1
|
Ahangari F, Soudi S, Ghaffari Khaligh S, Mirsanei Z, Soufihasanabad S, Ebadi Asl P, Mahmoud Hashemi S. Combinational therapy of mesenchymal stem cell-derived extracellular vesicles and azithromycin improves clinical and histopathological recovery in CLP sepsis model. Int Immunopharmacol 2024; 139:112732. [PMID: 39053229 DOI: 10.1016/j.intimp.2024.112732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Sepsis is a syndrome that occurs following an infection and marked by severe inflammatory responses, and if not treated in time, it can lead to multi-organ failure syndrome and death. This study examines the effects of a novel combination therapy using azithromycin and mesenchymal stem cell-derived extracellular vesicles (EVs) on a cecal ligation and puncture (CLP) model of sepsis. METHODS Human Wharton's jelly-mesenchymal stem cells were cultured, characterized, and used to extract EVs. The CLP sepsis model was induced in mice, followed by treatments: saline, AZM, EVs, and combination therapy (A+E). Clinical sepsis scores were recorded 24 h post-treatment. Serum, peritoneal fluid, and organ tissues (kidney, liver, lung) were collected and analyzed for biochemical parameters (AST ALT, and creatinine), inflammatory markers, bacterial load, and histopathological changes. RESULTS The A+E combined treatment improved the clinical scores of septic mice. The administration of A+E reduced bacterial loads in the peritoneum of septic mice, contributing to effective control of infection. Inflammatory markers of neutrophils-to-lymphocytes ratio (NLR) and TNF-α serum levels were significantly lower in the combinational therapy group, indicating significant anti-inflammatory effect of this combination. Additionally, combination of AZM and EVs alleviated organ damage mainly within liver, kidneys and lungs. Based on histopathological assessments and biochemical parameters, there was diminished tissue damage as well as reduced inflammation, which is correlated with improved functions of these vital organs. CONCLUSION The combined use of azithromycin and EVs offers a promising therapeutic approach for sepsis by effectively controlling infection and modulating the inflammatory response.
Collapse
Affiliation(s)
- Fatemeh Ahangari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Zahra Mirsanei
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Soufihasanabad
- Department of Animal Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Pedram Ebadi Asl
- Department of Medical Lab Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Nishiyama K, Ichikado K, Anan K, Nakamura K, Kawamura K, Suga M, Sakagami T. The ROX index (Index combining the respiratory rate with oxygenation) is a prognostic factor for acute respiratory distress syndrome. PLoS One 2023; 18:e0282241. [PMID: 36848346 PMCID: PMC9970069 DOI: 10.1371/journal.pone.0282241] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/10/2023] [Indexed: 03/01/2023] Open
Abstract
BACKGROUND There is no existing reliable and practical method for predicting the prognosis of acute respiratory distress syndrome (ARDS). OBJECTIVE We aimed to clarify the association between the ROX index, which is calculated as the ratio of peripheral oxygen saturation divided by the fraction of inspired oxygen to the respiratory rate, and the prognosis of patients with ARDS under ventilator support. METHODS In this single-center retrospective cohort study from prospectively collected database, eligible patients were categorized into three groups based on ROX tertiles. The primary outcome was the 28-day survival, and the secondary outcome was 28-day liberation from ventilator support. We performed multivariable analysis using the Cox proportional hazards model. RESULTS Among 93 eligible patients, 24 (26%) patients died. The patients were divided into three groups according to the ROX index (< 7.4, 7.4-11, ≥ 11), with 13, 7, and 4 patients dying in the groups, respectively. A higher ROX index was associated with lower mortality; adjusted hazard ratios [95% CIs] for increasing tertiles of ROX index: 1[reference], 0.54[0.21-1.41], 0.23[0.074-0.72] (P = 0.011 for trend) and a higher rate of successful 28-day liberation from ventilator support; adjusted hazard ratios [95% CIs] for increasing tertiles of ROX index: 1[reference], 1.41[0.68-2.94], 2.80[1.42-5.52] (P = 0.001 for trend). CONCLUSIONS The ROX index at 24 h after initiating ventilator support is a predictor of outcomes in patients with ARDS and might inform initiation of more advanced treatments.
Collapse
Affiliation(s)
- Kenta Nishiyama
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
- * E-mail:
| | - Kazuya Ichikado
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Keisuke Anan
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Kazunori Nakamura
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Kodai Kawamura
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Moritaka Suga
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Takuro Sakagami
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
3
|
Potruch A, Schwartz A, Ilan Y. The role of bacterial translocation in sepsis: a new target for therapy. Therap Adv Gastroenterol 2022; 15:17562848221094214. [PMID: 35574428 PMCID: PMC9092582 DOI: 10.1177/17562848221094214] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
Sepsis is a leading cause of death in critically ill patients, primarily due to multiple organ failures. It is associated with a systemic inflammatory response that plays a role in the pathogenesis of the disease. Intestinal barrier dysfunction and bacterial translocation (BT) play pivotal roles in the pathogenesis of sepsis and associated organ failure. In this review, we describe recent advances in understanding the mechanisms by which the gut microbiome and BT contribute to the pathogenesis of sepsis. We also discuss several potential treatment modalities that target the microbiome as therapeutic tools for patients with sepsis.
Collapse
|
4
|
Suzuki J, Sasabuchi Y, Hatakeyama S, Matsui H, Sasahara T, Morisawa Y, Yamada T, Fushimi K, Yasunaga H. Additional effect of azithromycin over β-lactam alone for severe community-acquired pneumonia-associated acute respiratory distress syndrome: a retrospective cohort study. Pneumonia (Nathan) 2022; 14:1. [PMID: 35012674 PMCID: PMC8744237 DOI: 10.1186/s41479-021-00093-8] [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: 08/05/2021] [Accepted: 12/12/2021] [Indexed: 11/24/2022] Open
Abstract
Background Community-acquired pneumonia (CAP) is the most common cause of acute respiratory distress syndrome (ARDS). Although previous studies have suggested that macrolide therapy is beneficial for ARDS, its benefit for severe CAP-associated ARDS remains uncertain. Previous studies were limited in that they had a small sample size and included patients with non-pulmonary ARDS and those with pulmonary ARDS. This study aimed to investigate the additional effect of azithromycin when used with β-lactam compared with the effect of β-lactam alone in mechanically ventilated patients with CAP-associated ARDS. Methods We identified mechanically ventilated patients with CAP-associated ARDS between July 2010 and March 2015 using data in the Diagnosis Procedure Combination database, a Japanese nationwide inpatient database. We performed propensity score matching analysis to assess 28-day mortality and in-hospital mortality in mechanically ventilated patients with CAP-associated ARDS who received β-lactam with and without azithromycin within hospital 2 days after admission. The inverse probability of treatment weighting analysis was also conducted. Results Eligible patients (n = 1257) were divided into the azithromycin group (n = 226) and the control group (n = 1031). The one-to-four propensity score matching analysis included 139 azithromycin users and 556 non-users. No significant difference was observed between the groups with respect to 28-day mortality (34.5% vs. 37.6%, p = 0.556) or in-hospital mortality (46.0% vs. 49.1%, p = 0.569). The inverse probability of treatment weighting analysis showed similar results. Conclusions Compared with treatment with β-lactam alone, treatment with azithromycin plus β-lactam had no significant additional effect on 28-day mortality or in-hospital mortality in mechanically ventilated patients with CAP-associated ARDS. To the best of our knowledge, this study is the first to determine the effect of azithromycin in mechanically ventilated patients with CAP-associated ARDS. Supplementary Information The online version contains supplementary material available at 10.1186/s41479-021-00093-8.
Collapse
Affiliation(s)
- Jun Suzuki
- Division of Infectious Diseases, Jichi Medical University Hospital, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| | - Yusuke Sasabuchi
- Center for Data Science, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Shuji Hatakeyama
- Division of Infectious Diseases, Jichi Medical University Hospital, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.,Division of General Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Hiroki Matsui
- Department of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Teppei Sasahara
- Division of Infectious Diseases, Jichi Medical University Hospital, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.,Department of Infection and Immunity, School of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Yuji Morisawa
- Division of Infectious Diseases, Jichi Medical University Hospital, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Toshiyuki Yamada
- Department of Clinical Laboratory Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Kiyohide Fushimi
- Department of Health Policy and Informatics, Tokyo Medical and Dental University Graduate School of Medicine, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Hideo Yasunaga
- Department of Clinical Epidemiology and Health Economics, School of Public Health, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
5
|
Sauer A, Peukert K, Putensen C, Bode C. Antibiotics as immunomodulators: a potential pharmacologic approach for ARDS treatment. Eur Respir Rev 2021; 30:210093. [PMID: 34615700 PMCID: PMC9489085 DOI: 10.1183/16000617.0093-2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/02/2021] [Indexed: 11/05/2022] Open
Abstract
First described in the mid-1960s, acute respiratory distress syndrome (ARDS) is a life-threatening form of respiratory failure with an overall mortality rate of approximately 40%. Despite significant advances in the understanding and treatment of ARDS, no substantive pharmacologic therapy has proven to be beneficial, and current management continues to be primarily supportive. Beyond their antibacterial activity, several antibiotics such as macrolides and tetracyclines exert pleiotropic immunomodulatory effects that might be able to rectify the dysregulated inflammatory response present in patients with ARDS. This review aims to provide an overview of preclinical and clinical studies that describe the immunomodulatory effects of antibiotics in ARDS. Moreover, the underlying mechanisms of their immunomodulatory properties will be discussed. Further studies are necessary to investigate their full therapeutic potential and to identify ARDS phenotypes which are most likely to benefit from their immunomodulatory effects.
Collapse
Affiliation(s)
- Andrea Sauer
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Konrad Peukert
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Putensen
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Christian Bode
- Dept of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
6
|
Pons S, Arrii E, Arnaud M, Loiselle M, Ferry J, Nouacer M, Lion J, Cohen S, Mooney N, Zafrani L. Immunomodulation of endothelial cells induced by macrolide therapy in a model of septic stimulation. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1656-1669. [PMID: 34636179 PMCID: PMC8589380 DOI: 10.1002/iid3.518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 01/18/2023]
Abstract
Objectives Sepsis is defined as the host's inflammatory response to a life‐threatening infection. The endothelium is implicated in immunoregulation during sepsis. Macrolides have been proposed to display immunomodulatory properties. The goal of this study was to analyze whether macrolides can exert immunomodulation of endothelial cells (ECs) in an experimental model of sepsis. Methods Human ECs were stimulated by proinflammatory cytokines and lipopolysaccharide before exposure to macrolides. ECs phenotypes were analyzed by flow cytometry. Cocultures of ECs and peripheral blood mononuclear cells (PBMCs) were performed to study the ECs ability to alter T‐cell viability and differentiation in the presence of macrolides. Soluble factor production was assessed. Results ECs act as non‐professional antigen presenting cells and expressed human leukocyte antigen (HLA) antigens, the adhesion molecules CD54, CD106, and the coinhibitory molecule CD274 after septic stimulation. Incubation with macrolides induced a significant decrease of HLA class I and HLA class II HLA‐DR on septic‐stimulated ECs, but did not alter either CD54, CD106, nor CD274 expression. Interleukin‐6 (IL‐6) and IL‐8 production by stimulated ECs were unaltered by incubation with macrolides, whereas Clarithromycin exposure significantly decreased IL‐6 gene expression. In cocultures of septic ECs with PBMCs, neither the proportion of CD4 + , CD8 + T nor their viability was altered by macrolides. T‐helper lymphocyte subsets Th1, Th17, and Treg polarization by stimulated ECs were unaltered by macrolides. Conclusion This study reports phenotypic and gene expression changes in septic‐stimulated ECs exposed to macrolides, without resulting in altered immunogenicity of ECs in co‐cultures with PBMCs. In vivo studies may help to further understand the impact of macrolide therapy on ECs immune homeostasis during sepsis.
Collapse
Affiliation(s)
- Stéphanie Pons
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France.,Department of Anesthesiology and Critical Care, Pitié-Salpêtrière Hospital, GRC 29, AP-HP, DMU DREAM, Sorbonne University, Paris, France
| | - Eden Arrii
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Marine Arnaud
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Maud Loiselle
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Juliette Ferry
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Manel Nouacer
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Julien Lion
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Shannon Cohen
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Nuala Mooney
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France
| | - Lara Zafrani
- Human Immunology, Pathophysiology, Immunotherapy (HIPI), INSERM U976, Université de Paris, Paris, France.,Medical Intensive Care Unit, AP-HP, Saint-Louis Teaching Hospital, Paris, France
| |
Collapse
|
7
|
Clinical phenotypes from fatal cases of acute respiratory distress syndrome caused by pneumonia. Sci Rep 2021; 11:20051. [PMID: 34625618 PMCID: PMC8501115 DOI: 10.1038/s41598-021-99540-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
There have been no report of objective clinical characteristics or prognostic factors that predict fatal outcome of acute respiratory distress syndrome (ARDS) since the Berlin definition was published. The aim of this study is to identify clinically available predictors that distinguish between two phenotypes of fatal ARDS due to pneumonia. In total, 104 cases of Japanese patients with pneumonia-induced ARDS were extracted from our prospectively collected database. Fatal cases were divided into early (< 7 days after diagnosis) and late (≥ 7 days) death groups, and clinical variables and prognostic factors were statistically evaluated. Of the 50 patients who died within 180 days, 18 (36%) and 32 (64%) were in the early (median 2 days, IQR [1, 5]) and late (median 16 days, IQR [13, 29]) death groups, respectively. According to multivariate regression analyses, the APACHE II score (HR 1.25, 95%CI 1.12–1.39, p < 0.001) and the disseminated intravascular coagulation score (HR 1.54, 95%CI 1.15–2.04, p = 0.003) were independent prognostic factors for early death. In contrast, late death was associated with high-resolution computed tomography (HRCT) score indicating early fibroproliferation (HR 1.28, 95%CI 1.13–1.42, p < 0.001) as well as the disseminated intravascular coagulation score (HR 1.24, 95%CI 1.01–1.52, p = 0.039). The extent of fibroproliferation on HRCT, and the APACHE II scores along with coagulation abnormalities, should be considered for use in predictive enrichment and personalized medicine for patients with ARDS due to pneumonia.
Collapse
|
8
|
Gyselinck I, Liesenborghs L, Landeloos E, Belmans A, Verbeke G, Verhamme P, Vos R, Janssens W. Direct antivirals working against the novel coronavirus: azithromycin (DAWn-AZITHRO), a randomized, multicenter, open-label, adaptive, proof-of-concept clinical trial of new antivirals working against SARS-CoV-2-azithromycin trial. Trials 2021; 22:126. [PMID: 33563325 PMCID: PMC7871018 DOI: 10.1186/s13063-021-05033-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 01/08/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The rapid emergence and the high disease burden of the novel coronavirus SARS-CoV-2 have created a medical need for readily available drugs that can decrease viral replication or blunt the hyperinflammatory state leading to severe COVID-19 disease. Azithromycin is a macrolide antibiotic, known for its immunomodulatory properties. It has shown antiviral effect specifically against SARS-CoV-2 in vitro and acts on cytokine signaling pathways that have been implicated in COVID-19. METHODS DAWn-AZITHRO is a randomized, open-label, phase 2 proof-of-concept, multicenter clinical trial, evaluating the safety and efficacy of azithromycin for treating hospitalized patients with COVID-19. It is part of a series of trials testing promising interventions for COVID-19, running in parallel and grouped under the name DAWn-studies. Patients hospitalized on dedicated COVID wards are eligible for study inclusion when they are symptomatic (i.e., clinical or radiological signs) and have been diagnosed with COVID-19 within the last 72 h through PCR (nasopharyngeal swab or bronchoalveolar lavage) or chest CT scan showing typical features of COVID-19 and without alternate diagnosis. Patients are block-randomized (9 patients) with a 2:1 allocation to receive azithromycin plus standard of care versus standard of care alone. Standard of care is mostly supportive, but may comprise hydroxychloroquine, up to the treating physician's discretion and depending on local policy and national health regulations. The treatment group receives azithromycin qd 500 mg during the first 5 consecutive days after inclusion. The trial will include 284 patients and recruits from 15 centers across Belgium. The primary outcome is time from admission (day 0) to life discharge or to sustained clinical improvement, defined as an improvement of two points on the WHO 7-category ordinal scale sustained for at least 3 days. DISCUSSION The trial investigates the urgent and still unmet global need for drugs that may impact the disease course of COVID-19. It will either provide support or else justify the discouragement of the current widespread, uncontrolled use of azithromycin in patients with COVID-19. The analogous design of other parallel trials of the DAWN consortium will amplify the chance of identifying successful treatment strategies and allow comparison of treatment effects within an identical clinical context. TRIAL REGISTRATION EU Clinical trials register EudraCT Nb 2020-001614-38 . Registered on 22 April 2020.
Collapse
Affiliation(s)
- Iwein Gyselinck
- Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Belgium.
| | - Laurens Liesenborghs
- Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Belgium
| | - Ewout Landeloos
- Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Belgium
| | - Ann Belmans
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Leuven, Belgium
| | - Geert Verbeke
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Leuven, Belgium
| | - Peter Verhamme
- Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Belgium
| | - Robin Vos
- Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Belgium
| | - W Janssens
- Katholieke Universiteit Leuven Universitaire Ziekenhuizen Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Gyselinck I, Janssens W, Verhamme P, Vos R. Rationale for azithromycin in COVID-19: an overview of existing evidence. BMJ Open Respir Res 2021; 8:e000806. [PMID: 33441373 PMCID: PMC7811960 DOI: 10.1136/bmjresp-2020-000806] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
Azithromycin has rapidly been adopted as a repurposed drug for the treatment of COVID-19, despite the lack of high-quality evidence. In this review, we critically appraise the current pharmacological, preclinical and clinical data of azithromycin for treating COVID-19. Interest in azithromycin has been fuelled by favourable treatment outcomes in other viral pneumonias, a documented antiviral effect on SARS-CoV-2 in vitro and uncontrolled case series early in the pandemic. Its antiviral effects presumably result from interfering with receptor mediated binding, viral lysosomal escape, intracellular cell-signalling pathways and enhancing type I and III interferon expression. Its immunomodulatory effects may mitigate excessive inflammation and benefit tissue repair. Currently, in vivo reports on azithromycin in COVID-19 are conflicting and do not endorse its widespread use outside of clinical trials. They are, however, mostly retrospective and therefore inherently biased. The effect size of azithromycin may depend on when it is started. Also, extended follow-up is needed to assess benefits in the recovery phase. Safety data warrant monitoring of drug-drug interactions and subsequent cardiac adverse events, especially with hydroxychloroquine. More prospective data of large randomised controlled studies are expected and much-needed. Uniform reporting of results should be strongly encouraged to facilitate data pooling with the many ongoing initiatives.
Collapse
Affiliation(s)
- Iwein Gyselinck
- Respiratory Diseases, KU Leuven University Hospitals, Leuven, Flanders, Belgium
- Department CHROMETA - Research group BREATHE, KU Leuven, Leuven, Flanders, Belgium
| | - Wim Janssens
- Respiratory Diseases, KU Leuven University Hospitals, Leuven, Flanders, Belgium
- Department CHROMETA - Research group BREATHE, KU Leuven, Leuven, Flanders, Belgium
| | - Peter Verhamme
- Cardiovascular Diseases, KU Leuven University Hospitals, Leuven, Flanders, Belgium
- Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Flanders, Belgium
| | - Robin Vos
- Respiratory Diseases, KU Leuven University Hospitals, Leuven, Flanders, Belgium
- Department CHROMETA - Research group BREATHE, KU Leuven, Leuven, Flanders, Belgium
| |
Collapse
|
10
|
Morris G, Bortolasci CC, Puri BK, Olive L, Marx W, O'Neil A, Athan E, Carvalho A, Maes M, Walder K, Berk M. Preventing the development of severe COVID-19 by modifying immunothrombosis. Life Sci 2021; 264:118617. [PMID: 33096114 PMCID: PMC7574725 DOI: 10.1016/j.lfs.2020.118617] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/01/2020] [Accepted: 10/13/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND COVID-19-associated acute respiratory distress syndrome (ARDS) is associated with significant morbidity and high levels of mortality. This paper describes the processes involved in the pathophysiology of COVID-19 from the initial infection and subsequent destruction of type II alveolar epithelial cells by SARS-CoV-2 and culminating in the development of ARDS. MAIN BODY The activation of alveolar cells and alveolar macrophages leads to the release of large quantities of proinflammatory cytokines and chemokines and their translocation into the pulmonary vasculature. The presence of these inflammatory mediators in the vascular compartment leads to the activation of vascular endothelial cells platelets and neutrophils and the subsequent formation of platelet neutrophil complexes. These complexes in concert with activated endothelial cells interact to create a state of immunothrombosis. The consequence of immunothrombosis include hypercoagulation, accelerating inflammation, fibrin deposition, migration of neutrophil extracellular traps (NETs) producing neutrophils into the alveolar apace, activation of the NLRP3 inflammazome, increased alveolar macrophage destruction and massive tissue damage by pyroptosis and necroptosis Therapeutic combinations aimed at ameliorating immunothrombosis and preventing the development of severe COVID-19 are discussed in detail.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | | | - Lisa Olive
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; School of Psychology, Deakin University, Geelong, Australia
| | - Wolfgang Marx
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Melbourne School of Population and Global Health, Melbourne, Australia
| | - Eugene Athan
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Barwon Health, Geelong, Australia
| | - Andre Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, University of Toronto, Toronto, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
11
|
Morris G, Athan E, Walder K, Bortolasci CC, O'Neil A, Marx W, Berk M, Carvalho AF, Maes M, Puri BK. Can endolysosomal deacidification and inhibition of autophagy prevent severe COVID-19? Life Sci 2020; 262:118541. [PMID: 33035581 PMCID: PMC7537668 DOI: 10.1016/j.lfs.2020.118541] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023]
Abstract
The possibility is examined that immunomodulatory pharmacotherapy may be clinically useful in managing the pandemic coronavirus disease 2019 (COVID-19), known to result from infection by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a positive-sense single-stranded RNA virus. The dominant route of cell entry of the coronavirus is via phagocytosis, with ensconcement in endosomes thereafter proceeding via the endosomal pathway, involving transfer from early (EEs) to late endosomes (LEs) and ultimately into lysosomes via endolysosomal fusion. EE to LE transportation is a rate-limiting step for coronaviruses. Hence inhibition or dysregulation of endosomal trafficking could potentially inhibit SARS-CoV-2 replication. Furthermore, the acidic luminal pH of the endolysosomal system is critical for the activity of numerous pH-sensitive hydrolytic enzymes. Golgi sub-compartments and Golgi-derived secretory vesicles also depend on being mildly acidic for optimal function and structure. Activation of endosomal toll-like receptors by viral RNA can upregulate inflammatory mediators and contribute to a systemic inflammatory cytokine storm, associated with a worsened clinical outcome in COVID-19. Such endosomal toll-like receptors could be inhibited by the use of pharmacological agents which increase endosomal pH, thereby reducing the activity of acid-dependent endosomal proteases required for their activity and/or assembly, leading to suppression of antigen-presenting cell activity, decreased autoantibody secretion, decreased nuclear factor-kappa B activity and decreased pro-inflammatory cytokine production. It is also noteworthy that SARS-CoV-2 inhibits autophagy, predisposing infected cells to apoptosis. It is therefore also suggested that further pharmacological inhibition of autophagy might encourage the apoptotic clearance of SARS-CoV-2-infected cells.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Eugene Athan
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Department of Infectious Disease, Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Victoria, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Wolf Marx
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Michael Berk
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, the Department of Psychiatry, the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - André F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - Michael Maes
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, Barwon Health, School of Medicine, Geelong, Victoria, Australia; Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | | |
Collapse
|
12
|
Brown SM, Peltan I, Kumar N, Leither L, Webb BJ, Starr N, Grissom CK, Buckel WR, Srivastava R, Butler AM, Groat D, Haaland B, Ying J, Harris E, Johnson S, Paine R, Greene T. Hydroxychloroquine vs. Azithromycin for Hospitalized Patients with COVID-19 (HAHPS): Results of a Randomized, Active Comparator Trial. Ann Am Thorac Soc 2020; 18:590-597. [PMID: 33166179 PMCID: PMC8009003 DOI: 10.1513/annalsats.202008-940oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022] Open
Abstract
RATIONALE The COVID-19 pandemic struck an immunologically naïve, globally interconnected population. In the face of a new infectious agent causing acute respiratory failure for which there were no known effective therapies, rapid, often pragmatic trials were necessary to evaluate potential treatments, frequently starting with medications that are already marketed for other indications. Early in the pandemic, hydroxychloroquine and azithromycin were two such candidates. OBJECTIVE Assess the relative efficacy of hydroxychloroquine and azithromycin among hospitalized patients with COVID-19. METHODS We performed a randomized clinical trial of hydroxychloroquine vs. azithromycin among hospitalized patients with COVID-19. Treatment was 5 days of study medication. The primary endpoint was the COVID Ordinal Outcomes scale at day 14. Secondary endpoints included hospital-, ICU-, and ventilator-free days at day 28. The trial was stopped early after enrollment of 85 patients when a separate clinical trial concluded that a clinically important effect of hydroxychloroquine over placebo was definitively excluded. Comparisons were made a priori using a proportional odds model from a Bayesian perspective. RESULTS We enrolled 85 patients at 13 hospitals over 11 weeks. Adherence to study medication was high. The estimated odds ratio for less favorable status on the ordinal scale for hydroxychloroquine vs. azithromycin from the primary analysis was 1.07, with a 95% credible interval from 0.63 to 1.83 with a posterior probability of 60% that hydroxychloroquine was worse than azithryomycin. Secondary outcomes displayed a similar, slight preference for azithromycin over hydroxychloroquine. QTc prolongation was rare and did not differ between groups. The twenty safety outcomes were similar between arms with the possible exception of post-randomization onset acute kidney injury, which was more common with hydroxychloroquine (15% vs. 0%). Patients in the hydroxychloroquine arm received remdesivir more often than in the azithromycin arm (19% vs. 2%). There was no apparent association between remdesivir use and acute kidney injury. CONCLUSIONS While early termination limits the precision of our results, we found no suggestion of substantial efficacy for hydroxychloroquine over azithromycin. Acute kidney injury may be more common with hydroxychloroquine than azithromycin, although this may be due to the play of chance. Differential use of remdesivir may have biased our results in favor of hydroxychloroquine. Our results are consistent with conclusions from other trials that hydroxychloroquine cannot be recommended for inpatients with COVID-19; azithromycin may merit additional investigation. CLINICAL TRIAL REGISTRATION This trial was prospectively registered (NCT04329832) before enrollment of the first patient.
Collapse
Affiliation(s)
- Samuel M Brown
- Intermountain Medical Center, Center for Humanizing Critical Care, Murray, Utah, United States;
| | - Ithan Peltan
- Intermountain Medical Center, 98078, Division of Pulmonary & Critical Care Medicine, Murray, Utah, United States
- University of Utah School of Medicine, 12348, Division of Pulmonary & Critical Care Medicine, Salt Lake City, Utah, United States
| | - Naresh Kumar
- Intermountain Medical Center, Office of Research, Murray, Utah, United States
| | - Lindsay Leither
- Intermountain Medical Center, 98078, Murray, Utah, United States
| | - Brandon J Webb
- Intermountain Medical Center, Division of Clinical Epidemiology and Infectious Diseases, Murray, Utah, United States
| | - Nathan Starr
- Intermountain Medical Center, Internal Medicine, Murray, Utah, United States
| | - Colin K Grissom
- Intermountain Medical Center, Critical Care Medicine, Murray, Utah, United States
| | - Whitney R Buckel
- Intermountain Medical Center, Pharmacy, Murray, Utah, United States
| | - Rajendu Srivastava
- Intermountain Medical Center, 98078, Office of Research, Murray, Utah, United States
| | - Allison M Butler
- Intermountain Healthcare, Statistical Data Center, Salt Lake City, Utah, United States
| | - Danielle Groat
- Intermountain Medical Center, Center for Humanizing Critical Care, Murray, Utah, United States
| | - Benjamin Haaland
- University of Utah, Biostatistics, Salt Lake City, Utah, United States
| | - Jian Ying
- University of Utah, Biostatistics, Salt Lake City, Utah, United States
| | - Estelle Harris
- University of Utah School of Medicine, 12348, Division of Pulmonary & Critical Care Medicine, Salt Lake City, Utah, United States
| | - Stacy Johnson
- University of Utah School of Medicine, 12348, Internal Medicine, Salt Lake City, Utah, United States
| | - Robert Paine
- University of Utah, Salt Lake City, Utah, United States
| | - Tom Greene
- University of Utah School of Medicine, 12348, Study Design and Biostatistics Center and Division of Epidemiology, Salt Lake City, Utah, United States
| |
Collapse
|
13
|
Lauriola M, Pani A, Ippoliti G, Mortara A, Milighetti S, Mazen M, Perseghin G, Pastori D, Grosso P, Scaglione F. Effect of Combination Therapy of Hydroxychloroquine and Azithromycin on Mortality in Patients With COVID-19. Clin Transl Sci 2020; 13:1071-1076. [PMID: 32926573 PMCID: PMC7719367 DOI: 10.1111/cts.12860] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 07/13/2020] [Indexed: 01/10/2023] Open
Abstract
Conflicting evidence regarding the use of hydroxychloroquine (HCQ) and azithromycin for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection do exist. We performed a retrospective single-center cohort study including 377 consecutive patients admitted for pneumonia related to coronavirus disease 2019 (COVID-19). Of these, 297 were in combination treatment, 17 were on HCQ alone, and 63 did not receive either of these 2 drugs because of contraindications. The primary end point was in-hospital death. Mean age was 71.8 ± 13.4 years and 34.2% were women. We recorded 146 deaths: 35 in no treatment, 7 in HCQ treatment group, and 102 in HCQ + azithromycin treatment group (log rank test for Kaplan-Meier curve P < 0.001). At multivariable Cox proportional hazard regression analysis, age (hazard ratio (HR) 1.057, 95% confidence interval (CI) 1.035-1.079, P < 0.001), mechanical ventilation/continuous positive airway pressure (HR 2.726, 95% CI 1.823-4.074, P < 0.001), and C reactive protein above the median (HR 2.191, 95% CI 1.479-3.246, P < 0.001) were directly associated with death, whereas use of HCQ + azithromycin (vs. no treatment; HR 0.265, 95% CI 0.171-0.412, P < 0.001) was inversely associated. In this study, we found a reduced in-hospital mortality in patients treated with a combination of HCQ and azithromycin after adjustment for comorbidities. A large randomized trial is necessary to confirm these findings.
Collapse
Affiliation(s)
| | - Arianna Pani
- Department of Oncology and Hemato‐OncologyUniversità degli Studi di MilanoMilanItaly
| | | | | | | | - Marjieh Mazen
- Internal Medicine DepartmentPoliclinico di MonzaMonzaItaly
| | - Gianluca Perseghin
- Internal Medicine DepartmentPoliclinico di MonzaMonzaItaly
- Department of Medicine and SurgeryUniversità degli Studi di Milano BicoccaMilanItaly
| | - Daniele Pastori
- Department of Clinical, Internal, Anesthesiologic, and Cardiovascular SciencesSapienza University of RomeRomeItaly
| | - Paolo Grosso
- Intensive Care UnitPoliclinico di MonzaMonzaItaly
| | - Francesco Scaglione
- Department of Oncology and Hemato‐OncologyUniversità degli Studi di MilanoMilanItaly
| |
Collapse
|
14
|
Brown SM, Peltan ID, Webb B, Kumar N, Starr N, Grissom C, Buckel WR, Srivastava R, Harris ES, Leither LM, Johnson SA, Paine R, Greene T. Hydroxychloroquine versus Azithromycin for Hospitalized Patients with Suspected or Confirmed COVID-19 (HAHPS). Protocol for a Pragmatic, Open-Label, Active Comparator Trial. Ann Am Thorac Soc 2020; 17:1008-1015. [PMID: 32425051 PMCID: PMC7393787 DOI: 10.1513/annalsats.202004-309sd] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/18/2020] [Indexed: 01/04/2023] Open
Abstract
Coronavirus disease (COVID-19) is a potentially fatal illness with no proven therapy beyond excellent supportive care. Treatments are urgently sought. Adaptations to traditional trial logistics and design to allow rapid implementation, evaluation of trials within a global trials context, flexible interim monitoring, and access outside traditional research hospitals (even in settings where formal placebos are unavailable) may be helpful. Thoughtful adaptations to traditional trial designs, especially within the global context of related studies, may also foster collaborative relationships among government, community, and the research enterprise. Here, we describe the protocol for a pragmatic, active comparator trial in as many as 300 patients comparing two current "off-label" treatments for COVID-19-hydroxychloroquine and azithromycin-in academic and nonacademic hospitals in Utah. We developed the trial in response to local pressures for widespread, indiscriminate off-label use of these medications. We used a hybrid Bayesian-frequentist design for interim monitoring to allow rapid, contextual assessment of the available evidence. We also developed an inference grid for interpreting the range of possible results from this trial within the context of parallel trials and prepared for a network meta-analysis of the resulting data. This trial was prospectively registered (ClinicalTrials.gov Identifier: NCT04329832) before enrollment of the first patient.Clinical trial registered with www.clinicaltrials.gov (NCT04329832).
Collapse
Affiliation(s)
- Samuel M Brown
- Pulmonary and Critical Care Medicine
- Pulmonary and Critical Care Medicine
| | - Ithan D Peltan
- Pulmonary and Critical Care Medicine
- Pulmonary and Critical Care Medicine
| | - Brandon Webb
- Infectious Disease, and
- Infectious Disease
- Infectious Disease, Stanford University, Stanford, California; and
| | - Naresh Kumar
- Pulmonary and Critical Care Medicine
- Infectious Disease, and
| | - Nathan Starr
- Hospital Medicine, Intermountain Medical Center, Murray, Utah
| | - Colin Grissom
- Pulmonary and Critical Care Medicine
- Pulmonary and Critical Care Medicine
| | | | - Raj Srivastava
- Inpatient Medicine, Department of Pediatrics
- Healthcare Delivery Institute, Intermountain Healthcare, Murray, Utah
| | | | - Lindsay M Leither
- Pulmonary and Critical Care Medicine
- Pulmonary and Critical Care Medicine
| | | | | | - Tom Greene
- Biostatistics, University of Utah, Salt Lake City, Utah
| |
Collapse
|
15
|
A Scoring System with High-Resolution Computed Tomography to Predict Drug-Associated Acute Respiratory Distress Syndrome: Development and Internal Validation. Sci Rep 2019; 9:8601. [PMID: 31197186 PMCID: PMC6565715 DOI: 10.1038/s41598-019-45063-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 05/29/2019] [Indexed: 12/23/2022] Open
Abstract
Drugs can cause acute respiratory distress syndrome (ARDS). However, there is no established clinical prediction rule for drug-associated ARDS (DARDS). We aimed to develop and validate a scoring system for DARDS prediction. We analysed data collected from a prospective, single-centre, cohort study that included ARDS patients. The ARDS diagnosis was based on the American-European Consensus Conference or Berlin definition. Drug-associated acute lung injury (DALI) was defined as previous exposure to drugs which cause ALI and presence of traditional risk factors for ALI. High-resolution computed tomography (HRCT; indicating extent of lung damage with fibroproliferation), Acute Physiology and Chronic Health Evaluation (APACHE) II, and disseminated intravascular coagulation (DIC; indicating multiorgan failure) scores and PaO2/FiO2 were evaluated for their ability to predict DARDS. Twenty-nine of 229 patients had DARDS. The HRCT, APACHE II, and DIC scores and PaO2/FiO2 were assessed. The model-based predicted probability of DARDS fitted well with the observed data, and discrimination ability, assessed through bootstrap with an area under the receiver-operating curve, improved from 0.816 to 0.875 by adding the HRCT score. A simple clinical scoring system consisting of the APACHE II score, PaO2/FiO2, and DIC and HRCT scores can predict DARDS. This model may facilitate more appropriate clinical decision-making.
Collapse
|
16
|
Weis S, Heindl M, Carvalho T, Jentho E, Lorenz J, Sommerer I, Mössner J, Hoffmeister A. Azithromycin does not improve disease severity in acute experimental pancreatitis. PLoS One 2019; 14:e0216614. [PMID: 31075097 PMCID: PMC6510415 DOI: 10.1371/journal.pone.0216614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 04/24/2019] [Indexed: 12/31/2022] Open
Abstract
Acute pancreatitis is a severe systemic disease triggered by a sterile inflammation and initial local tissue damage of the pancreas. Immune cells infiltrating into the pancreas are main mediators of acute pancreatitis pathogenesis. In addition to their antimicrobial potency, macrolides possess anti-inflammatory and immunomodulatory properties which are routinely used in patients with chronic airway infections and might also beneficial in the treatment of acute lung injury. We here tested the hypothesis that the macrolide antibiotic azithromycin can improve the course of acute experimental pancreatitis via ameliorating the damage imposed by sterile inflammation, and could be used as a disease specific therapy. However, our data show that azithromycin does not have influence on caerulein induced acute pancreatitis in terms of reduction of organ damage, and disease severity. Furthermore Infiltration of immune cells into the pancreas or the lungs was not attenuated by azithromycin as compared to controls or ampicillin treated animals with acute experimental pancreatitis. We conclude that in the chosen model, azithromycin does not have any beneficial effects and that its immunomodulatory properties cannot be used to decrease disease severity in the model of caerulein-induced pancreatitis in mice.
Collapse
Affiliation(s)
- Sebastian Weis
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Institute for Infectious Disease and Infection Control, Jena University Hospital, Jena, Germany
- Division of Gastroenterology, University Hospital Leipzig, Leipzig, Germany
| | - Mario Heindl
- Division of Gastroenterology, University Hospital Leipzig, Leipzig, Germany
| | - Tania Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Elisa Jentho
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Jana Lorenz
- Division of Gastroenterology, University Hospital Leipzig, Leipzig, Germany
| | - Ines Sommerer
- Division of Gastroenterology, University Hospital Leipzig, Leipzig, Germany
| | - Joachim Mössner
- Division of Gastroenterology, University Hospital Leipzig, Leipzig, Germany
| | - Albrecht Hoffmeister
- Division of Gastroenterology, University Hospital Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
17
|
Pons S, Timsit JF, Ruckly S, Schwebel C, Papazian L, Azoulay E, Reignier J, Zafrani L. Impact of macrolide therapy in critically ill patients with acute respiratory failure: a desirability of outcome ranking analysis to investigate the OUTCOMEREA database. Intensive Care Med 2019; 45:1043-1045. [PMID: 30874823 DOI: 10.1007/s00134-019-05586-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2019] [Indexed: 10/27/2022]
Affiliation(s)
- Stéphanie Pons
- INSERM U976, Saint-Louis University Hospital, Paris, France
| | - Jean-François Timsit
- Infectious and Medical Intensive Care Unit, Bichat Claude-Bernard University Hospital, Paris, France.,INSERM IAME, U1137, Team DesCID, Paris, France
| | | | - Carole Schwebel
- Polyvalent Intensive Care Unit, Grenoble University Hospital, Grenoble, France
| | - Laurent Papazian
- Respiratory and Infectious Diseases Intensive Care Unit, North University Hospital, Marseille, France
| | - Elie Azoulay
- Medical Intensive Care Unit, Saint-Louis Hospital, 1, Avenue Claude Vellefaux, 75010, Paris, France
| | - Jean Reignier
- Medical Intensive Care Unit, Nantes University Hospital Center, Nantes, France
| | - Lara Zafrani
- INSERM U976, Saint-Louis University Hospital, Paris, France. .,Medical Intensive Care Unit, Saint-Louis Hospital, 1, Avenue Claude Vellefaux, 75010, Paris, France.
| |
Collapse
|
18
|
Mussbacher M, Salzmann M, Brostjan C, Hoesel B, Schoergenhofer C, Datler H, Hohensinner P, Basílio J, Petzelbauer P, Assinger A, Schmid JA. Cell Type-Specific Roles of NF-κB Linking Inflammation and Thrombosis. Front Immunol 2019; 10:85. [PMID: 30778349 PMCID: PMC6369217 DOI: 10.3389/fimmu.2019.00085] [Citation(s) in RCA: 375] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 01/11/2019] [Indexed: 12/22/2022] Open
Abstract
The transcription factor NF-κB is a central mediator of inflammation with multiple links to thrombotic processes. In this review, we focus on the role of NF-κB signaling in cell types within the vasculature and the circulation that are involved in thrombo-inflammatory processes. All these cells express NF-κB, which mediates important functions in cellular interactions, cell survival and differentiation, as well as expression of cytokines, chemokines, and coagulation factors. Even platelets, as anucleated cells, contain NF-κB family members and their corresponding signaling molecules, which are involved in platelet activation, as well as secondary feedback circuits. The response of endothelial cells to inflammation and NF-κB activation is characterized by the induction of adhesion molecules promoting binding and transmigration of leukocytes, while simultaneously increasing their thrombogenic potential. Paracrine signaling from endothelial cells activates NF-κB in vascular smooth muscle cells and causes a phenotypic switch to a “synthetic” state associated with a decrease in contractile proteins. Monocytes react to inflammatory situations with enforced expression of tissue factor and after differentiation to macrophages with altered polarization. Neutrophils respond with an extension of their life span—and upon full activation they can expel their DNA thereby forming so-called neutrophil extracellular traps (NETs), which exert antibacterial functions, but also induce a strong coagulatory response. This may cause formation of microthrombi that are important for the immobilization of pathogens, a process designated as immunothrombosis. However, deregulation of the complex cellular links between inflammation and thrombosis by unrestrained NET formation or the loss of the endothelial layer due to mechanical rupture or erosion can result in rapid activation and aggregation of platelets and the manifestation of thrombo-inflammatory diseases. Sepsis is an important example of such a disorder caused by a dysregulated host response to infection finally leading to severe coagulopathies. NF-κB is critically involved in these pathophysiological processes as it induces both inflammatory and thrombotic responses.
Collapse
Affiliation(s)
- Marion Mussbacher
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Manuel Salzmann
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Christine Brostjan
- Department of Surgery, General Hospital, Medical University of Vienna, Vienna, Austria
| | - Bastian Hoesel
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | | | - Hannes Datler
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Philipp Hohensinner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - José Basílio
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Peter Petzelbauer
- Skin and Endothelial Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Johannes A Schmid
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
19
|
Ling Y, Li ZZ, Zhang JF, Zheng XW, Lei ZQ, Chen RY, Feng JH. RETRACTED: MicroRNA-494 inhibition alleviates acute lung injury through Nrf2 signaling pathway via NQO1 in sepsis-associated acute respiratory distress syndrome. Life Sci 2018; 210:1-8. [PMID: 30121199 PMCID: PMC9673760 DOI: 10.1016/j.lfs.2018.08.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 11/24/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figures 5G,H+I, which appear to have a similar phenotype as many other publications, as detailed here: https://pubpeer.com/publications/7C9483B2551952AD53CCFCE206C4EB; and here: https://docs.google.com/spreadsheets/d/1r0MyIYpagBc58BRF9c3luWNlCX8VUvUuPyYYXzxWvgY/edit#gid=262337249. The journal requested that the corresponding author comment on these concerns and provide the raw data. The authors did not respond to this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Yun Ling
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| | - Zheng-Zhao Li
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| | - Jian-Feng Zhang
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China.
| | - Xiao-Wen Zheng
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| | - Zhuo-Qing Lei
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| | - Ru-Yan Chen
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| | - Ji-Hua Feng
- Department of Emergency, the Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, PR China
| |
Collapse
|
20
|
Anan K, Kawamura K, Suga M, Ichikado K. Clinical differences between pulmonary and extrapulmonary acute respiratory distress syndrome: a retrospective cohort study of prospectively collected data in Japan. J Thorac Dis 2018; 10:5796-5803. [PMID: 30505487 DOI: 10.21037/jtd.2018.09.73] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Although acute respiratory distress syndrome (ARDS) reportedly shows various clinical phenotypes with different risk and prognostic factors, few studies have assessed the clinical features and prognosis of pulmonary and extrapulmonary ARDS. The aim of the present study was to investigate clinical differences between pulmonary and extrapulmonary ARDS. Methods In total, 200 patients who met the Berlin criteria and were diagnosed with ARDS between October 2004 and September 2017 were included. We classified the patients into pulmonary and extrapulmonary ARDS groups. Both groups were assessed for 60-day mortality, duration of ventilation, and other clinical features. Results There were 150 and 50 patients in the pulmonary and extrapulmonary ARDS groups, respectively. The two groups showed no significant differences in any assessment parameters except the serum lactate dehydrogenase (LDH) level, which was higher in the extrapulmonary ARDS group (P=0.01). After adjustment for potentially confounding covariates, there were no significant differences in 60-day mortality (P=0.99) and the duration of ventilation (P=0.45) between the two groups. Mortality was significantly associated with the disseminated intravascular coagulation (DIC) score, high-resolution computed tomography (HRCT) score, and serum LDH level in the pulmonary ARDS group and the DIC score and HRCT score in the extrapulmonary ARDS group. Conclusions Pulmonary and extrapulmonary ARDS may be comparable in terms of the prognosis and duration of ventilation. DIC and HRCT scores may be common clinical predictors of mortality with ARDS.
Collapse
Affiliation(s)
- Keisuke Anan
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Kodai Kawamura
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Moritaka Suga
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Kazuya Ichikado
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| |
Collapse
|
21
|
Kawamura K, Ichikado K, Takaki M, Eguchi Y, Anan K, Suga M. Adjunctive therapy with azithromycin for moderate and severe acute respiratory distress syndrome: a retrospective, propensity score-matching analysis of prospectively collected data at a single center. Int J Antimicrob Agents 2018; 51:918-924. [PMID: 29501821 DOI: 10.1016/j.ijantimicag.2018.02.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/10/2018] [Accepted: 02/14/2018] [Indexed: 01/22/2023]
Abstract
Effective pharmacological therapy has not been established for patients with acute respiratory distress syndrome (ARDS). Macrolides are antibiotics with potent immunomodulatory and anti-inflammatory effects that may be beneficial in ARDS treatment. The objective of this study was to determine the adjunctive effect of azithromycin on survival for patients with ARDS. This single-center, retrospective cohort evaluation of hospitalized patients with moderate or severe ARDS was conducted to assess the impact of intravenous azithromycin on clinical outcomes using a propensity score analysis. All data were collected prospectively as part of ongoing research on the utility of high-resolution computed tomography in ARDS. The primary outcome was 90-day mortality, and the secondary analysis assessed the effect of azithromycin on time to successful discontinuation of mechanical ventilation and 28-day mortality. Of 191 eligible patients with severe or moderate ARDS, 62 were treated with azithromycin. The 62 patients treated with azithromycin and 62 not treated with azithromycin were matched and analysed. Azithromycin use was associated with a statistically significant improvement in 90-day survival rate (Hazard ratio [HR], 0.49; 95% confidence interval [CI], 0.27-0.87; P = 0.015) and a shorter time to successful discontinuation of mechanical ventilation (HR, 1.74; 95% CI, 1.07-2.81; P = 0.026). The 28-day mortality rate tended to be higher in the azithromycin cohort than in the non-azithromycin cohort, but this was not statistically significant. Adjunctive intravenous azithromycin therapy was effective in patients with moderate or severe ARDS. Further prospective randomized controlled trials are needed to confirm this result.
Collapse
Affiliation(s)
- Kodai Kawamura
- Division of Respiratory Medicine, Social Welfare Organization Saiseikai Imperial Gift Foundation, Inc., Saiseikai Kumamoto Hospital, Kumamoto 861-4193, Japan.
| | - Kazuya Ichikado
- Division of Respiratory Medicine, Social Welfare Organization Saiseikai Imperial Gift Foundation, Inc., Saiseikai Kumamoto Hospital, Kumamoto 861-4193, Japan
| | - Makoto Takaki
- Division of Respiratory Medicine, Social Welfare Organization Saiseikai Imperial Gift Foundation, Inc., Saiseikai Kumamoto Hospital, Kumamoto 861-4193, Japan
| | - Yoshitomo Eguchi
- Division of Respiratory Medicine, Social Welfare Organization Saiseikai Imperial Gift Foundation, Inc., Saiseikai Kumamoto Hospital, Kumamoto 861-4193, Japan
| | - Keisuke Anan
- Division of Respiratory Medicine, Social Welfare Organization Saiseikai Imperial Gift Foundation, Inc., Saiseikai Kumamoto Hospital, Kumamoto 861-4193, Japan
| | - Moritaka Suga
- Division of Respiratory Medicine, Social Welfare Organization Saiseikai Imperial Gift Foundation, Inc., Saiseikai Kumamoto Hospital, Kumamoto 861-4193, Japan
| |
Collapse
|
22
|
Hussain M, Xu C, Ahmad M, Majeed A, Lu M, Wu X, Tang L, Wu X. Acute Respiratory Distress Syndrome: Bench-to-Bedside Approaches to Improve Drug Development. Clin Pharmacol Ther 2018; 104:484-494. [PMID: 29484641 PMCID: PMC7162218 DOI: 10.1002/cpt.1034] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/11/2018] [Accepted: 01/21/2018] [Indexed: 12/20/2022]
Abstract
Despite 50 years of extensive research, no definite drug is currently available to treat acute respiratory distress syndrome (ARDS), and the supportive therapies remain the mainstay of treatment. To improve drug development for ARDS, researchers need to deeply analyze the “omics” approaches, reevaluate the suitable therapeutic targets, resolve the problems of inadequate animal modeling, develop the strategies to reduce the heterogeneity, and reconsider new therapeutic and analytical approaches for better designs of clinical trials.
Collapse
Affiliation(s)
- Musaddique Hussain
- Department of Pharmacology, Hangzhou City, 310058, China.,The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City, 310058, China
| | - Chengyun Xu
- Department of Pharmacology, Hangzhou City, 310058, China.,The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City, 310058, China
| | - Mashaal Ahmad
- Department of Pharmacology, Hangzhou City, 310058, China.,The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City, 310058, China
| | - Abdul Majeed
- Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Meiping Lu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City, 310006, China
| | - Xiling Wu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City, 310006, China
| | - Lanfang Tang
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City, 310006, China
| | - Ximei Wu
- Department of Pharmacology, Hangzhou City, 310058, China.,The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City, 310058, China
| |
Collapse
|
23
|
Simonis FD, de Iudicibus G, Cremer OL, Ong DSY, van der Poll T, Bos LD, Schultz MJ. Macrolide therapy is associated with reduced mortality in acute respiratory distress syndrome (ARDS) patients. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:24. [PMID: 29430441 DOI: 10.21037/atm.2017.12.25] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Background Macrolides have been associated with favorable immunological effects in various inflammatory disease states. We investigated the association between macrolide therapy and mortality in patients with the acute respiratory distress syndrome (ARDS). Methods This was an unplanned secondary analysis of patients with ARDS within a large prospective observational study of critically ill patients in the intensive care units (ICUs) of two university-affiliated hospitals in the Netherlands. The exposure of interest was low-dose macrolide use prescribed for another reason than infection; we excluded patients who received high-dose macrolides for an infection. The primary endpoint was 30-day mortality. The association between macrolide therapy and mortality was determined in the whole cohort, as well as in a propensity score matched cohort; the association was compared between pulmonary versus non-pulmonary ARDS, and between two biological phenotypes based on plasma levels of 20 biomarkers. Results In total, 873 patients with ARDS were analyzed, of whom 158 patients (18%) received macrolide therapy during stay in ICU for a median duration of 3 (interquartile range, 1-4) days. Erythromycin was the most frequent prescribed macrolide (97%). Macrolide therapy was associated with reduced 30-day mortality in the whole cohort [22.8% vs. 31.6%; crude odds ratio (OR), 0.64 (interquartile range, 0.43-0.96), P=0.03]. The association in the propensity score matched cohort remained significant [22.8% vs. 32.9%; OR, 0.62 (interquartile range, 0.39-0.96), P=0.03]. Propensity matched associations with mortality were different in patients with non-pulmonary ARDS vs. pulmonary ARDS and also varied by biological phenotype. Conclusions These data together show that low-dose macrolide therapy prescribed for another reason than infection is associated with decreased mortality in patients with ARDS.
Collapse
Affiliation(s)
- Fabienne D Simonis
- Department of Intensive Care, Academic Medical Center, Amsterdam, The Netherlands.,Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Academic Medical Center, Amsterdam, The Netherlands
| | | | - Olaf L Cremer
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - David S Y Ong
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Medical Microbiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine (C.E.M.M.), Academic Medical Center, Amsterdam, The Netherlands.,Division of Infectious Diseases, Academic Medical Center, Amsterdam, The Netherlands
| | - Lieuwe D Bos
- Department of Intensive Care, Academic Medical Center, Amsterdam, The Netherlands.,Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Academic Medical Center, Amsterdam, The Netherlands.,Department of Pulmonology, Academic Medical Center, Amsterdam, The Netherlands
| | - Marcus J Schultz
- Department of Intensive Care, Academic Medical Center, Amsterdam, The Netherlands.,Laboratory for Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Academic Medical Center, Amsterdam, The Netherlands.,Mahidol Oxford Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | |
Collapse
|
24
|
Anan K, Ichikado K, Kawamura K, Johkoh T, Fujimoto K, Suga M. Clinical characteristics and prognosis of drug-associated acute respiratory distress syndrome compared with non-drug-associated acute respiratory distress syndrome: a single-centre retrospective study in Japan. BMJ Open 2017; 7:e015330. [PMID: 29122783 PMCID: PMC5695405 DOI: 10.1136/bmjopen-2016-015330] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES To report the clinical features and prognosis of drug-associatedacute respiratory distress syndrome (ARDS). DESIGN A retrospective analysis of data collected during a prospective cohort study. SETTING Intensive care unit in a teaching hospital. PARTICIPANTS A total of 197 Japanese patients with ARDS diagnosed by the Berlin definition who were admitted to the Division of Respiratory Medicine from October 2004 to December 2015 were enrolled in the study and were classified as two groups according to their causes: a drug-associated ARDS group (n=27) and a non-drug-associated ARDS group (n=170). Primary outcome measure is 28-day mortality, and the secondaryoutcome measure is ventilator-free days. RESULTS The Acute Physiology and Chronic Health Evaluation II scores were significantly lower in the drug-associated ARDS group than in the non-drug-associated ARDS group (median (IQR): 18.0 (16.5-21.0) vs 23.0 (18.0-26.0), p<0.001), and the arterial oxygen tension/fractional inspired oxygen ratio was higher (148.0 (114.1-177.5) vs 101.0 (71.5-134.0), p=0.003). In the drug-associated ARDS group, although high-resolution CT scores indicative of the extent of fibroproliferation (301.6 (244.1-339.8) vs 208.3 (183.4-271.6), p<0.001), serum lactate dehydrogenase levels (477 (365-585) vs 322 (246-434), p=0.003) and the McCabe scores (score 1/2/3, n (%): 20 (74)/4 (15)/3 (11)vs154 (91)/7 (4)/9 (5), p=0.04) were significantly higher, ventilator weaning was earlier (p<0.001) and 28-day mortality was better (p=0.043). After adjusting for potentially confounding covariates, drug-associated ARDS group was associated with lower 28-day mortality (adjusted HR (HR) 0.275; 95% CI 0.106 to 0.711; p=0.008). CONCLUSIONS Although more severe lung damage with fibroproliferation was observed in patients with drug-associated ARDS, ventilator weaning was earlier, and their prognosis was better than the others. Further well-designed prospective studies are needed.
Collapse
Affiliation(s)
- Keisuke Anan
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Kazuya Ichikado
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Kodai Kawamura
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Takeshi Johkoh
- Department of Radiology, Kiniki Central Hospital of Mutual Aid Association of Public School Teachers, Itami, Hyogo, Japan
| | - Kiminori Fujimoto
- Department of Radiology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
- Center for Diagnostic Imaging, Kurume University Hospital, Kurume, Fukuoka, Japan
| | - Moritaka Suga
- Division of Respiratory Medicine, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| |
Collapse
|
25
|
Kawamura K, Ichikado K, Yasuda Y, Anan K, Suga M. Azithromycin for idiopathic acute exacerbation of idiopathic pulmonary fibrosis: a retrospective single-center study. BMC Pulm Med 2017. [PMID: 28629448 PMCID: PMC5477160 DOI: 10.1186/s12890-017-0437-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background Acute exacerbation (AE) of idiopathic pulmonary fibrosis (IPF) is a fatal condition without an established pharmaceutical treatment. Most patients are treated with high-dose corticosteroids and broad-spectrum antibiotics. Azithromycin is a macrolide with immunomodulatory activity and may be beneficial for treatment of acute lung injury. The objective of this study was to determine the effect of azithromycin on survival of patients with idiopathic AE of IPF. Methods We evaluated 85 consecutive patients hospitalized in our department for idiopathic AE of IPF from April 2005 to August 2016. The initial 47 patients were treated with a fluoroquinolone-based regimen (control group), and the following 38 consecutive patients were treated with azithromycin (500 mg/day) for 5 days. Idiopathic AE of IPF was defined using the criteria established by the 2016 International Working Group. Results Mortality in patients treated with azithromycin was significantly lower than in those treated with fluoroquinolones (azithromycin, 26% vs. control, 70%; p < 0.001). Multivariate analysis revealed that the two variables were independently correlated with 60-day mortality as determined by the Acute Physiology and Chronic Health Evaluation II score (p = 0.002) and azithromycin use (p < 0.001). Conclusion Azithromycin may improve survival in patients with idiopathic AE of IPF.
Collapse
Affiliation(s)
- Kodai Kawamura
- Division of Respiratory Medicine, Social Welfare Organization Saiseikai Imperial Gift Foundation, Inc., Saiseikai Kumamoto Hospital, Kumamoto, Kumamoto, 861-4193, Japan.
| | - Kazuya Ichikado
- Division of Respiratory Medicine, Social Welfare Organization Saiseikai Imperial Gift Foundation, Inc., Saiseikai Kumamoto Hospital, Kumamoto, Kumamoto, 861-4193, Japan
| | - Yuko Yasuda
- Division of Respiratory Medicine, Social Welfare Organization Saiseikai Imperial Gift Foundation, Inc., Saiseikai Kumamoto Hospital, Kumamoto, Kumamoto, 861-4193, Japan
| | - Keisuke Anan
- Division of Respiratory Medicine, Social Welfare Organization Saiseikai Imperial Gift Foundation, Inc., Saiseikai Kumamoto Hospital, Kumamoto, Kumamoto, 861-4193, Japan
| | - Moritaka Suga
- Division of Respiratory Medicine, Social Welfare Organization Saiseikai Imperial Gift Foundation, Inc., Saiseikai Kumamoto Hospital, Kumamoto, Kumamoto, 861-4193, Japan
| |
Collapse
|
26
|
Cramer CL, Patterson A, Alchakaki A, Soubani AO. Immunomodulatory indications of azithromycin in respiratory disease: a concise review for the clinician. Postgrad Med 2017; 129:493-499. [PMID: 28116959 DOI: 10.1080/00325481.2017.1285677] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Azithromycin has a well-characterized bacteriostatic activity. However, it also has a robust immunomodulatory effect that has proven beneficial in a variety of chronic illnesses. This effect results in decreased production of pro-inflammatory cytokines in the acute phase and promotes resolution of chronic inflammation in the later phases. Specifically, azithromycin has direct activity on airway epithelial cells to maintain their function and reduce mucus secretion. These characteristics have resulted in the use of azithromycin in the management of a variety of chronic lung diseases including chronic obstructive pulmonary disease, cystic fibrosis (CF), non-CF bronchiectasis, bronchiolitis obliterans syndrome, diffuse panbronchiolitis, and asthma. In this review, we present the evidence supporting the role of azithromycin in these conditions with an emphasis on the clinical aspects for the practicing physician.
Collapse
Affiliation(s)
- Cassondra L Cramer
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| | - Allie Patterson
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| | - Abdulrazak Alchakaki
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| | - Ayman O Soubani
- a Division of Pulmonary, Critical Care and Sleep Medicine , Wayne State University School of Medicine , Detroit , MI , USA
| |
Collapse
|