provide decision-making basis for clinical medical staff to choose physical ACT corresponding to the patient's condition.
Collapse
Affiliation(s)
- Y X Zhang
- Department of Burns and Wound Repair, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - M J Lan
- Department of Nursing, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - S Y Liang
- Department of Nursing, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, China
| | - C M Han
- Department of Burns and Wound Repair, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
11
|
Sikora A, Rafiei A, Rad MG, Keats K, Smith SE, Devlin JW, Murphy DJ, Murray B, Kamaleswaran R, MRC-ICU Investigator Team. Pharmacophenotype identification of intensive care unit medications using unsupervised cluster analysis of the ICURx common data model. Crit Care 2023; 27:167. [PMID: 37131200 PMCID: PMC10155304 DOI: 10.1186/s13054-023-04437-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/10/2023] [Indexed: 05/04/2023] Open
Abstract
BACKGROUND Identifying patterns within ICU medication regimens may help artificial intelligence algorithms to better predict patient outcomes; however, machine learning methods incorporating medications require further development, including standardized terminology. The Common Data Model for Intensive Care Unit (ICU) Medications (CDM-ICURx) may provide important infrastructure to clinicians and researchers to support artificial intelligence analysis of medication-related outcomes and healthcare costs. Using an unsupervised cluster analysis approach in combination with this common data model, the objective of this evaluation was to identify novel patterns of medication clusters (termed 'pharmacophenotypes') correlated with ICU adverse events (e.g., fluid overload) and patient-centered outcomes (e.g., mortality). METHODS This was a retrospective, observational cohort study of 991 critically ill adults. To identify pharmacophenotypes, unsupervised machine learning analysis with automated feature learning using restricted Boltzmann machine and hierarchical clustering was performed on the medication administration records of each patient during the first 24 h of their ICU stay. Hierarchical agglomerative clustering was applied to identify unique patient clusters. Distributions of medications across pharmacophenotypes were described, and differences among patient clusters were compared using signed rank tests and Fisher's exact tests, as appropriate. RESULTS A total of 30,550 medication orders for the 991 patients were analyzed; five unique patient clusters and six unique pharmacophenotypes were identified. For patient outcomes, compared to patients in Clusters 1 and 3, patients in Cluster 5 had a significantly shorter duration of mechanical ventilation and ICU length of stay (p < 0.05); for medications, Cluster 5 had a higher distribution of Pharmacophenotype 1 and a smaller distribution of Pharmacophenotype 2, compared to Clusters 1 and 3. For outcomes, patients in Cluster 2, despite having the highest severity of illness and greatest medication regimen complexity, had the lowest overall mortality; for medications, Cluster 2 also had a comparably higher distribution of Pharmacophenotype 6. CONCLUSION The results of this evaluation suggest that patterns among patient clusters and medication regimens may be observed using empiric methods of unsupervised machine learning in combination with a common data model. These results have potential because while phenotyping approaches have been used to classify heterogenous syndromes in critical illness to better define treatment response, the entire medication administration record has not been incorporated in those analyses. Applying knowledge of these patterns at the bedside requires further algorithm development and clinical application but may have the future potential to be leveraged in guiding medication-related decision making to improve treatment outcomes.
Collapse
Affiliation(s)
- Andrea Sikora
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Augusta, GA USA
| | - Alireza Rafiei
- Department of Computer Science and Informatics, Emory University, Atlanta, GA USA
| | - Milad Ghiasi Rad
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA USA
| | - Kelli Keats
- Department of Pharmacy, Augusta University Medical Center, Augusta, GA USA
| | - Susan E. Smith
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Augusta, GA USA
| | - John W. Devlin
- Northeastern University School of Pharmacy, Boston, MA USA
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Care Medicine, Boston, MA USA
| | - David J. Murphy
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA USA
| | - Brian Murray
- Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, NC USA
| | - Rishikesan Kamaleswaran
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA USA
| | - MRC-ICU Investigator Team
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Augusta, GA USA
- Department of Computer Science and Informatics, Emory University, Atlanta, GA USA
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA USA
- Department of Pharmacy, Augusta University Medical Center, Augusta, GA USA
- Northeastern University School of Pharmacy, Boston, MA USA
- Brigham and Women’s Hospital, Division of Pulmonary and Critical Care Medicine, Boston, MA USA
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Emory University, Atlanta, GA USA
- Department of Pharmacy, University of North Carolina Medical Center, Chapel Hill, NC USA
- Department of Biomedical Informatics, Emory University School of Medicine, Atlanta, GA USA
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA USA
| |
Collapse
|
12
|
Blanchard F, James A, Assefi M, Kapandji N, Constantin JM. Personalized medicine targeting different ARDS phenotypes: The future of pharmacotherapy for ARDS? Expert Rev Respir Med 2023; 17:41-52. [PMID: 36724878 DOI: 10.1080/17476348.2023.2176302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
INTRODUCTION Acute respiratory distress syndrome (ARDS) still represents a major challenge with high mortality rates and altered quality of life. Many well-designed studies have failed to improve ARDS outcomes. Heterogeneity of etiologies, mechanisms of lung damage, different lung mechanics, and different treatment approaches may explain these failures. At the era of personalized medicine, ARDS phenotyping is not only a field of research, but a bedside consideration when implementing therapy. ARDS has moved from being a simple syndrome to a more complex area of subgrouping. Intensivists must understand these phenotypes and therapies associated with a better outcome. AREAS COVERED After a brief sum-up of the different type of ARDS phenotypes, we will present some relevant therapy that may be impacted by phenotyping. A focus on pharmacotherapy will be realized before a section on non-pharmaceutical strategies. Eventually, we will highlight the limits of our knowledge of phenotyping and the pitfalls of personalized medicine. EXPERT OPINION Biological and morphological ARDS phenotypes are now well studied. The future of ARDS therapy will go through phenotyping that allows a personalized medication for each patient. However, a better assessment of these phenotypes is required, and clinical trials should be conducted with an ad-hoc phenotyping before randomization.
Collapse
Affiliation(s)
- Florian Blanchard
- Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France.,Antimicrobial Stewardship Team GH Paris Centre, Cochin Hospital, APHP, Paris, France
| | - Arthur James
- Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France
| | - Mona Assefi
- Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France
| | - Natacha Kapandji
- Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France
| | - Jean-Michel Constantin
- Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
13
|
Wu L, Lei Q, Gao Z, Zhang W. Research Progress on Phenotypic Classification of Acute Respiratory Distress Syndrome: A Narrative Review. Int J Gen Med 2022; 15:8767-8774. [PMID: 36601648 PMCID: PMC9807128 DOI: 10.2147/ijgm.s391969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a clinical syndrome that is characterized by an acute onset and refractory hypoxemia. It remains an important contributor to high mortality in critically ill patients, and the majority of clinical randomized controlled trials on ARDS provide underwhelming findings, which is attributed in large part to its pathophysiological and clinical heterogeneity, among other aspects. It is now widely accepted that ARDS is highly heterogeneous, growing evidences support this. ARDS phenotypic and subphenotypic studies aim to further differentiate and identify ARDS heterogeneity in the hope that clinicians can benefit from it, then can diagnose ARDS faster and more accurately and provide targeted treatments. This review collates and evaluates the major phenotype-related research advances of recent years, with a specific focus on ARDS biomarkers and clinical factors.
Collapse
Affiliation(s)
- Linlin Wu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Qian Lei
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Zirong Gao
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China
| | - Wei Zhang
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People’s Republic of China,Correspondence: Wei Zhang, Email
| |
Collapse
|
14
|
Meng L, Liao X, Wang Y, Chen L, Gao W, Wang M, Dai H, Yan N, Gao Y, Wu X, Wang K, Liu Q. Pharmacologic therapies of ARDS: From natural herb to nanomedicine. Front Pharmacol 2022; 13:930593. [PMID: 36386221 PMCID: PMC9651133 DOI: 10.3389/fphar.2022.930593] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 10/03/2022] [Indexed: 12/15/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common critical illness in respiratory care units with a huge public health burden. Despite tremendous advances in the prevention and treatment of ARDS, it remains the main cause of intensive care unit (ICU) management, and the mortality rate of ARDS remains unacceptably high. The poor performance of ARDS is closely related to its heterogeneous clinical syndrome caused by complicated pathophysiology. Based on the different pathophysiology phases, drugs, protective mechanical ventilation, conservative fluid therapy, and other treatment have been developed to serve as the ARDS therapeutic methods. In recent years, there has been a rapid development in nanomedicine, in which nanoparticles as drug delivery vehicles have been extensively studied in the treatment of ARDS. This study provides an overview of pharmacologic therapies for ARDS, including conventional drugs, natural medicine therapy, and nanomedicine. Particularly, we discuss the unique mechanism and strength of nanomedicine which may provide great promises in treating ARDS in the future.
Collapse
Affiliation(s)
- Linlin Meng
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Department of Critical Care Medicine, Shanghai East Hospital, School of medicine, Tongji University, China
| | - Ximing Liao
- Department of Critical Care Medicine, Shanghai East Hospital, School of medicine, Tongji University, China
| | - Yuanyuan Wang
- Department of Critical Care Medicine, Shanghai East Hospital, School of medicine, Tongji University, China
| | - Liangzhi Chen
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wei Gao
- Department of Critical Care Medicine, Shanghai East Hospital, School of medicine, Tongji University, China
| | - Muyun Wang
- Department of Critical Care Medicine, Shanghai East Hospital, School of medicine, Tongji University, China
| | - Huiling Dai
- Department of Critical Care Medicine, Shanghai East Hospital, School of medicine, Tongji University, China
| | - Na Yan
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yixuan Gao
- Department of Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xu Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Kun Wang
- Department of Critical Care Medicine, Shanghai East Hospital, School of medicine, Tongji University, China
- *Correspondence: Kun Wang, ; Qinghua Liu,
| | - Qinghua Liu
- Department of Critical Care Medicine, Shanghai East Hospital, School of medicine, Tongji University, China
- *Correspondence: Kun Wang, ; Qinghua Liu,
| |
Collapse
|
15
|
Boerger E, Russ M, von Platen P, Taher M, Hinken L, Pomprapa A, Koebrich R, Konietschke F, Graw JA, Lachmann B, Braun W, Leonhardt S, Pickerodt PA, Francis RCE. Induction of severe hypoxemia and low lung recruitability for the evaluation of therapeutic ventilation strategies: a translational model of combined surfactant-depletion and ventilator-induced lung injury. Intensive Care Med Exp 2022; 10:32. [PMID: 35902450 PMCID: PMC9334469 DOI: 10.1186/s40635-022-00456-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/09/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Models of hypoxemic lung injury caused by lavage-induced pulmonary surfactant depletion are prone to prompt recovery of blood oxygenation following recruitment maneuvers and have limited translational validity. We hypothesized that addition of injurious ventilation following surfactant-depletion creates a model of the acute respiratory distress syndrome (ARDS) with persistently low recruitability and higher levels of titrated "best" positive end-expiratory pressure (PEEP) during protective ventilation. METHODS Two types of porcine lung injury were induced by lung lavage and 3 h of either protective or injurious ventilation, followed by 3 h of protective ventilation (N = 6 per group). Recruitment maneuvers (RM) and decremental PEEP trials comparing oxygenation versus dynamic compliance were performed after lavage and at 3 h intervals of ventilation. Pulmonary gas exchange function, respiratory mechanics, and ventilator-derived parameters were assessed after each RM to map the course of injury severity and recruitability. RESULTS Lung lavage impaired respiratory system compliance (Crs) and produced arterial oxygen tensions (PaO2) of 84±13 and 80±15 (FIO2 = 1.0) with prompt increase after RM to 270-395 mmHg in both groups. After subsequent 3 h of either protective or injurious ventilation, PaO2/FIO2 was 104±26 vs. 154±123 and increased to 369±132 vs. 167±87 mmHg in response to RM, respectively. After additional 3 h of protective ventilation, PaO2/FIO2 was 120±15 vs. 128±37 and increased to 470±68 vs. 185±129 mmHg in response to RM, respectively. Subsequently, decremental PEEP titration revealed that Crs peaked at 36 ± 10 vs. 25 ± 5 ml/cm H2O with PEEP of 12 vs. 16 cmH2O, and PaO2/FIO2 peaked at 563 ± 83 vs. 334 ± 148 mm Hg with PEEP of 16 vs. 22 cmH2O in the protective vs. injurious ventilation groups, respectively. The large disparity of recruitability between groups was not reflected in the Crs nor the magnitude of mechanical power present after injurious ventilation, once protective ventilation was resumed. CONCLUSION Addition of transitory injurious ventilation after lung lavage causes prolonged acute lung injury with diffuse alveolar damage and low recruitability yielding high titrated PEEP levels. Mimicking lung mechanical and functional characteristics of ARDS, this porcine model rectifies the constraints of single-hit lavage models and may enhance the translation of experimental research on mechanical ventilation strategies.
Collapse
Affiliation(s)
- Emilia Boerger
- Department of Anesthesiology and Intensive Care Medicine CCM/CVK, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13351, Berlin, Germany
| | - Martin Russ
- Department of Anesthesiology and Intensive Care Medicine CCM/CVK, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13351, Berlin, Germany
| | - Philip von Platen
- Chair for Medical Information Technology, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
| | - Mahdi Taher
- Department of Anesthesiology and Intensive Care Medicine CCM/CVK, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13351, Berlin, Germany
| | - Lea Hinken
- Fritz Stephan GmbH, Kirchstr. 19, 56412, Gackenbach, Germany
| | - Anake Pomprapa
- Chair for Medical Information Technology, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
| | - Rainer Koebrich
- EKU Elektronik GmbH, Am Sportplatz, 56291, Leiningen, Germany
| | - Frank Konietschke
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Jan Adriaan Graw
- Department of Anesthesiology and Intensive Care Medicine CCM/CVK, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13351, Berlin, Germany
| | - Burkhard Lachmann
- Department of Anesthesiology and Intensive Care Medicine CCM/CVK, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13351, Berlin, Germany
| | - Wolfgang Braun
- Fritz Stephan GmbH, Kirchstr. 19, 56412, Gackenbach, Germany
| | - Steffen Leonhardt
- Chair for Medical Information Technology, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, 52074, Aachen, Germany
| | - Philipp A Pickerodt
- Department of Anesthesiology and Intensive Care Medicine CCM/CVK, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13351, Berlin, Germany
| | - Roland C E Francis
- Department of Anesthesiology and Intensive Care Medicine CCM/CVK, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13351, Berlin, Germany.
| |
Collapse
|
16
|
Chiumello D, Pozzi T, Mereto E, Fratti I, Chiodaroli E, Gattinoni L, Coppola S. Long term feasibility of ultraprotective lung ventilation with low-flow extracorporeal carbon dioxide removal in ARDS patients. J Crit Care 2022; 71:154092. [PMID: 35714453 DOI: 10.1016/j.jcrc.2022.154092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/02/2022] [Accepted: 05/26/2022] [Indexed: 11/19/2022]
Abstract
PURPOSE To explore the feasibility of long-term application of ultraprotective ventilation with low flow ECCO2R support in moderate-severe ARDS patients and the reduction of mechanical power (MP) compared to lung protective ventilation. MATERIAL AND METHODS ARDS patients with PaO2/FiO2 < 200, PEEP of 10 cmH2O, tidal volume 6 ml/Kg of predicted body weight (PBW), plateau pressure > 24 cmH2O, MP > 17 J/min were prospectively enrolled. After 2 h tidal volume was reduced to 4-5 ml/kg, respiratory rate (RR) and PEEP were changed to maintain similar minute ventilation and mean airway pressure (MAP) to those obtained at baseline. After 2 h, ECCO2R support was started, RR was decreased and PEEP was increased to maintain similar PaCO2 and MAP, respectively. RESULTS The only reduction of tidal volume with the increase in RR did not decrease MP. The application of low flow ECCO2R support allowed a reduction of RR from 25 [24-30] to 11 [9-14] bpm and MP from 18 [13-23] to 8 [7-11] J/min. During the following 5 days no changes in mechanics variables and gas exchange occurred. CONCLUSIONS The application of low flow ECCO2R support with ultraprotective ventilation was feasible minimizing the MP without deterioration in oxygenation in ARDS patients.
Collapse
Affiliation(s)
- Davide Chiumello
- Department of Anesthesia and Intensive Care, ASST Santi Paolo e Carlo, San Paolo University Hospital, Via Di Rudini 9, Milan, Italy; Department of Health Sciences, University of Milan, Milano, Italy; Coordinated Research Center on Respiratory Failure, University of Milan, Milan, Italy.
| | - Tommaso Pozzi
- Department of Health Sciences, University of Milan, Milano, Italy
| | - Elisa Mereto
- Department of Health Sciences, University of Milan, Milano, Italy
| | - Isabella Fratti
- Department of Health Sciences, University of Milan, Milano, Italy
| | - Elena Chiodaroli
- Department of Anesthesia and Intensive Care, ASST Santi Paolo e Carlo, San Paolo University Hospital, Via Di Rudini 9, Milan, Italy
| | - Luciano Gattinoni
- Department of Anesthesiology, University Medical Center of Göttingen, Göttingen, Germany
| | - Silvia Coppola
- Department of Anesthesia and Intensive Care, ASST Santi Paolo e Carlo, San Paolo University Hospital, Via Di Rudini 9, Milan, Italy
| |
Collapse
|
17
|
Gong KQ, Mikacenic C, Long ME, Frevert CW, Birkland TP, Charron J, Gharib SA, Manicone AM. MAP2K2 Delays Recovery in Murine Models of Acute Lung Injury and Associates with Acute Respiratory Distress Syndrome Outcome. Am J Respir Cell Mol Biol 2022; 66:555-563. [PMID: 35157553 PMCID: PMC9116357 DOI: 10.1165/rcmb.2021-0252oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 12/30/2021] [Indexed: 12/15/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) remains a significant problem in need of new pharmaceutical approaches to improve its resolution. Studies comparing gene expression signatures in rodents and humans with lung injury reveal conserved pathways, including MAPK (mitogen-activated protein kinase)/ERK (extracellular signal-related protein kinase) activation. In preclinical acute lung injury (ALI) models, inhibition of MAP2K1 (MAPK kinase 1)/MAP2K2 (MAPK kinase 2) improves measures of ALI. Myeloid cell deletion of MAP2K1 results in sustained MAP2K2 activation and nonresolving ALI, suggesting that MAP2K2 deactivation may be a key driver of ALI resolution. We used human genomic data from the iSPAAR (Identification of SNPs Predisposing to Altered Acute Lung Injury Risk) Consortium to assess genetic variants in MAP2K1 and MAP2K2 for association with mortality from ARDS. To determine the role of MAP2K2 in ALI recovery, we studied mice deficient in Map2k2 (Mek2-/-) and wild-type control mice in ALI models. We identified a MAP2K2 variant that was associated with death in ARDS and MAP2K2 expression. In Pseudomonas aeruginosa ALI, Mek2-/- mice had similar early alveolar neutrophilic recruitment but faster resolution of alveolar neutrophilia and vascular leak. Gene expression analysis revealed a role for MAP2K2 in promoting and sustaining select proinflammatory pathway activation in ALI. Bone marrow chimera studies indicate that leukocyte MAP2K2 is the key regulator of ALI duration. These studies implicate a role for MAP2K2 in ALI duration via transcriptional regulation of inflammatory programming with potential relevance to ARDS. Targeting leukocyte MAP2K2 may be an effective strategy to promote ALI resolution.
Collapse
Affiliation(s)
- Ke-Qin Gong
- Division of Pulmonary, Critical Care and Sleep Medicine, and
| | - Carmen Mikacenic
- Division of Pulmonary, Critical Care and Sleep Medicine, and
- Benaroya Research Institute, Seattle, Washington
| | - Matthew E. Long
- Division of Pulmonary, Critical Care and Sleep Medicine, and
- Division of Pulmonary, Critical Care and Sleep Medicine, the Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Charles W. Frevert
- Division of Pulmonary, Critical Care and Sleep Medicine, and
- Department of Comparative Medicine, University of Washington, Seattle, Washington
| | | | - Jean Charron
- Oncology Division, Quebec University Hospital Center–Laval University Research Center, Laval University Research Center and Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec City, Quebec, Canada
| | - Sina A. Gharib
- Division of Pulmonary, Critical Care and Sleep Medicine, and
| | | |
Collapse
|
18
|
Wijeysundera DN, Mistry N, Mazer CD. The Future of Clinical Trials Methodology: Accomplishments and Challenges Ahead. Anesth Analg 2022; 134:664-667. [PMID: 35299204 DOI: 10.1213/ane.0000000000005935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Duminda N Wijeysundera
- From the Department of Anesthesia, St Michael's Hospital, Toronto, Ontario, Canada.,Department of Anesthesiology and Pain Medicine, Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Nikhil Mistry
- Department of Anesthesia, St Michael's Hospital, Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - C David Mazer
- From the Department of Anesthesia, St Michael's Hospital, Toronto, Ontario, Canada.,Departments of Anesthesiology and Pain Medicine, and Physiology, Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Belbasis L, Bellou V, Ioannidis JPA. Conducting umbrella reviews. BMJ MEDICINE 2022; 1:e000071. [PMID: 36936579 PMCID: PMC9951359 DOI: 10.1136/bmjmed-2021-000071] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 09/23/2022] [Indexed: 11/23/2022]
Abstract
In this article, Lazaros Belbasis and colleagues explain the rationale for umbrella reviews and the key steps involved in conducting an umbrella review, using a working example.
Collapse
Affiliation(s)
- Lazaros Belbasis
- Meta-Research Innovation Center Berlin, QUEST Center, Berlin Institute of Health, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Vanesa Bellou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - John P A Ioannidis
- Meta-Research Innovation Center Berlin, QUEST Center, Berlin Institute of Health, Charité Universitätsmedizin Berlin, Berlin, Germany
- Meta-Research Innovation Center at Stanford, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford University Medical School, Stanford, CA, USA
- Department of Epidemiology and Population Health, Stanford University Medical School, Stanford, CA, USA
- Department of Health Research and Policy, Stanford University Medical School, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University Medical School, Stanford, CA, USA
| |
Collapse
|
20
|
Villar J, González-Martín JM, Ambrós A, Mosteiro F, Martínez D, Fernández L, Soler JA, Parra L, Solano R, Soro M, Del Campo R, González-Luengo RI, Civantos B, Montiel R, Pita-García L, Vidal A, Añón JM, Ferrando C, Díaz-Domínguez FJ, Mora-Ordoñez JM, Fernández MM, Fernández C, Fernández RL, Rodríguez-Suárez P, Steyerberg EW, Kacmarek RM. Stratification for Identification of Prognostic Categories In the Acute RESpiratory Distress Syndrome (SPIRES) Score. Crit Care Med 2021; 49:e920-e930. [PMID: 34259448 DOI: 10.1097/ccm.0000000000005142] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To develop a scoring model for stratifying patients with acute respiratory distress syndrome into risk categories (Stratification for identification of Prognostic categories In the acute RESpiratory distress syndrome score) for early prediction of death in the ICU, independent of the underlying disease and cause of death. DESIGN A development and validation study using clinical data from four prospective, multicenter, observational cohorts. SETTING A network of multidisciplinary ICUs. PATIENTS One-thousand three-hundred one patients with moderate-to-severe acute respiratory distress syndrome managed with lung-protective ventilation. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS The study followed Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis guidelines for prediction models. We performed logistic regression analysis, bootstrapping, and internal-external validation of prediction models with variables collected within 24 hours of acute respiratory distress syndrome diagnosis in 1,000 patients for model development. Primary outcome was ICU death. The Stratification for identification of Prognostic categories In the acute RESpiratory distress syndrome score was based on patient's age, number of extrapulmonary organ failures, values of end-inspiratory plateau pressure, and ratio of Pao2 to Fio2 assessed at 24 hours of acute respiratory distress syndrome diagnosis. The pooled area under the receiver operating characteristic curve across internal-external validations was 0.860 (95% CI, 0.831-0.890). External validation in a new cohort of 301 acute respiratory distress syndrome patients confirmed the accuracy and robustness of the scoring model (area under the receiver operating characteristic curve = 0.870; 95% CI, 0.829-0.911). The Stratification for identification of Prognostic categories In the acute RESpiratory distress syndrome score stratified patients in three distinct prognostic classes and achieved better prediction of ICU death than ratio of Pao2 to Fio2 at acute respiratory distress syndrome onset or at 24 hours, Acute Physiology and Chronic Health Evaluation II score, or Sequential Organ Failure Assessment scale. CONCLUSIONS The Stratification for identification of Prognostic categories In the acute RESpiratory distress syndrome score represents a novel strategy for early stratification of acute respiratory distress syndrome patients into prognostic categories and for selecting patients for therapeutic trials.
Collapse
Affiliation(s)
- Jesús Villar
- CIBER de Enfermedades Respiratorias, Instituto Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | - Jesús M González-Martín
- Division of Biostatistics, Research Unit, Hospital Universitario Dr. Negrín, Las Palmas, Spain
| | - Alfonso Ambrós
- Intensive Care Unit, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Fernando Mosteiro
- Intensive Care Unit, Hospital Universitario A Coruña, La Coruña, Spain
| | - Domingo Martínez
- Intensive Care Unit, Hospital Universitario Virgen de Arrixaca, Murcia, Spain
| | - Lorena Fernández
- Intensive Care Unit, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Juan A Soler
- Intensive Care Unit, Hospital Universitario Virgen de Arrixaca, Murcia, Spain
| | - Laura Parra
- Intensive Care Unit, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Rosario Solano
- Intensive Care Unit, Hospital Virgen de la Luz, Cuenca, Spain
| | - Marina Soro
- Department of Anesthesiology, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Rafael Del Campo
- Intensive Care Unit, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | | | - Belén Civantos
- Intensive Care Unit, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Raquel Montiel
- Intensive Care Unit, Hospital Universitario NS de Candelaria, Santa Cruz de Tenerife, Spain
| | - Lidia Pita-García
- Intensive Care Unit, Hospital Universitario A Coruña, La Coruña, Spain
| | - Anxela Vidal
- Intensive Care Unit, Fundación Hospital Universitario Jiménez Díaz, Madrid, Spain
| | - José M Añón
- CIBER de Enfermedades Respiratorias, Instituto Salud Carlos III, Madrid, Spain
- Intensive Care Unit, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - Carlos Ferrando
- CIBER de Enfermedades Respiratorias, Instituto Salud Carlos III, Madrid, Spain
- Department of Anesthesiology and Critical Care, Hospital Clinic, Institut D'investigació August Pi i Sunyer, Barcelona, Spain
| | | | - Juan M Mora-Ordoñez
- Intensive Care Unit, Hospital Regional Universitario de Málaga Carlos Haya, Málaga, Spain
| | - M Mar Fernández
- Intensive Care Unit, Hospital Universitario Mutua Terrassa, Terrassa, Barcelona, Spain
| | - Cristina Fernández
- Research Unit, Hospital Universitario Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | - Rosa L Fernández
- CIBER de Enfermedades Respiratorias, Instituto Salud Carlos III, Madrid, Spain
- Research Unit, Hospital Universitario Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | - Pedro Rodríguez-Suárez
- Department of Thoracic Surgery, Hospital Universitario Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | - Ewout W Steyerberg
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
- Department of Public Health, Erasmus MC, Rotterdam, The Netherlands
| | - Robert M Kacmarek
- Department of Respiratory Care, Massachusetts General Hospital, Boston, MA
- Department of Anesthesiology, Harvard University, Boston, MA
| |
Collapse
|
21
|
Xu H, Xiao J. ACE2 Promotes the Synthesis of Pulmonary Surfactant to Improve AT II Cell Injury via SIRT1/eNOS Pathway. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2021; 2021:7710129. [PMID: 34471421 PMCID: PMC8405332 DOI: 10.1155/2021/7710129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/19/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE We aimed to explore the level of PS, cell viability, inflammatory factors, and apoptosis in neonatal respiratory distress syndrome (ARDS). Besides, we explored the potential relationship between ACE2, SIRT1/eNOS pathway, and hypoxia-induced AT II cell damage. METHODS The hUC-MSC-derived AT II cells were verified by IF and ICC, whereas qRT-PCR was used for PS and AT II cell marker (CK-8 and KGF). The AT II cell damage model was established by hypoxia exposure. The enhanced expression of ACE2 was tested after transfection with pcDNA3.1-ACE2 by western blot. The effects of hypoxia and ACE2 on AT II cells were evaluated by MTT, western blot, ELISA, and flow cytometry. The involvement of the SIRT1/eNOS pathway in AT II cell's protective functions against NRDS was verified with the addition of SIRT1 inhibitor EX527. RESULTS Based on the successful differentiation of AT II cells from hUC-MSCs and the buildup of AT II cell damage model, the overexpressed ACE2 impeded the hypoxia-induced cellular damage of AT II cells. It also counteracted the inhibitory effects of hypoxia on the secretion of PS. Overexpression of ACE2 rescued the cell viability and suppressed the secretion of inflammatory cytokines and the apoptosis of AT II cells triggered by hypoxia. And ACE2 activated the SIRT1/eNOS pathway to play its cell-protective and anti-inflammatory roles. CONCLUSION Our findings provided information that ACE2 prevented AT II cells from inflammatory damage through activating the SIRT1/eNOS pathway, which suggested that ACE2 might become a novel protective agent applied in the protection and treatment of NRDS.
Collapse
Affiliation(s)
- Hailing Xu
- Department of Respiratory Medicine, Laizhou People's Hospital of Shandong Province, China
| | - Jianguang Xiao
- Department of Thoracic Surgery, Laizhou People's Hospital of Shandong Province, China
| |
Collapse
|
22
|
Hamama KM, Fathy SM, AbdAlrahman RS, Alsherif SEDI, Ahmed SA. Driving pressure-guided ventilation versus protective lung ventilation in ARDS patients: A prospective randomized controlled study. EGYPTIAN JOURNAL OF ANAESTHESIA 2021. [DOI: 10.1080/11101849.2021.1930401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Khaled M. Hamama
- Anesthesiology, Surgical Intensive Care and Pain Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Sameh M. Fathy
- Anesthesiology, Surgical Intensive Care and Pain Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Reda S. AbdAlrahman
- Anesthesiology, Surgical Intensive Care and Pain Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Salah El-Din I. Alsherif
- Anesthesiology, Surgical Intensive Care and Pain Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Sameh Abdelkhalik Ahmed
- Anesthesiology, Surgical Intensive Care and Pain Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
23
|
Juschten J, Tuinman PR, Guo T, Juffermans NP, Schultz MJ, Loer SA, Girbes ARJ, de Grooth HJ. Between-trial heterogeneity in ARDS research. Intensive Care Med 2021; 47:422-434. [PMID: 33713156 PMCID: PMC7955690 DOI: 10.1007/s00134-021-06370-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/15/2021] [Indexed: 02/07/2023]
Abstract
Purpose Most randomized controlled trials (RCTs) in patients with acute respiratory distress syndrome (ARDS) revealed indeterminate or conflicting study results. We aimed to systematically evaluate between-trial heterogeneity in reporting standards and trial outcome. Methods A systematic review of RCTs published between 2000 and 2019 was performed including adult ARDS patients receiving lung-protective ventilation. A random-effects meta-regression model was applied to quantify heterogeneity (non-random variability) and to evaluate trial and patient characteristics as sources of heterogeneity. Results In total, 67 RCTs were included. The 28-day control-group mortality rate ranged from 10 to 67% with large non-random heterogeneity (I2 = 88%, p < 0.0001). Reported baseline patient characteristics explained some of the outcome heterogeneity, but only six trials (9%) reported all four independently predictive variables (mean age, mean lung injury score, mean plateau pressure and mean arterial pH). The 28-day control group mortality adjusted for patient characteristics (i.e. the residual heterogeneity) ranged from 18 to 45%. Trials with significant benefit in the primary outcome reported a higher control group mortality than trials with an indeterminate outcome or harm (mean 28-day control group mortality: 44% vs. 28%; p = 0.001). Conclusion Among ARDS RCTs in the lung-protective ventilation era, there was large variability in the description of baseline characteristics and significant unexplainable heterogeneity in 28-day control group mortality. These findings signify problems with the generalizability of ARDS research and underline the urgent need for standardized reporting of trial and baseline characteristics. Supplementary Information The online version of this article (10.1007/s00134-021-06370-w) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- J Juschten
- Department of Intensive Care, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, mail stop ZH 7D-172, 1081HV, Amsterdam, The Netherlands. .,Research VUmc Intensive Care (REVIVE), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands. .,Department of Anesthesiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| | - P R Tuinman
- Department of Intensive Care, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, mail stop ZH 7D-172, 1081HV, Amsterdam, The Netherlands.,Research VUmc Intensive Care (REVIVE), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - T Guo
- Department of Intensive Care, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, mail stop ZH 7D-172, 1081HV, Amsterdam, The Netherlands.,Research VUmc Intensive Care (REVIVE), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Division of System Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, The Netherlands
| | - N P Juffermans
- Laboratory of Experimental Intensive Care and Anesthesiology (LEICA), Amsterdam UMC, Universiteit Van Amsterdam, Amsterdam, The Netherlands.,Department of Intensive Care, OLVG Hospital, Amsterdam, The Netherlands
| | - M J Schultz
- Department of Intensive Care, Amsterdam UMC, Universiteit Van Amsterdam, Amsterdam, The Netherlands.,Mahidol-Oxford Tropical Medicine Research Unit (MORU), Mahidol University, Bangkok, Thailand.,Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - S A Loer
- Department of Anesthesiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - A R J Girbes
- Department of Intensive Care, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, mail stop ZH 7D-172, 1081HV, Amsterdam, The Netherlands.,Research VUmc Intensive Care (REVIVE), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - H J de Grooth
- Department of Anesthesiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
24
|
Wang X, Zhou X, Xia X, Zhang Y. Estradiol attenuates LPS-induced acute lung injury via induction of aquaporins AQP1 and AQP5. EUR J INFLAMM 2021; 19. [DOI: 10.1177/20587392211049197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024] Open
Abstract
Background Acute lung injury (ALI) is associated with increased inflammation and oxidative stress. Estradiol is produced by the ovaries and is the most active hormone of estrogen. Our aim was to investigate whether estradiol contributes to protect against lipopolysaccharide (LPS)-induced ALI via induction of aquaporins AQP1 and AQP5 and the underlying mechanisms. Methods and results For induction of ALI, LPS was applied once by intraperitoneal injection in SD rats 14 days after oophorectomy. To assess the therapeutic effects of estradiol on LPS-induced ALI, estradiol was subcutaneously injected for 1 h prior to LPS challenge. Estradiol can significantly attenuate the lung edema reflected by decreasing wet-to-dry weight ratio and permeability of lung and total protein concentration of bronchial lavage fluid (BALF). Results of histological detection showed that estradiol attenuated the lung injury reflected by reducing edema, congestion, and thickening pulmonary septal of lung tissues. In addition, estradiol attenuated TNF-α, IL-1β, and IL-6 and oxidative stress in lung tissues. Estradiol was more effective than estradiol associated with ERα antagonist or ERβ antagonist in protecting against LPS-induced ALI in rats. Mechanistically, we investigate whether estradiol regulates the expression of AQP1 and AQP5 in lung tissues. Of interest, estradiol upregulates AQP1 and AQP5 mRNA and protein expression. Taken together, these results demonstrate that estradiol can increase the expression of AQP1 and AQP5, which plays a critical role in ameliorating oxidative stress and downregulating inflammatory responses induced by LPS.Conclusion Therefore, these findings strongly suggest that AQP1 and AQP5 mediate the anti-inflammatory and antioxidant effects of estradiol.
Collapse
Affiliation(s)
- Xiaobo Wang
- Internal Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiuyun Zhou
- Blood Purification Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiumei Xia
- Department of Imaging Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Yili Zhang
- Health Management Center, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
25
|
Actualización de la Declaración de consenso en medicina critica para la atención multidisciplinaria del paciente con sospecha o confirmación diagnóstica de COVID-19. ACTA COLOMBIANA DE CUIDADO INTENSIVO 2020; 20:1-112. [PMCID: PMC7538086 DOI: 10.1016/j.acci.2020.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Antecedentes y objetivos La enfermedad por coronavirus de 2019 (COVID-19) es una enfermedad ocasionada por el nuevo coronavirus del síndrome respiratorio agudo grave (SARS-CoV-2). Se identificó por primera vez en diciembre de 2019 en la ciudad de Wuhan, en los meses siguientes se expandió rápidamente a todos los continentes y la Organización Mundial de la Salud (OMS) la reconoció como una pandemia global el 11 de marzo de 2020. La mayoría de los individuos son asintomáticos pero una baja proporción ingresan a cuidados intensivos con una alta morbimortalidad. Este consenso tiene como objetivo actualizar la declaratoria inicial emitida por la Asociación Colombiana de Medicina Crítica (AMCI) para el manejo del paciente críticamente enfermo con COVID-19, dentro de las áreas críticas de las instituciones de salud. Métodos Este estudio utilizó dos técnicas de consenso formal para construir las recomendaciones finales: Delphi modificada y grupos nominales. Se construyeron preguntas por la estrategia PICO. 10 grupos nominales desarrollaron recomendaciones para cada unidad temática. El producto del consenso fue evaluado y calificado en una ronda Delphi y se discutió de forma virtual por los relatores de cada núcleo y los representantes de sociedades médicas científicas afines al manejo del paciente con COVID-19. Resultados 80 expertos nacionales participaron en la actualización del consenso AMCI, especialistas en Medicina Critica y Cuidados Intensivos, Nefrología, Neurología, Neumología, bioeticistas, Medicina interna, Anestesia, Cirugía General, Cirugía de cabeza y cuello, Cuidados Paliativos, Enfermeras Especialistas en Medicina crítica, Terapeutas respiratorias especialistas en medicina crítica y Fisioterapia, con experiencia clínica en la atención del paciente críticamente enfermo. La declaratoria emite recomendaciones en los ámbitos más relevantes para la atención en salud de los casos de COVID-19, al interior de las unidades de cuidados intensivos, en el contexto nacional de Colombia. Conclusiones Un grupo significativo multidisciplinario de profesionales expertos en medicina crítica emiten, mediante técnicas de consenso formal, recomendaciones sobre la mejor práctica para la atención del paciente críticamente enfermo con COVID-19. Las recomendaciones deben ser adaptadas a las condiciones específicas, administrativas y estructurales de las distintas unidades de cuidados intensivos del país.
Collapse
|
26
|
Li B, Li D, Huang W, Che Y. Effect of lung recruitment on blood gas index, hemodynamics, lung compliance, and rehabilitation index in children with acute respiratory distress syndrome. Transl Pediatr 2020; 9:795-801. [PMID: 33457301 PMCID: PMC7804486 DOI: 10.21037/tp-20-383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a common pediatric disease, with an increasing mortality rate in recent years. This study aims to explore the effects of lung recruitment on blood gas indexes, hemodynamics, lung compliance, and rehabilitation index in children with ARDS. METHODS Seventy children with ARDS admitted to our hospital from December 2017 to December 2018 were selected as the study subjects, and were divided into a study group (35 cases, treated with lung recruitment strategy) and a control group (35 cases, treated with routine therapy). The changes of blood gas indexes, such as partial pressure of oxygen (PO2), partial pressure of carbon dioxide (PCO2), and partial pressure of oxygen/fraction of inspired oxygen (PO2/FiO2) levels, as well as hemodynamic indexes, including cardiac output (CO), heart rate (HR), mean arterial pressure (MAP), central venous pressure (CVP), were compared before and after treatment in the two groups. RESULTS Results showed that the difference in blood gas indexes between the two groups was statistically significant after treatment (P<0.05), and that the levels of PaO2, PaCO2, pondus Hydrogenii (pH), and PO2/FiO2 in the study group were all higher compared to the control group (P<0.05). The hemodynamic indexes showed that CO was significantly different between the two groups (P<0.05), but HR, MAP, and CVP were not (P>0.05). The lung compliance values of the two groups continued to increase at different time points after treatment (P<0.05), and the lung compliance of the study group was higher than that of the control group immediately after recruitment, as well as at 10 and 60 min of lung recruitment (P<0.05). In addition, the ventilator use, ICU stay, and hospital stay times of the study group were shorter than those in the control group (P<0.05), and the mortality rate of the study group was lower than that of the control group (P>0.05). CONCLUSIONS The lung recruitment strategy has a significant therapeutic effect on children with ARDS. It can effectively improve blood and gas function and lung compliance, and has a positive effect on the hemodynamic stability of children with ARDS.
Collapse
Affiliation(s)
- Bo Li
- Pediatric Intensive Care Unit of Maternity and Child Health Care of Zaozhuang, Zaozhuang, China
| | - Duoling Li
- Pediatric Intensive Care Unit of Tongxu People's Hospital, Kaifeng, China
| | - Wei Huang
- Pediatric Intensive Care Unit of Zaozhuang Municipal Hospital, Zaozhuang, China
| | - Yuanyuan Che
- Department of Coronary Heart Disease of Zaozhuang Municipal Hospital, Zaozhuang, China
| |
Collapse
|
27
|
Matera MG, Rogliani P, Bianco A, Cazzola M. Pharmacological management of adult patients with acute respiratory distress syndrome. Expert Opin Pharmacother 2020; 21:2169-2183. [PMID: 32783481 DOI: 10.1080/14656566.2020.1801636] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION There is still no definite drug for acute respiratory distress syndrome (ARDS) that is capable of reducing either short-term or long-term mortality. Therefore, great efforts are being made to identify a pharmacological approach that can be really effective. AREAS COVERED This review focuses on current challenges and future directions in the pharmacological management of ARDS, regardless of anti-infective treatments. The authors have excluded small randomized controlled trials (RCTs) with less than 60 patients because those studies do not have statistical power for outcome data, and also anecdotal trials but have considered the last meta-analysis on any drug. EXPERT OPINION There has been substantial progress in our knowledge of ARDS over the past two decades and many drugs have been used in its treatment. Nevertheless, effective targeted pharmacological treatments for ARDS are still lacking. The likely reason why a pharmacological approach is beneficial for some patients, but harmful for others is that ARDS is an extremely heterogeneous syndrome. To overcome this issue, a precision approach for ARDS, whereby therapies are specifically targeted to patients most likely to benefit, has been proposed. At present, however, the application of this approach seems to be a difficult task.
Collapse
Affiliation(s)
- Maria Gabriella Matera
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli" , Naples, Italy
| | - Paola Rogliani
- Department of Experimental Medicine, University of Rome "Tor Vergata" , Rome, Italy
| | - Andrea Bianco
- Department of Translational Medical Sciences, University of Campania "Luigi Vanvitelli"/Monaldi Hospital , Naples, Italy
| | - Mario Cazzola
- Department of Experimental Medicine, University of Rome "Tor Vergata" , Rome, Italy
| |
Collapse
|
28
|
Zhao X, Li Y, Ge Y, Shi Y, Lv P, Zhang J, Fu G, Zhou Y, Jiang K, Lin N, Bai T, Jin R, Wu Y, Yang X, Li X. Evaluation of Nutrition Risk and Its Association With Mortality Risk in Severely and Critically Ill COVID-19 Patients. JPEN J Parenter Enteral Nutr 2020; 45:32-42. [PMID: 32613660 PMCID: PMC7361906 DOI: 10.1002/jpen.1953] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/16/2020] [Accepted: 06/23/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND The nutrition status of coronavirus disease 2019 patients is unknown. This study evaluates clinical and nutrition characteristics of severely and critically ill patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and investigates the relationship between nutrition risk and clinical outcomes. METHODS A retrospective, observational study was conducted at West Campus of Union Hospital in Wuhan. Patients confirmed with SARS-CoV-2 infection by a nucleic acid-positive test and identified as severely or critically ill were enrolled in this study. Clinical data and outcomes information were collected and nutrition risk was assessed using Nutritional Risk Screening 2002 (NRS). RESULTS In total, 413 patients were enrolled in this study, including 346 severely and 67 critically ill patients. Most patients, especially critically ill patients, had significant changes in nutrition-related parameters and inflammatory markers. As for nutrition risk, the critically ill patients had significantly higher proportion of high NRS scores (P < .001), which were correlated with inflammatory and nutrition-related markers. Among 342 patients with NRS score ≥3, only 84 (of 342, 25%) received nutrition support. Critically ill patients and those with higher NRS score had a higher risk of mortality and longer stay in hospital. In logistic regression models, 1-unit increase in NRS score was associated with the risk of mortality increasing by 1.23 times (adjusted odds ratio, 2.23; 95% CI, 1.10-4.51; P = .026). CONCLUSIONS Most severely and critically ill patients infected with SARS-CoV-2 are at nutrition risk. The patients with higher nutrition risk have worse outcome and require nutrition therapy.
Collapse
Affiliation(s)
- Xiaobo Zhao
- Department of Paediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Yan Li
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Yanyan Ge
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Yuxin Shi
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Ping Lv
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Jianchu Zhang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Gui Fu
- Department of Paediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Yanfen Zhou
- Department of Paediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Ke Jiang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Nengxing Lin
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Tao Bai
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Runming Jin
- Department of Paediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Yuanjue Wu
- Department of Clinical Nutrition, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| | - Xuefeng Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xin Li
- Department of Paediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P. R. China
| |
Collapse
|
29
|
Rogers CJ, Harman RJ, Bunnell BA, Schreiber MA, Xiang C, Wang FS, Santidrian AF, Minev BR. Rationale for the clinical use of adipose-derived mesenchymal stem cells for COVID-19 patients. J Transl Med 2020; 18:203. [PMID: 32423449 PMCID: PMC7232924 DOI: 10.1186/s12967-020-02380-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 05/14/2020] [Indexed: 02/08/2023] Open
Abstract
In late 2019, a novel coronavirus (SARS-CoV-2) emerged in Wuhan, capital city of Hubei province in China. Cases of SARS-CoV-2 infection quickly grew by several thousand per day. Less than 100 days later, the World Health Organization declared that the rapidly spreading viral outbreak had become a global pandemic. Coronavirus disease 2019 (COVID-19) is typically associated with fever and respiratory symptoms. It often progresses to severe respiratory distress and multi-organ failure which carry a high mortality rate. Older patients or those with medical comorbidities are at greater risk for severe disease. Inflammation, pulmonary edema and an over-reactive immune response can lead to hypoxia, respiratory distress and lung damage. Mesenchymal stromal/stem cells (MSCs) possess potent and broad-ranging immunomodulatory activities. Multiple in vivo studies in animal models and ex vivo human lung models have demonstrated the MSC's impressive capacity to inhibit lung damage, reduce inflammation, dampen immune responses and aid with alveolar fluid clearance. Additionally, MSCs produce molecules that are antimicrobial and reduce pain. Upon administration by the intravenous route, the cells travel directly to the lungs where the majority are sequestered, a great benefit for the treatment of pulmonary disease. The in vivo safety of local and intravenous administration of MSCs has been demonstrated in multiple human clinical trials, including studies of acute respiratory distress syndrome (ARDS). Recently, the application of MSCs in the context of ongoing COVID-19 disease and other viral respiratory illnesses has demonstrated reduced patient mortality and, in some cases, improved long-term pulmonary function. Adipose-derived stem cells (ASC), an abundant type of MSC, are proposed as a therapeutic option for the treatment of COVID-19 in order to reduce morbidity and mortality. Additionally, when proven to be safe and effective, ASC treatments may reduce the demand on critical hospital resources. The ongoing COVID-19 outbreak has resulted in significant healthcare and socioeconomic burdens across the globe. There is a desperate need for safe and effective treatments. Cellular based therapies hold great promise for the treatment of COVID-19. This literature summary reviews the scientific rationale and need for clinical studies of adipose-derived stem cells and other types of mesenchymal stem cells in the treatment of patients who suffer with COVID-19.
Collapse
Affiliation(s)
| | | | - Bruce A. Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA USA
| | - Martin A. Schreiber
- Department of Surgery, Oregon Health and Science University, Portland, OR USA
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Fu-Sheng Wang
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center, Beijing, 100039 China
| | | | - Boris R. Minev
- Calidi Biotherapeutics, Inc., San Diego, CA USA
- Department of Radiation Medicine and Applied Sciences, Moores UCSD Cancer Center, San Diego, CA USA
| |
Collapse
|
30
|
Abstract
Although lung transplant remains the only option for patients suffering from end-stage lung failure, donor supply is insufficient to meet demand. Static cold preservation is the most common method to preserve lungs in transport to the recipient; however, this method does not improve lung quality and only allows for 8 h of storage. This results in lungs which become available for donation but cannot be used due to failure to meet physiologic criteria or an inability to store them for a sufficient time to find a suitable recipient. Therefore, lungs lost due to failure to meet physiological or compatibility criteria may be mitigated through preservation methods which improve lung function and storage durations. Ex situ lung perfusion (ESLP) is a recently developed method which allows for longer storage times and has been demonstrated to improve lung function such that rejected lungs can be accepted for donation. Although greater use of ESLP will help to improve donor lung utilization, the ability to cryopreserve lungs would allow for organ banking to better utilize donor lungs. However, lung cryopreservation research remains underrepresented in the literature despite its unique advantages for cryopreservation over other organs. Therefore, this review will discuss the current techniques for lung preservation, static cold preservation and ESLP, and provide a review of the cryopreservation challenges and advantages unique to lungs.
Collapse
|
31
|
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 2020; 200:42-50. [PMID: 30645145 DOI: 10.1164/rccm.201809-1808oc] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Rationale: Two biologic phenotypes of acute respiratory distress syndrome (ARDS) have been identified based on plasma protein markers in four previous studies. Objectives: To determine if blood leukocyte gene expression is different between the "reactive" and "uninflamed" phenotype. Methods: This is a new study adding blood leukocyte transcriptomics and bioinformatics analysis to an existing patient cohort of ARDS in patients with sepsis admitted to two ICUs during a 1.5-year period. Canonical pathway analysis was performed. Measurements and Main Results: A total of 210 patients with sepsis and ARDS were included, of whom 128 had a reactive and 82 an uninflamed phenotype. A total of 3,332/11,443 (29%) transcripts were significantly different between the phenotypes. Canonical pathway analysis showed upregulation of oxidative phosphorylation genes indicative of mitochondrial dysfunction (52% of genes in pathway). The uninflamed phenotype was characterized by upregulation of mitogen-activated protein kinase pathways. Conclusions: A third of genes are differentially expressed between biologic phenotypes of ARDS supporting the observation that the subgroups of ARDS are incomparable in terms of pathophysiology. These data provide additional support for biologic heterogeneity in patients with ARDS and suggests that a personalized approach to intervention focusing on oxidative phosphorylation is pivotal in this condition.
Collapse
Affiliation(s)
- Lieuwe D J Bos
- 1 Intensive Care, Laboratory of Experimental Intensive Care and Anesthesiology.,2 Department of Respiratory Medicine
| | - Brendon P Scicluna
- 3 Center of Experimental Molecular Medicine, and.,4 Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Infection and Immunity, Amsterdam University Medical Center, location Academic Medical Center, Amsterdam, the Netherlands
| | - David S Y Ong
- 5 Department of Microbiology, Julius Center for Health Sciences and Primary Care.,6 Department of Epidemiology, Julius Center for Health Sciences and Primary Care, and
| | - Olaf Cremer
- 7 Intensive Care, University Medical Center Utrecht, Utrecht, the Netherlands; and
| | | | - Marcus J Schultz
- 1 Intensive Care, Laboratory of Experimental Intensive Care and Anesthesiology.,8 Mahidol Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| |
Collapse
|
32
|
Contemporary strategies to improve clinical trial design for critical care research: insights from the First Critical Care Clinical Trialists Workshop. Intensive Care Med 2020; 46:930-942. [PMID: 32072303 PMCID: PMC7224097 DOI: 10.1007/s00134-020-05934-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/11/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Conducting research in critically-ill patient populations is challenging, and most randomized trials of critically-ill patients have not achieved pre-specified statistical thresholds to conclude that the intervention being investigated was beneficial. METHODS In 2019, a diverse group of patient representatives, regulators from the USA and European Union, federal grant managers, industry representatives, clinical trialists, epidemiologists, and clinicians convened the First Critical Care Clinical Trialists (3CT) Workshop to discuss challenges and opportunities in conducting and assessing critical care trials. Herein, we present the advantages and disadvantages of available methodologies for clinical trial design, conduct, and analysis, and a series of recommendations to potentially improve future trials in critical care. CONCLUSION The 3CT Workshop participants identified opportunities to improve critical care trials using strategies to optimize sample size calculations, account for patient and disease heterogeneity, increase the efficiency of trial conduct, maximize the use of trial data, and to refine and standardize the collection of patient-centered and patient-informed outcome measures beyond mortality.
Collapse
|
33
|
A Prognostic Enrichment Strategy for Selection of Patients With Acute Respiratory Distress Syndrome in Clinical Trials. Crit Care Med 2020; 47:377-385. [PMID: 30624279 DOI: 10.1097/ccm.0000000000003624] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Incomplete or ambiguous evidence for identifying high-risk patients with acute respiratory distress syndrome for enrollment into randomized controlled trials has come at the cost of an unreasonable number of negative trials. We examined a set of selected variables early in acute respiratory distress syndrome to determine accurate prognostic predictors for selecting high-risk patients for randomized controlled trials. DESIGN A training and testing study using a secondary analysis of data from four prospective, multicenter, observational studies. SETTING A network of multidisciplinary ICUs. PATIENTS We studied 1,200 patients with moderate-to-severe acute respiratory distress syndrome managed with lung-protective ventilation. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We evaluated different thresholds for patient's age, PaO2/FIO2, plateau pressure, and number of extrapulmonary organ failures to predict ICU outcome at 24 hours of acute respiratory distress syndrome diagnosis. We generated 1,000 random scenarios as training (n = 900, 75% of population) and testing (n = 300, 25% of population) datasets and averaged the logistic coefficients for each scenario. Thresholds for age (< 50, 50-70, > 70 yr), PaO2/FIO2 (≤ 100, 101-150, > 150 mm Hg), plateau pressure (< 29, 29-30, > 30 cm H2O), and number of extrapulmonary organ failure (< 2, 2, > 2) stratified accurately acute respiratory distress syndrome patients into categories of risk. The model that included all four variables proved best to identify patients with the highest or lowest risk of death (area under the receiver operating characteristic curve, 0.86; 95% CI, 0.84-0.88). Decision tree analyses confirmed the accuracy and robustness of this enrichment model. CONCLUSIONS Combined thresholds for patient's age, PaO2/FIO2, plateau pressure, and extrapulmonary organ failure provides prognostic enrichment accuracy for stratifying and selecting acute respiratory distress syndrome patients for randomized controlled trials.
Collapse
|
34
|
Distinctive Roles and Mechanisms of Human Neutrophil Peptides in Experimental Sepsis and Acute Respiratory Distress Syndrome. Crit Care Med 2019; 46:e921-e927. [PMID: 29979223 DOI: 10.1097/ccm.0000000000003265] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To examine the effects and mechanisms of human neutrophil peptides in systemic infection and noninfectious inflammatory lung injury. DESIGN Prospective experimental study. SETTING University hospital-based research laboratory. SUBJECTS In vitro human cells and in vivo mouse models. INTERVENTIONS Wild-type (Friend virus B-type) and conditional leukocyte human neutrophil peptides transgenic mice were subjected to either sepsis induced by cecal ligation and puncture or acute lung injury by intratracheal instillation of hydrochloric acid followed by mechanical ventilation. Using human neutrophil peptides as bait, the basal cell adhesion molecule (CD239) and the purinergic P2Y purinoceptor 6 receptor were identified as the putative human neutrophil peptides receptor complex in human lung epithelial cells. MEASUREMENTS AND MAIN RESULTS In the cecal ligation and puncture sepsis model, Friend virus B-type mice exhibited higher systemic bacterial load, cytokine production, and lung injury than human neutrophil peptides transgenic mice. Conversely, an increased lung cytokine production was seen in Friend virus B-type mice, which was further enhanced in human neutrophil peptides transgenic mice in response to two-hit lung injury induced by hydrochloric acid and mechanical ventilation. The human neutrophil peptides-mediated inflammatory response was mediated through the basal cell adhesion molecule-P2Y purinoceptor 6 receptor signal pathway in human lung epithelial cells. CONCLUSIONS Human neutrophil peptides are critical in host defense against infectious sepsis by their cationic antimicrobial properties but may exacerbate tissue injury when neutrophil-mediated inflammatory responses are excessive in noninfectious lung injury. Targeting the basal cell adhesion molecule/P2Y purinoceptor 6 signaling pathway may serve as a novel approach to attenuate the neutrophil-mediated inflammatory responses and injury while maintaining the antimicrobial function of human neutrophil peptides in critical illness.
Collapse
|
35
|
Is Overall Mortality the Right Composite Endpoint in Clinical Trials of Acute Respiratory Distress Syndrome? Crit Care Med 2019; 46:892-899. [PMID: 29420341 DOI: 10.1097/ccm.0000000000003022] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Overall mortality in patients with acute respiratory distress syndrome is a composite endpoint because it includes death from multiple causes. In most acute respiratory distress syndrome trials, it is unknown whether reported deaths are due to acute respiratory distress syndrome or the underlying disease, unrelated to the specific intervention tested. We investigated the causes of death after contracting acute respiratory distress syndrome in a large cohort. DESIGN A secondary analysis from three prospective, multicenter, observational studies. SETTING A network of multidisciplinary ICUs. PATIENTS We studied 778 patients with moderate-to-severe acute respiratory distress syndrome treated with lung-protective ventilation. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We examined death in the ICU from individual causes. Overall ICU mortality was 38.8% (95% CI, 35.4-42.3). Causes of acute respiratory distress syndrome modified the risk of death. Twenty-three percent of deaths occurred from refractory hypoxemia due to nonresolving acute respiratory distress syndrome. Most patients died from causes unrelated to acute respiratory distress syndrome: 48.7% of nonsurvivors died from multisystem organ failure, and cancer or brain injury was involved in 37.1% of deaths. When quantifying the true burden of acute respiratory distress syndrome outcome, we identified 506 patients (65.0%) with one or more exclusion criteria for enrollment into current interventional trials. Overall ICU mortality of the "trial cohort" (21.3%) was markedly lower than the parent cohort (relative risk, 0.55; 95% CI, 0.43-0.70; p < 0.000001). CONCLUSIONS Most deaths in acute respiratory distress syndrome patients are not directly related to lung damage but to extrapulmonary multisystem organ failure. It would be challenging to prove that specific lung-directed therapies have an effect on overall survival.
Collapse
|
36
|
Mesenchymal Stem Cells From Bone Marrow, Adipose Tissue, and Lung Tissue Differentially Mitigate Lung and Distal Organ Damage in Experimental Acute Respiratory Distress Syndrome. Crit Care Med 2019; 46:e132-e140. [PMID: 29116998 DOI: 10.1097/ccm.0000000000002833] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Mesenchymal stem cells-based therapies have shown promising effects in experimental acute respiratory distress syndrome. Different mesenchymal stem cells sources may result in diverse effects in respiratory diseases; however, there is no information regarding the best source of mesenchymal stem cells to treat pulmonary acute respiratory distress syndrome. We tested the hypothesis that mesenchymal stem cells derived from bone marrow, adipose tissue, and lung tissue would lead to different beneficial effects on lung and distal organ damage in experimental pulmonary acute respiratory distress syndrome. DESIGN Animal study and primary cell culture. SETTING Laboratory investigation. SUBJECTS Seventy-five Wistar rats. INTERVENTIONS Wistar rats received saline (control) or Escherichia coli lipopolysaccharide (acute respiratory distress syndrome) intratracheally. On day 2, acute respiratory distress syndrome animals were further randomized to receive saline or bone marrow, adipose tissue, or lung tissue mesenchymal stem cells (1 × 10 cells) IV. Lung mechanics, histology, and protein levels of inflammatory mediators and growth factors were analyzed 5 days after mesenchymal stem cells administration. RAW 264.7 cells (a macrophage cell line) were incubated with lipopolysaccharide followed by coculture or not with bone marrow, adipose tissue, and lung tissue mesenchymal stem cells (10 cells/mL medium). MEASUREMENTS AND MAIN RESULTS Regardless of mesenchymal stem cells source, cells administration improved lung function and reduced alveolar collapse, tissue cellularity, collagen, and elastic fiber content in lung tissue, as well as decreased apoptotic cell counts in liver. Bone marrow and adipose tissue mesenchymal stem cells administration also reduced levels of tumor necrosis factor-α, interleukin-1β, keratinocyte-derived chemokine, transforming growth factor-β, and vascular endothelial growth factor, as well as apoptotic cell counts in lung and kidney, while increasing expression of keratinocyte growth factor in lung tissue. Additionally, mesenchymal stem cells differently modulated the secretion of biomarkers by macrophages depending on their source. CONCLUSIONS Mesenchymal stem cells from different sources led to variable responses in lungs and distal organs. Bone marrow and adipose tissue mesenchymal stem cells yielded greater beneficial effects than lung tissue mesenchymal stem cells. These findings may be regarded as promising in clinical trials.
Collapse
|
37
|
Kubat Ö, Gökçek E, Kaydu A. An Analysis of Patients Followed Up in the Intensive Care Unit with the Diagnosis of Acute Respiratory Distress Syndrome. Turk J Anaesthesiol Reanim 2019; 47:62-68. [PMID: 31276113 DOI: 10.5152/tjar.2018.27122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 07/03/2018] [Indexed: 01/11/2023] Open
Abstract
Objective To examine the factors thought to have an effect on the mortality of patients with acute respiratory distress syndrome (ARDS) in the intensive care unit (ICU). Methods A retrospective evaluation of 100 patients diagnosed with ARDS in the ICU between January 2009 and January 2013 was made. Surviving and deceased patients were compared with respect to the effect of the general characteristics, aetiological and prognostic factors, mechanical ventilation (MV) applications (especially permissive hypercapnia resulting from the restriction of the tidal volume predicted to avoid excessive distention of the alveoli), laboratory test values, multiorgan dysfunction rates, Acute Physiologic Assessment and Chronic Health Evaluation II score, Lung Injury Score, Glasgow Coma Score, Sequential Organ Failure Assessment scores, arterial blood gas parameters and partial pressure of arterial oxygen/fraction of inspired oxygen ratio values on mortality. Results There were 100 patients with ARDS comprising 61 males and 39 females with a mean age of 57.0±13.0 (range: 20-82) years and length of stay in the ICU of 38.7±13 days. The aetiological causes of ARDS were determined as pneumonia in 37 patients, trauma (traffic accidents inside or outside the vehicle and other accidents) in 14, sepsis in 19, pulmonary contusion in 9, non-pulmonary infection in 6, intoxication in 5, multiple blood transfusions in 4, firearms injury in 4 and acute pancreatitis in 2. Forty-four patients died. Conclusion Survival rates were increased in patients with ARDS with early diagnosis and ICU support, lung protective MV strategy and permissive hypercapnia.
Collapse
Affiliation(s)
- Ömer Kubat
- Department of Anaesthesiology and Reanimation, Diyarbakır State Hospital, Diyarbakır, Turkey
| | - Erhan Gökçek
- Department of Anaesthesiology and Reanimation, Diyarbakır State Hospital, Diyarbakır, Turkey
| | - Ayhan Kaydu
- Department of Anaesthesiology and Reanimation, Diyarbakır State Hospital, Diyarbakır, Turkey
| |
Collapse
|
38
|
Cardinal-Fernandez P, Ortiz G, Chang CH, Kao KC, Bertreau E, Philipponnet C, Casero-Alonso VM, Souweine B, Charbonney E, Guérin C. Predicting the Impact of Diffuse Alveolar Damage through Open Lung Biopsy in Acute Respiratory Distress Syndrome-The PREDATOR Study. J Clin Med 2019; 8:jcm8060829. [PMID: 31212621 PMCID: PMC6616523 DOI: 10.3390/jcm8060829] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/24/2019] [Accepted: 05/31/2019] [Indexed: 12/16/2022] Open
Abstract
The aim of this retrospective and international study is to identify those clinical variables associated with diffuse alveolar damage (DAD), and to explore the impact of DAD on hospital mortality risk. Inclusion criteria were: adult patients with acute respiratory distress syndrome (ARDS) undergoing open lung biopsy (OLB) during their intensive care unit (ICU) management. The main end-points were: DAD and hospital mortality. In the training (n = 193) and validation cohorts (n = 65), the respiratory rate (odd ratio (OR) 0.956; confidence interval (CI) 95% 0.918; 0.995) and coronary ischemia (OR 5.974; CI95% 1.668; 21.399) on the day of ARDS had an average area under the receiver operating characteristic curve (AUROC) of 0.660 (CI95% 0.585; 0.736) and 0.562 (0.417; 0.706), respectively. PEEP (OR 1.131; CI95% 1.051; 1.218) and coronary ischemia (OR 6.820; CI95% 1.856; 25.061) on the day of OLB had an average AUROC of 0.696 (CI95% 0.621; 0.769) and 0.534 (CI95% 0.391; 0.678), respectively, to predict DAD. DAD (OR 2.296; CI95% 1.228; 4.294), diabetes mellitus requiring insulin (OR 0.081; CI95% 0.009; 0.710) and the respiratory rate (OR 1.045; CI95% 1.001; 1.091) on the day of ARDS had an average AUROC of 0.659 (CI95% 0.583; 0.737) and 0.513 (CI95% 0.361; 0.664) to predict hospital mortality and DAD (OR 2.081; CI95% 1.053; 4.114), diabetes mellitus requiring insulin (OR 0.093; CI95% 0.009; 0.956), PaCO2 (OR 1.051; CI95% 1.019; 1.084), and platelets count (OR 0.999; CI95% 0.999; 0.999) the day of OLB had an average AUROC of 0.778 (CI95% 0.710; 0.843) and 0.634 (CI95%0.481; 0.787) to predict hospital mortalty in the training and validation cohorts, respectively. In conclusion, DAD could not to be predicted clinically and was significantly associated with hospital mortality.
Collapse
Affiliation(s)
| | - Guillermo Ortiz
- Universidad del Bosque, Bogotá #131a2, Colombia.
- Universidad de Barcelona, 08007 Barcelona, Spain.
| | | | - Kuo-Chin Kao
- Chang Gung Memorial Hospital, Taoyuan 33005, Taiwan.
| | | | | | | | | | - Emmanuel Charbonney
- Centre de Recherche Hôpital du Sacré-Cœur de Montréal (HSCM), 5400 boul. Gouin Ouest, Montréal, QC H4J 1C5, Canada.
| | - Claude Guérin
- Réanimation Médicale Hôpital de la Croix Rousse, 69004 Lyon, France.
- Lyon East Faculty of Medicine, University of Lyon, 69100 Lyon, France.
- INSERM, 955 Créteil, France.
- Hospices Civils de Lyon, 69003 Lyon, France.
| |
Collapse
|
39
|
Griffiths MJD, McAuley DF, Perkins GD, Barrett N, Blackwood B, Boyle A, Chee N, Connolly B, Dark P, Finney S, Salam A, Silversides J, Tarmey N, Wise MP, Baudouin SV. Guidelines on the management of acute respiratory distress syndrome. BMJ Open Respir Res 2019; 6:e000420. [PMID: 31258917 PMCID: PMC6561387 DOI: 10.1136/bmjresp-2019-000420] [Citation(s) in RCA: 285] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/01/2019] [Indexed: 12/16/2022] Open
Abstract
The Faculty of Intensive Care Medicine and Intensive Care Society Guideline Development Group have used GRADE methodology to make the following recommendations for the management of adult patients with acute respiratory distress syndrome (ARDS). The British Thoracic Society supports the recommendations in this guideline. Where mechanical ventilation is required, the use of low tidal volumes (<6 ml/kg ideal body weight) and airway pressures (plateau pressure <30 cmH2O) was recommended. For patients with moderate/severe ARDS (PF ratio<20 kPa), prone positioning was recommended for at least 12 hours per day. By contrast, high frequency oscillation was not recommended and it was suggested that inhaled nitric oxide is not used. The use of a conservative fluid management strategy was suggested for all patients, whereas mechanical ventilation with high positive end-expiratory pressure and the use of the neuromuscular blocking agent cisatracurium for 48 hours was suggested for patients with ARDS with ratio of arterial oxygen partial pressure to fractional inspired oxygen (PF) ratios less than or equal to 27 and 20 kPa, respectively. Extracorporeal membrane oxygenation was suggested as an adjunct to protective mechanical ventilation for patients with very severe ARDS. In the absence of adequate evidence, research recommendations were made for the use of corticosteroids and extracorporeal carbon dioxide removal.
Collapse
Affiliation(s)
| | - Danny Francis McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, UK
| | - Gavin D Perkins
- Warwick Clinical Trials Unit, University of Warwick, Coventry, West Midlands, UK
| | | | - Bronagh Blackwood
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, UK
| | - Andrew Boyle
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, UK
| | - Nigel Chee
- Academic Department of Critical Care, Queen Alexandra Hospital, Portsmouth Hospitals NHS Trust, Portsmouth, UK
| | | | - Paul Dark
- Division of Infection, Immunity and Respiratory Medicine, NIHR Biomedical Research Centre, University of Manchester, Manchester, Greater Manchester, UK
| | - Simon Finney
- Peri-Operative Medicine, Barts Health NHS Trust, London, UK
| | - Aemun Salam
- Peri-Operative Medicine, Barts Health NHS Trust, London, UK
| | - Jonathan Silversides
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, UK
| | - Nick Tarmey
- Academic Department of Critical Care, Queen Alexandra Hospital, Portsmouth Hospitals NHS Trust, Portsmouth, UK
| | | | - Simon V Baudouin
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
40
|
Heras MJL, Dianti J, Tisminetzky M, Svetliza G, Giannasi SE, Roman ES. Cryoprobe biopsy for the diagnosis of acute hypoxemic respiratory failure of undetermined origin. J Intensive Care Soc 2019; 21:119-123. [PMID: 32489407 DOI: 10.1177/1751143719847323] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Rationale Acute hypoxemic respiratory failure is a condition that comprises a wide array of entities. Obtaining a histological lung sample might help reach a diagnosis and direct an appropriate treatment in a select group of patients. Objective To describe our experience in the use of cryobiopsy for the diagnosis of acute hypoxemic respiratory failure of undetermined origin. Methods Retrospective analysis of case series of patients with acute hypoxemic respiratory failure who underwent lung cryobiopsy at the Intensive Care Unit of the Hospital Italiano de Buenos Aires, Argentina. Results Cryobiopsy yielded a histological diagnosis in all patients (n = 10, 100%). This led to either a change in therapy or continuation of a specific treatment in eight of these patients. Cryobiopsy was found to be contributive in all the patients who did not meet Berlin criteria for acute respiratory distress syndrome. No major complications were associated with the procedure. Conclusions Cryobiopsy is a safe procedure with a high diagnostic yield in a selected group of patients.
Collapse
Affiliation(s)
- Marcos J Las Heras
- Servicio de Terapia Intensiva de Adultos, Hospital Italiano de Buenos Aires, Peron, Argentina.,Servicio de Clínica Médica, Sección de Neumonología, Hospital Italiano de Buenos Aires, Peron, Argentina
| | - Jose Dianti
- Servicio de Terapia Intensiva de Adultos, Hospital Italiano de Buenos Aires, Peron, Argentina
| | - Manuel Tisminetzky
- Servicio de Terapia Intensiva de Adultos, Hospital Italiano de Buenos Aires, Peron, Argentina
| | - Graciela Svetliza
- Servicio de Clínica Médica, Sección de Neumonología, Hospital Italiano de Buenos Aires, Peron, Argentina
| | - Sergio E Giannasi
- Servicio de Terapia Intensiva de Adultos, Hospital Italiano de Buenos Aires, Peron, Argentina
| | - Eduardo San Roman
- Servicio de Terapia Intensiva de Adultos, Hospital Italiano de Buenos Aires, Peron, Argentina
| |
Collapse
|
41
|
Kumada Y, Takahashi T, Shimizu H, Nakamura R, Omori E, Inoue K, Morimatsu H. Therapeutic effect of carbon monoxide-releasing molecule-3 on acute lung injury after hemorrhagic shock and resuscitation. Exp Ther Med 2019; 17:3429-3440. [PMID: 30988722 PMCID: PMC6447800 DOI: 10.3892/etm.2019.7390] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/11/2019] [Indexed: 01/14/2023] Open
Abstract
Hemorrhagic shock and resuscitation (HSR) induces a pulmonary inflammatory response and frequently causes acute lung injury. Carbon monoxide-releasing molecule-3 (CORM-3) has been reported to liberate and deliver CO under physiological conditions, which exerts organ-protective effects during systemic insults. The present study aimed to determine whether the administration of CORM-3 following HSR exerts a therapeutic effect against HSR-induced lung injury without any detrimental effects on oxygenation and hemodynamics. To induce hemorrhagic shock, rats were bled to a mean arterial blood pressure of 30 mmHg for 45 min and then resuscitated with the shed blood. CORM-3 or a vehicle was intravenously administered immediately following the completion of resuscitation. The rats were divided into four groups, including sham, HSR, HSR/CORM-3 and HSR/inactive CORM-3 groups. Arterial blood gas parameters and vital signs were recorded during HSR. The histopathological changes to the lungs were evaluated using a lung injury score, while pulmonary edema was evaluated on the basis of the protein concentration in bronchoalveolar lavage fluid and the lung wet/dry ratio. We also investigated the pulmonary expression levels of inflammatory mediators and apoptotic markers such as cleaved caspase-3 and transferase-mediated dUTP-fluorescein isothiocyanate nick-end labeling (TUNEL) staining. Although HSR caused significant lung histopathological damage and pulmonary edema, CORM-3 significantly ameliorated this damage. CORM-3 also attenuated the HSR-induced upregulation of tumor necrosis factor-α, inducible nitric oxide synthase and interleukin-1β genes, and the expression of interleukin-1β and macrophage inflammatory protein-2. In addition, the expression of interleukin-10, an anti-inflammatory cytokine, was inversely enhanced by CORM-3, which also reduced the number of TUNEL-positive cells and the expression of cleaved caspase-3 following HSR. Although CORM-3 was administered during the acute phase of HSR, it did not exert any influence on arterial blood gas analysis data and vital signs during HSR. Therefore, treatment with CORM-3 ameliorated HSR-induced lung injury, at least partially, through anti-inflammatory and anti-apoptotic effects, without any detrimental effects on oxygenation and hemodynamics.
Collapse
Affiliation(s)
- Yuta Kumada
- Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Toru Takahashi
- Faculty of Health and Welfare Science, Okayama Prefectural University, Soja, Okayama 719-1197, Japan
| | - Hiroko Shimizu
- Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Ryu Nakamura
- Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Emiko Omori
- Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Kazuyoshi Inoue
- Department of Anesthesiology, Kagawa Prefectural Central Hospital, Takamatsu, Kagawa 760-8557, Japan
| | - Hiroshi Morimatsu
- Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
42
|
Kogelmann K, Scheller M, Drüner M, Jarczak D. Use of hemoadsorption in sepsis-associated ECMO-dependent severe ARDS: A case series. J Intensive Care Soc 2019; 21:183-190. [PMID: 32489416 DOI: 10.1177/1751143718818992] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Introduction Acute respiratory distress syndrome in the context of severe sepsis and septic shock represents a serious clinical disorder. A recent case series in patients with septic shock and renal failure receiving hemoadsorption treatment showed rapid hemodynamic stabilization and increased survival, particularly in pneumonia patients and in those where therapy was started early. We hypothesized that patients suffering from pneumonia and refractory acute respiratory distress syndrome to the extent that they required extracorporeal membrane oxygenation support could possibly demonstrate the most pronounced benefit from the treatment. Methods We assessed the association of hemoadsorption treatment with hemodynamics, ventilation, and outcome variables in a set of patients with septic shock, acute respiratory distress syndrome, need for veno-venous extracorporeal membrane oxygenation, and continuous renal replacement therapy. Results Key observations include a significant stabilization in hemodynamics as evidenced by a marked decrease in catecholamine need, which was paralleled by a clear reduction in hyperlactatemia. Respiratory variables improved significantly. In addition, severity of illness and overall organ dysfunction showed a considerable decrease during the course of treatment. Observed mortality was approximately half as predicted by APACHE II. Treatment with CytoSorb was safe and well tolerated with no device-related adverse events. Discussion This is the first case series reporting on outcome variables associated to CytoSorb therapy in critically ill patients with septic shock, acute respiratory distress syndrome, veno-venous extracorporeal membrane oxygenation, and continuous renal replacement therapy. Based on our observations in this small case series, CytoSorb might represent a potentially promising therapy option for patients with refractory extracorporeal membrane oxygenation-dependent acute respiratory distress syndrome in the context of septic shock.
Collapse
Affiliation(s)
- Klaus Kogelmann
- Department of Anesthesiology and Intensive Care Medicine, Klinikum Emden, Emden, Germany
| | - Morten Scheller
- Department of Anesthesiology and Intensive Care Medicine, Klinikum Emden, Emden, Germany
| | - Matthias Drüner
- Department of Anesthesiology and Intensive Care Medicine, Klinikum Emden, Emden, Germany
| | - Dominik Jarczak
- Department of Intensive Care Medicine, University Hospital Hamburg, Hamburg, Germany
| |
Collapse
|
43
|
Weathington NM, Álvarez D, Sembrat J, Radder J, Cárdenes N, Noda K, Gong Q, Wong H, Kolls J, D'Cunha J, Mallampalli RK, Chen BB, Rojas M. Ex vivo lung perfusion as a human platform for preclinical small molecule testing. JCI Insight 2018; 3:95515. [PMID: 30282819 DOI: 10.1172/jci.insight.95515] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/17/2018] [Indexed: 12/14/2022] Open
Abstract
The acute respiratory distress syndrome (ARDS) causes an estimated 70,000 US deaths annually. Multiple pharmacologic interventions for ARDS have been tested and failed. An unmet need is a suitable laboratory human model to predictively assess emerging therapeutics on organ function in ARDS. We previously demonstrated that the small molecule BC1215 blocks actions of a proinflammatory E3 ligase-associated protein, FBXO3, to suppress NF-κB signaling in animal models of lung injury. Ex vivo lung perfusion (EVLP) is a clinical technique that maintains lung function for possible transplant after organ donation. We used human lungs unacceptable for transplant to model endotoxemic injury with EVLP for 6 hours. LPS infusion induced inflammatory injury with impaired oxygenation of pulmonary venous circulation. BC1215 treatment after LPS rescued oxygenation and decreased inflammatory cytokines in bronchoalveolar lavage. RNA sequencing transcriptomics from biopsies taken during EVLP revealed robust inflammatory gene induction by LPS with a strong signal for NF-κB-associated transcripts. BC1215 treatment reduced the LPS induction of genes associated with inflammatory and host defense gene responses by Gene Ontology (GOterm) and pathways analysis. BC1215 also significantly antagonized LPS-mediated NF-κB activity. EVLP may provide a unique human platform for preclinical study of chemical entities such as FBXO3 inhibitors on tissue physiology.
Collapse
Affiliation(s)
| | - Diana Álvarez
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine.,Simmons Center for Interstitial Lung Disease, and
| | - John Sembrat
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine.,Simmons Center for Interstitial Lung Disease, and
| | - Josiah Radder
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine
| | - Nayra Cárdenes
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine.,Simmons Center for Interstitial Lung Disease, and
| | - Kentaro Noda
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Qiaoke Gong
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine
| | - Hesper Wong
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine
| | - Jay Kolls
- Department of Medicine, Tulane University, New Orleans, Louisiana
| | - Jonathan D'Cunha
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Rama K Mallampalli
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,The Veterans Affairs Pittsburgh Health System, Pittsburgh, Pennsylvania, USA
| | - Bill B Chen
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine.,Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mauricio Rojas
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine.,Simmons Center for Interstitial Lung Disease, and.,The University of Pittsburgh McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
44
|
Xie J, Liu L, Yang Y, Yu W, Li M, Yu K, Zheng R, Yan J, Wang X, Cai G, Li J, Gu Q, Zhao H, Mu X, Ma X, Qiu H. A modified acute respiratory distress syndrome prediction score: a multicenter cohort study in China. J Thorac Dis 2018; 10:5764-5773. [PMID: 30505484 DOI: 10.21037/jtd.2018.09.117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Background Early recognition of the risks of acute respiratory distress syndrome (ARDS) and prevention of the development of ARDS may be more effective in improving patient outcomes. We performed the present study to determine the ARDS risk factors in a Chinese population and validate a score to predict the development of ARDS. Methods This was an observational multicenter cohort study performed in 13 tertiary hospitals in China. Patients admitted into participating intensive care units (ICUs) from January 1 to January 31, 2012, and from January 1 to January 10, 2013, were enrolled in a retrospective derivation cohort and a prospective validation cohort, respectively. In the derivation cohort, the potential risk factors of ARDS were collected. The confirmed risk factors were determined with univariate and multivariate logistic regression analyses, and then the modified ARDS prediction score (MAPS) was established. We prospectively enrolled patients to verify the accuracy of MAPS. Results A total of 479 and 198 patients were enrolled into the retrospective derivation cohort and the prospective validation cohort, respectively. A total of 93 (19.4%) patients developed ARDS in the derivation cohort. Acute pancreatitis, pneumonia, hypoalbuminemia, acidosis, and high respiratory rate were the risk factors for ARDS. The MAPS discriminated patients who developed ARDS from those who did not, with an area under the curve (AUC) of 0.809 [95% confidence interval (CI), 0.758-0.859, P<0.001]. In the prospective validation cohort, performance of the MAPS was similar to the retrospective derivation cohort, with an AUC of 0.792 (95% CI, 0.717-0.867, P<0.001). The lung injury prediction score (LIPS) showed a predicted value of an AUC of 0.770 (95% CI, 0.728-0.812, P<0.001) in our patients, which was significantly lower than our score (P<0.046). Conclusions The MAPS based on risk factors could help the clinician to predict patients who will develop ARDS. Trial registration ClinicalTrials.gov NCT01666834.
Collapse
Affiliation(s)
- Jianfeng Xie
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Ling Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yi Yang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | - Wenkui Yu
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Maoqin Li
- Department of Critical Care Medicine, Xuzhou Central Hospital, Xuzhou 221009, China
| | - Kaijiang Yu
- Department of Critical Care Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin 150040, China.,Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ruiqiang Zheng
- Department of Critical Care Medicine, Subei People's Hospital, School of Medicine, Yangzhou University, Yangzhou 225001, China
| | - Jie Yan
- Department of Critical Care Medicine, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Xue Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Guolong Cai
- Department of Critical Care Medicine, Zhejiang Hospital, Hangzhou 310013, China
| | - Jianguo Li
- Department of Critical Care Medicine, Zhongnan Hospital, Wuhan University, 430071, Wuhan, China
| | - Qin Gu
- Department of Critical Care Medicine, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Hongsheng Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Xinwei Mu
- Department of Critical Care Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xiaochun Ma
- Department of Critical Care Medicine, The First Hospital of China Medical University, Shenyang 110001, China
| | - Haibo Qiu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
45
|
Negative trials in critical care: why most research is probably wrong. THE LANCET RESPIRATORY MEDICINE 2018; 6:659-660. [DOI: 10.1016/s2213-2600(18)30279-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022]
|
46
|
Horie S, Gonzalez HE, Laffey JG, Masterson CH. Cell therapy in acute respiratory distress syndrome. J Thorac Dis 2018; 10:5607-5620. [PMID: 30416812 DOI: 10.21037/jtd.2018.08.28] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute respiratory distress syndrome (ARDS) is driven by a severe pro-inflammatory response resulting in lung damage, impaired gas exchange and severe respiratory failure. There is no specific treatment that effectively improves outcome in ARDS. However, in recent years, cell therapy has shown great promise in preclinical ARDS studies. A wide range of cells have been identified as potential candidates for use, among these are mesenchymal stromal cells (MSCs), which are adult multi-lineage cells that can modulate the immune response and enhance repair of damaged tissue. The therapeutic potential of MSC therapy for sepsis and ARDS has been demonstrated in multiple in vivo models. The therapeutic effect of these cells seems to be due to two different mechanisms; direct cellular interaction, and paracrine release of different soluble products such as extracellular vesicles (EVs)/exosomes. Different approaches have also been studied to enhance the therapeutic effect of these cells, such as the over-expression of anti-inflammatory or pro-reparative molecules. Several clinical trials (phase I and II) have already shown safety of MSCs in ARDS and other diseases. However, several translational issues still need to be addressed, such as the large-scale production of cells, and their potentiality and variability, before the therapeutic potential of stem cells therapies can be realized.
Collapse
Affiliation(s)
- Shahd Horie
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - Hector Esteban Gonzalez
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| | - John G Laffey
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland.,Department of Anesthesia and Intensive Care Medicine, Galway University Hospitals, SAOLTA Hospital Group, Ireland
| | - Claire H Masterson
- Regenerative Medicine Institute (REMEDI), CÚRAM Centre for Research in Medical Devices, Biomedical Sciences Building, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
47
|
Janiaud P, Cristea IA, Ioannidis JPA. Industry-funded versus non-profit-funded critical care research: a meta-epidemiological overview. Intensive Care Med 2018; 44:1613-1627. [PMID: 30151688 PMCID: PMC6182357 DOI: 10.1007/s00134-018-5325-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 07/16/2018] [Indexed: 12/24/2022]
Abstract
Purpose To study the landscape of funding in intensive care research and assess whether the reported outcomes of industry-funded randomized controlled trials (RCTs) are more favorable. Methods We systematically assembled meta-analyses evaluating any type of intervention in the critical care setting and reporting the source of funding for each included RCT. Furthermore, when the intervention was a drug or biologic, we searched also the original RCT articles, when their funding information was unavailable in the meta-analysis. We then qualitatively summarized the sources of funding. For binary outcomes, separate summary odds ratios were calculated for trials with and without industry funding. We then calculated the ratio of odds ratios (RORs) and the summary ROR (sROR) across topics. ROR < 1 implies that the experimental intervention is relatively more favorable in trials with industry funding compared with trials without industry funding. For RCTs included in the ROR analysis, we also examined the conclusions of their abstract. Results Across 67 topics with 568 RCTs, 88 were funded by industry and another 73 had both industry and non-profit funding. Across 33 topics with binary outcomes, the sROR was 1.10 [95% CI (0.96–1.26), I2 = 1%]. Conclusions were not significantly more commonly unfavorable for the experimental arm interventions in industry-funded trials (21.3%) compared with trials without industry funding (18.2%). Conclusion Industry-funded RCTs are the minority in intensive care. We found no evidence that industry-funded trials in intensive care yield more favorable results or are less likely to reach unfavorable conclusions. Electronic supplementary material The online version of this article (10.1007/s00134-018-5325-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Perrine Janiaud
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, 94305, USA
| | - Ioana-Alinea Cristea
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, 94305, USA.,Department of Clinical Psychology and Psychotherapy, Babes-Bolyai University, Cluj-Napoca, Romania
| | - John P A Ioannidis
- Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, 94305, USA. .,Department of Clinical Psychology and Psychotherapy, Babes-Bolyai University, Cluj-Napoca, Romania. .,Department of Medicine, Stanford University, Stanford, CA, 94305, USA. .,Department of Health Research and Policy, Stanford University, Stanford, CA, 94305, USA. .,Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA. .,Department of Statistics, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
48
|
Lin S, Wu H, Wang C, Xiao Z, Xu F. Regulatory T Cells and Acute Lung Injury: Cytokines, Uncontrolled Inflammation, and Therapeutic Implications. Front Immunol 2018; 9:1545. [PMID: 30038616 PMCID: PMC6046379 DOI: 10.3389/fimmu.2018.01545] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/21/2018] [Indexed: 12/24/2022] Open
Abstract
Acute respiratory distress syndrome/acute lung injury (ALI) was described in 1967. The uncontrolled inflammation is a central issue of the syndrome. The regulatory T cells (Tregs), formerly known as suppressor T cells, are a subpopulation of T cells. Tregs indirectly limits immune inflammation-inflicted tissue damage by employing multiple mechanisms and creating the appropriate immune environment for successful tissue repair. And it plays a central role in the resolution of ALI. Accordingly, for this review, we will focus on Treg populations which are critical for inflammatory immunity of ALI, and the effect of interaction between Treg subsets and cytokines on ALI. And then explore the possibility of cytokines as beneficial factors in inflammation resolution of ALI.
Collapse
Affiliation(s)
- Shihui Lin
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Wu
- Center for Cognitive and Neurobiological Imaging, Stanford University, Stanford, CA, United States
| | - Chuanjiang Wang
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhibo Xiao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Xu
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
49
|
Ortiz G, Garay M, Capelozzi V, Cardinal-Fernández P. Airway Pathological Alterations Selectively Associated With Acute Respiratory Distress Syndrome and Diffuse Alveolar Damage - Narrative Review. Arch Bronconeumol 2018; 55:31-37. [PMID: 29853259 DOI: 10.1016/j.arbres.2018.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/16/2018] [Accepted: 03/07/2018] [Indexed: 12/12/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a frequent and life-threatening entity. Recently, it has been demonstrated that diffuse alveolar damage (DAD), which is considered the histological hallmark in spite of presenting itself in only half of living patients with ARDS, exerts a relevant effect in the ARDS outcome. Despite the fact that the bronchial tree constitutes approximately 1% of the lung volume, discovering a relation between DAD and bronchial tree findings could be of paramount importance for a few reasons; (a) it could improve the description of ARDS with DAD as a clinical-pathological entity, (b) it could subrogate DAD findings with the advantage of their more accessible and safer analysis and (c) it could allow the discovery of new therapeutic targets. This narrative review is focused on pathological airway changes associated to Diffuse Alveolar Damage in the context of Acute Respiratory Distress Syndrome. It is organized into five sections: main anatomical and functional features of the human airway, why it is necessary to study airway features associated to DAD in patients with ARDS, pathological airway changes associated with DAD in animal models of ARDS, pathological airway changes associated with DAD in patients with ARDS, and the newest techniques for studying the histology of the bronchial tree and lung parenchyma.
Collapse
Affiliation(s)
- Guillermo Ortiz
- Universidad del Bosque, Bogotá, Colombia; Universidad de Barcelona, Barcelona, Spain
| | | | - Vera Capelozzi
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Pablo Cardinal-Fernández
- Emergency Department, Hospital Universitario HM Sanchinarro, Madrid, Spain; HM Research Foundation, Madrid, Spain.
| |
Collapse
|
50
|
Bonniaud P, Fabre A, Frossard N, Guignabert C, Inman M, Kuebler WM, Maes T, Shi W, Stampfli M, Uhlig S, White E, Witzenrath M, Bellaye PS, Crestani B, Eickelberg O, Fehrenbach H, Guenther A, Jenkins G, Joos G, Magnan A, Maitre B, Maus UA, Reinhold P, Vernooy JHJ, Richeldi L, Kolb M. Optimising experimental research in respiratory diseases: an ERS statement. Eur Respir J 2018; 51:13993003.02133-2017. [PMID: 29773606 DOI: 10.1183/13993003.02133-2017] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 04/02/2018] [Indexed: 12/15/2022]
Abstract
Experimental models are critical for the understanding of lung health and disease and are indispensable for drug development. However, the pathogenetic and clinical relevance of the models is often unclear. Further, the use of animals in biomedical research is controversial from an ethical perspective.The objective of this task force was to issue a statement with research recommendations about lung disease models by facilitating in-depth discussions between respiratory scientists, and to provide an overview of the literature on the available models. Focus was put on their specific benefits and limitations. This will result in more efficient use of resources and greater reduction in the numbers of animals employed, thereby enhancing the ethical standards and translational capacity of experimental research.The task force statement addresses general issues of experimental research (ethics, species, sex, age, ex vivo and in vitro models, gene editing). The statement also includes research recommendations on modelling asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, lung infections, acute lung injury and pulmonary hypertension.The task force stressed the importance of using multiple models to strengthen validity of results, the need to increase the availability of human tissues and the importance of standard operating procedures and data quality.
Collapse
Affiliation(s)
- Philippe Bonniaud
- Service de Pneumologie et Soins Intensifs Respiratoires, Centre Hospitalo-Universitaire de Bourgogne, Dijon, France.,Faculté de Médecine et Pharmacie, Université de Bourgogne-Franche Comté, Dijon, France.,INSERM U866, Dijon, France
| | - Aurélie Fabre
- Dept of Histopathology, St Vincent's University Hospital, UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Nelly Frossard
- Laboratoire d'Innovation Thérapeutique, Université de Strasbourg, Strasbourg, France.,CNRS UMR 7200, Faculté de Pharmacie, Illkirch, France.,Labex MEDALIS, Université de Strasbourg, Strasbourg, France
| | - Christophe Guignabert
- INSERM UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Sud and Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Mark Inman
- Dept of Medicine, Firestone Institute for Respiratory Health at St Joseph's Health Care MDCL 4011, McMaster University, Hamilton, ON, Canada
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Tania Maes
- Dept of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Wei Shi
- Developmental Biology and Regenerative Medicine Program, The Saban Research Institute of Children's Hospital Los Angeles, Los Angeles, CA, USA.,Dept of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Martin Stampfli
- Dept of Medicine, Firestone Institute for Respiratory Health at St Joseph's Health Care MDCL 4011, McMaster University, Hamilton, ON, Canada.,Dept of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University
| | - Stefan Uhlig
- Institute of Pharmacology and Toxicology, RWTH Aachen University, Aachen, Germany
| | - Eric White
- Division of Pulmonary and Critical Care Medicine, Dept of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Martin Witzenrath
- Dept of Infectious Diseases and Respiratory Medicine And Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Pierre-Simon Bellaye
- Département de Médecine nucléaire, Plateforme d'imagerie préclinique, Centre George-François Leclerc (CGFL), Dijon, France
| | - Bruno Crestani
- Assistance Publique-Hôpitaux de Paris, Hôpital Bichat, DHU FIRE, Service de Pneumologie A, Paris, France.,INSERM UMR 1152, Paris, France.,Université Paris Diderot, Paris, France
| | - Oliver Eickelberg
- Division of Pulmonary Sciences and Critical Care Medicine, Dept of Medicine, University of Colorado, Aurora, CO, USA
| | - Heinz Fehrenbach
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany.,Member of the Leibniz Research Alliance Health Technologies
| | - Andreas Guenther
- Justus-Liebig-University Giessen, Universitary Hospital Giessen, Agaplesion Lung Clinic Waldhof-Elgershausen, German Center for Lung Research, Giessen, Germany
| | - Gisli Jenkins
- Nottingham Biomedical Research Centre, Respiratory Research Unit, City Campus, University of Nottingham, Nottingham, UK
| | - Guy Joos
- Dept of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Antoine Magnan
- Institut du thorax, CHU de Nantes, Université de Nantes, Nantes, France
| | - Bernard Maitre
- Hôpital H Mondor, AP-HP, Centre Hospitalier Intercommunal de Créteil, Service de Pneumologie et de Pathologie Professionnelle, DHU A-TVB, Université Paris Est - Créteil, Créteil, France
| | - Ulrich A Maus
- Hannover School of Medicine, Division of Experimental Pneumology, Hannover, Germany
| | - Petra Reinhold
- Institute of Molecular Pathogenesis at the 'Friedrich-Loeffler-Institut' (Federal Research Institute for Animal Health), Jena, Germany
| | - Juanita H J Vernooy
- Dept of Respiratory Medicine, Maastricht University Medical Center+ (MUMC+), AZ Maastricht, The Netherlands
| | - Luca Richeldi
- UOC Pneumologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario "A. Gemelli", Rome, Italy
| | - Martin Kolb
- Dept of Medicine, Firestone Institute for Respiratory Health at St Joseph's Health Care MDCL 4011, McMaster University, Hamilton, ON, Canada
| |
Collapse
|