1
|
Shao XT, Wang YS, Gong ZF, Li YY, Lin JG, Wang DG. A feasibility study on cortisol and cortisone as biomarkers for psychological stress in wastewater-based epidemiology. WATER RESEARCH 2025; 273:123022. [PMID: 39742636 DOI: 10.1016/j.watres.2024.123022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/03/2025]
Abstract
Psychological stress has a significant impact on individuals' quality of life and health. Traditionally, psychological stress assessment relies on self-reported tools such as the Perceived Stress Scale (PSS), which are inherently subjective. This study aims to evaluate the feasibility of using wastewater-based epidemiology (WBE) to assess cortisol and cortisone as biomarkers for psychological stress. We conducted sampling and monitoring of cortisol and cortisone concentrations at both a small-scale campus setting (five weeks) and a large-scale municipal wastewater treatment plant (12 months), calculating the mass loads of these hormones. At the campus level, while the mass load of cortisone was higher during exam weeks compared to regular class weeks, and higher in females than in males, no significant differences were observed in the mass load of cortisol. The mass load results of cortisone were consistent with the findings of the PSS-14 questionnaire. These results suggest that cortisone is a more suitable biomarker for psychological stress assessment. In the large-scale municipal wastewater samples, seasonal variations were observed, with higher levels of cortisol and cortisone in winter compared to summer, likely due to the COVID-19 outbreak in winter and the presence of external pharmaceutical sources. The results indicate that cortisone is more suitable for small-scale stress assessments, as larger-scale evaluations may be more significantly influenced by wastewater transport or sampling methodologies.
Collapse
Affiliation(s)
- Xue-Ting Shao
- College of Environmental Science and Engineering, Dalian Maritime University, No. 1 Linghai Road, Dalian, , 116026, China
| | - Yan-Song Wang
- College of Environmental Science and Engineering, Dalian Maritime University, No. 1 Linghai Road, Dalian, , 116026, China
| | - Zhen-Fang Gong
- College of Environmental Science and Engineering, Dalian Maritime University, No. 1 Linghai Road, Dalian, , 116026, China
| | - Yan-Ying Li
- College of Environmental Science and Engineering, Dalian Maritime University, No. 1 Linghai Road, Dalian, , 116026, China
| | - Jian-Guo Lin
- College of Environmental Science and Engineering, Dalian Maritime University, No. 1 Linghai Road, Dalian, , 116026, China
| | - De-Gao Wang
- College of Environmental Science and Engineering, Dalian Maritime University, No. 1 Linghai Road, Dalian, , 116026, China.
| |
Collapse
|
2
|
Bansal V, Jain NK, Lal A, Khedr A, Tekin A, Jama AB, Attallah N, Hassan E, Mushtaq HA, Robinson S, Kondori MJ, Koritala T, Armaignac DL, Christie AB, Raju U, Khanna A, Cartin-Ceba R, Sanghavi DK, La Nou A, Boman K, Kumar V, Walkey AJ, Domecq JP, Kashyap R, Khan SA. The association between early corticosteroid use and the risk of secondary infections in hospitalized patients with COVID-19: a double-edged sword. Results from the international SCCM discovery viral infection and respiratory illness universal study (VIRUS) COVID-19 registry. Front Med (Lausanne) 2025; 12:1466346. [PMID: 40027890 PMCID: PMC11868930 DOI: 10.3389/fmed.2025.1466346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/15/2025] [Indexed: 03/05/2025] Open
Abstract
Background Corticosteroids improve survival in hospitalized COVID-19 patients needing supplemental oxygen. However, concern exists about increased risk of secondary infections. This study investigated the impact of early corticosteroids use on these infections. Methods Data from the Society of Critical Care Medicine Discovery Viral Infection and Respiratory Illness Universal Study (VIRUS): COVID-19 registry were analyzed for adult patients, stratified by early corticosteroid use (within 48 h of admission). The primary outcome was documented secondary infections, including bacteremia, bacterial pneumonia, empyema, meningitis/encephalitis, septic shock, and ventilator-associated pneumonia. Univariate and multivariable logistic regression models were used to assess the association between early corticosteroids and these outcomes. Results Among 17,092 eligible patients, with 13.5% developed at least one secondary bacterial infection during hospitalization. Patients receiving early corticosteroids were older (median 63 years) compared to those who did not (median 60 years), with a similar gender distribution (42.5% vs. 44.2% female). Unadjusted analysis revealed a higher risk for any secondary infection (OR 1.93, 95% CI 1.76-2.12). This association persisted for specific infections including bacteremia (OR 2.0, 95% CI 1.58-2.54), bacterial pneumonia (OR 1.5, 95% CI 1.27-1.77), and septic shock (OR 1.67, 95% CI 1.44-1.93). However, the effect on meningitis/encephalitis (OR 0.62, 95% CI 0.24-1.57) and ventilator-associated pneumonia (VAP; OR 1.08, 95% CI 0.75-1.57) was non-significant. Adjusted analysis maintained significance for any secondary infection (OR 1.15, 95% CI 1.02-1.29), bacteremia (OR 1.43, 95% CI 1.09-1.88), and infections with unknown sources (OR 1.63, 95% CI 1.31-2.02). Notably, the association weakened and became non-significant for bacterial pneumonia (OR 0.98, 95% CI 0.81-1.20) and septic shock (OR 0.94, 95% CI 0.79-1.11), while it became significant for meningitis/encephalitis (OR 0.26, 95% CI 0.08-0.82). VAP remained non-significant (OR 0.87, 95% CI 0.56-1.34). Conclusion Early use of corticosteroids increased overall secondary infection risk in hospitalized COVID-19 patients, but the impact varied. Risk of bacteremia was notably increased, while the association with bacterial pneumonia and septic shock weakened after adjustment becoming non-significant and surprisingly reduced meningitis/encephalitis risk was noted suggesting the complexity of corticosteroid effects. Further research is needed to understand how corticosteroids influence specific secondary infections, and thereby optimize the treatment strategies.
Collapse
Affiliation(s)
- Vikas Bansal
- Division of Nephrology and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Nitesh K. Jain
- Department of Critical Care Medicine, Mayo Clinic Health System, Mankato, MN, United States
- Department of Critical Care Medicine, North East Georgia Health System, Gainesville, GA, United States
| | - Amos Lal
- Department of Critical Care Medicine, North East Georgia Health System, Gainesville, GA, United States
| | - Anwar Khedr
- Department of Critical Care Medicine, Mayo Clinic Health System, Mankato, MN, United States
| | - Aysun Tekin
- Division of Nephrology and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Abbas B. Jama
- Department of Critical Care Medicine, Mayo Clinic Health System, Mankato, MN, United States
| | - Noura Attallah
- Department of Critical Care Medicine, Mayo Clinic Health System, Mankato, MN, United States
| | - Esraa Hassan
- Department of Critical Care Medicine, Mayo Clinic Health System, Mankato, MN, United States
| | - Hisham Ahmed Mushtaq
- Department of Critical Care Medicine, Mayo Clinic Health System, Mankato, MN, United States
| | - Sara Robinson
- Department of Family Medicine, Mayo Clinic Health System, Mankato, MN, United States
| | - Marjan Jahani Kondori
- Department of Family Medicine, Mayo Clinic Health System, Mankato, MN, United States
| | - Thoyaja Koritala
- Department of Critical Care Medicine, Mayo Clinic Health System, Mankato, MN, United States
| | - Donna Lee Armaignac
- Center for Advanced Analytics, Baptist Health South Florida, Miami, FL, United States
| | - Amy B. Christie
- Department of Trauma Critical Care, The Medical Center Navicent Health, Mercer University School of Medicine, Macon, GA, United States
| | | | - Ashish Khanna
- Section on Critical Care Medicine, Department of Anesthesiology, Wake Forest University School of Medicine, Atrium Health Wake Forest Baptist Medical Center, Winston-Salem, NC, United States
| | - Rodrigo Cartin-Ceba
- Division of Critical Care Medicine, Department of Pulmonary Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Devang K. Sanghavi
- Department of Critical Care Medicine, Mayo Clinic Florida, Jacksonville, FL, United States
| | - Abigail La Nou
- Department of Critical Care Medicine, Mayo Clinic Health System, Eau Claire, WI, United States
| | - Karen Boman
- Society of Critical Care Medicine, Mount Prospect, IL, United States
| | - Vishakha Kumar
- Society of Critical Care Medicine, Mount Prospect, IL, United States
| | - Allan J. Walkey
- Pulmonary Center, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Juan Pablo Domecq
- Division of Nephrology and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
- Department of Critical Care Medicine, Mayo Clinic Health System, Mankato, MN, United States
| | - Rahul Kashyap
- Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Care, Mayo Clinic, Rochester, MN, United States
- Department of Medicine and Medical, Research, WellSpan Health, York, PA, United States
| | - Syed Anjum Khan
- Department of Critical Care Medicine, Mayo Clinic Health System, Mankato, MN, United States
| | | |
Collapse
|
3
|
Eldien HMS, Almaeen AH, El Fath AA, Taha AE, Ahmed R, Elfadil H, Hetta HF. Unlocking the Potential of RNA Sequencing in COVID-19: Toward Accurate Diagnosis and Personalized Medicine. Diagnostics (Basel) 2025; 15:229. [PMID: 39857114 PMCID: PMC11763845 DOI: 10.3390/diagnostics15020229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/08/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
COVID-19 has caused widespread morbidity and mortality, with its effects extending to multiple organ systems. Despite known risk factors for severe disease, including advanced age and underlying comorbidities, patient outcomes can vary significantly. This variability complicates efforts to predict disease progression and tailor treatment strategies. While diagnostic and therapeutic approaches are still under debate, RNA sequencing (RNAseq) has emerged as a promising tool to provide deeper insights into the pathophysiology of COVID-19 and guide personalized treatment. A comprehensive literature review was conducted using PubMed, Scopus, Web of Science, and Google Scholar. We employed Medical Subject Headings (MeSH) terms and relevant keywords to identify studies that explored the role of RNAseq in COVID-19 diagnostics, prognostics, and therapeutics. RNAseq has proven instrumental in identifying molecular biomarkers associated with disease severity in patients with COVID-19. It allows for the differentiation between asymptomatic and symptomatic individuals and sheds light on the immune response mechanisms that contribute to disease progression. In critically ill patients, RNAseq has been crucial for identifying key genes that may predict patient outcomes, guiding therapeutic decisions, and assessing the long-term effects of the virus. Additionally, RNAseq has helped in understanding the persistence of viral RNA after recovery, offering new insights into the management of post-acute sequelae, including long COVID. RNA sequencing significantly improves COVID-19 management, particularly for critically ill patients, by enhancing diagnostic accuracy, personalizing treatment, and predicting therapeutic responses. It refines patient stratification, improving outcomes, and holds promise for targeted interventions in both acute and long COVID.
Collapse
Affiliation(s)
- Heba M. Saad Eldien
- Department of Anatomy, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia
| | - Abdulrahman H. Almaeen
- Department of Pathology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia;
| | - Ahmed Abo El Fath
- Tropical Medicine and Gastroenterology Department, Assiut University Hospital, Assiut 71515, Egypt;
| | - Ahmed E. Taha
- Microbiology and Immunology Unit, Department of Pathology, College of Medicine, Jouf University, Sakaka 72388, Saudi Arabia;
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Rehab Ahmed
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| | - Hassabelrasoul Elfadil
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| | - Helal F. Hetta
- Division of Microbiology, Immunology and Biotechnology, Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia; (R.A.); (H.E.)
| |
Collapse
|
4
|
Hu H, Wei S, Huang J, Sharma L, Chang D. Beneficial effects of early low-dose methylprednisolone with long-term treatment in ARDS. Eur J Intern Med 2024; 129:137-139. [PMID: 38879352 DOI: 10.1016/j.ejim.2024.06.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 11/05/2024]
Affiliation(s)
- Haiming Hu
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing, 100853, PR China
| | - Shuting Wei
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100037, PR China
| | - Jiayin Huang
- Department of Microbiology, Army Medical University (the Third Military Medical University), Chongqing 400037, PR China
| | - Lokesh Sharma
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, PA 15213, USA
| | - De Chang
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing, 100853, PR China.
| |
Collapse
|
5
|
Xu J, Li Y, Yang X, Li H, Xiao X, You J, Li H, Zheng L, Yi C, Li Z, Huang Y. Quercetin inhibited LPS-induced cytokine storm by interacting with the AKT1-FoxO1 and Keap1-Nrf2 signaling pathway in macrophages. Sci Rep 2024; 14:20913. [PMID: 39245773 PMCID: PMC11381534 DOI: 10.1038/s41598-024-71569-y] [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: 05/10/2024] [Accepted: 08/29/2024] [Indexed: 09/10/2024] Open
Abstract
Cytokine storm (CS) emerges as an exacerbated inflammatory response triggered by various factors such as pathogens and excessive immunotherapy, posing a significant threat to life if left unchecked. Quercetin, a monomer found in traditional Chinese medicine, exhibits notable anti-inflammatory and antiviral properties. This study endeavors to explore whether quercetin intervention could mitigate CS through a combination of network pharmacology analysis and experimental validation. First, common target genes and potential mechanisms affected by quercetin and CS were identified through network pharmacology, and molecular docking experiments confirmed quercetin and core targets. Subsequently, in vitro experiments of Raw264.7 cells stimulated by lipopolysaccharide (LPS) showed that quercetin could effectively inhibit the overexpression of pro-inflammatory mediators and regulate the AKT1-FoxO1 signaling pathway. At the same time, quercetin can reduce ROS through the Keap1-Nrf2 signaling pathway. In addition, in vivo studies of C57BL/6 mice injected with LPS further confirmed quercetin's inhibitory effect on CS. In conclusion, this investigation elucidated novel target genes and signaling pathways implicated in the therapeutic effects of quercetin on CS. Moreover, it provided compelling evidence supporting the efficacy of quercetin in reversing LPS-induced CS, primarily through the regulation of the AKT1-FoxO1 and Keap1-Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Jingyi Xu
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, No.17, Section3, Renmin South Road, Chengdu, 610044, People's Republic of China
| | - Yue Li
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, No.17, Section3, Renmin South Road, Chengdu, 610044, People's Republic of China
| | - Xi Yang
- Department of Medical Oncology, West China Hospital, Cancer Center, Sichuan University, No.37 Guoxue Lane, Chengdu, 610041, China
| | - Hong Li
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, No.17, Section3, Renmin South Road, Chengdu, 610044, People's Republic of China
| | - Xi Xiao
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, No.17, Section3, Renmin South Road, Chengdu, 610044, People's Republic of China
| | - Jia You
- Department of Medical Oncology, West China Hospital, Cancer Center, Sichuan University, No.37 Guoxue Lane, Chengdu, 610041, China
| | - Huawei Li
- Department of Integrated Traditional Chinese and Western Medicine, School of Medicine, Cancer Hospital, University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Lingnan Zheng
- Department of Medical Oncology, West China Hospital, Cancer Center, Sichuan University, No.37 Guoxue Lane, Chengdu, 610041, China
| | - Cheng Yi
- Department of Medical Oncology, West China Hospital, Cancer Center, Sichuan University, No.37 Guoxue Lane, Chengdu, 610041, China.
| | - Zhaojun Li
- Department of Radiation Oncology, Hainan Affiliated Hospital of Hainan Medical University (Hainan General Hospital), No.17, Xiuhua Road, Haikou, 570100, China.
| | - Ying Huang
- West China School of Basic Medical Science and Forensic Medicine, Sichuan University, No.17, Section3, Renmin South Road, Chengdu, 610044, People's Republic of China.
| |
Collapse
|
6
|
Navas Boada P, Chamorro K, Ballaz S. Survival analysis of COVID-19 versus non-COVID-19 patients requiring intensive care for acute respiratory distress syndrome: An observational retrospective study. CANADIAN JOURNAL OF RESPIRATORY THERAPY : CJRT = REVUE CANADIENNE DE LA THERAPIE RESPIRATOIRE : RCTR 2024; 60:112-121. [PMID: 39268526 PMCID: PMC11392457 DOI: 10.29390/001c.122402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/07/2024] [Indexed: 09/15/2024]
Abstract
Background/Aim This study analyzed clinical factors impacting the survival of COVID-19 patients with acute respiratory distress síndrome, or ARDS (CARDS) to ICU compared to non-COVID-19 ARDS patients. Methods Clinical variables from 1,008 CARDS cases and 332 ARDS cases were computed using learning algorithms. The multivariable Cox proportional hazards regression models with the enter method evaluated risk factors and ICU mortality relationships. The survival analysis was completed with Kaplan-Meier and the log-rank tests. Results A Random Forest model revealed that mechanical ventilation-related factors, oxygenation, blood pH, superinfection, shock, and ICU length of stay have the greatest effects on ICU survival. According to a multivariate Cox model, reintubation and a high-flow nasal cannula were essential for survival in CARDS patients during the ICU stay. The length of stay in the ICU diminishes in patients older than 45 years, regardless of the source of ARDS. Conclusion This study gives recommendations for the respiratory care of ARDS in COVID-19 patients.
Collapse
Affiliation(s)
- Paulo Navas Boada
- School of Biological Sciences and Engineering Universidad Yachay Tech
| | - Kevin Chamorro
- School of Mathematics and Computational Sciences Universidad Yachay Tech
| | | |
Collapse
|
7
|
Yu Q, Zhou X, Kapini R, Arsecularatne A, Song W, Li C, Liu Y, Ren J, Münch G, Liu J, Chang D. Cytokine Storm in COVID-19: Insight into Pathological Mechanisms and Therapeutic Benefits of Chinese Herbal Medicines. MEDICINES (BASEL, SWITZERLAND) 2024; 11:14. [PMID: 39051370 PMCID: PMC11270433 DOI: 10.3390/medicines11070014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/20/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024]
Abstract
Cytokine storm (CS) is the main driver of SARS-CoV-2-induced acute respiratory distress syndrome (ARDS) in severe coronavirus disease-19 (COVID-19). The pathological mechanisms of CS are quite complex and involve multiple critical molecular targets that turn self-limited and mild COVID-19 into a severe and life-threatening concern. At present, vaccines are strongly recommended as safe and effective treatments for preventing serious illness or death from COVID-19. However, effective treatment options are still lacking for people who are at the most risk or hospitalized with severe disease. Chinese herbal medicines have been shown to improve the clinical outcomes of mild to severe COVID-19 as an adjunct therapy, particular preventing the development of mild to severe ARDS. This review illustrates in detail the pathogenesis of CS-involved ARDS and its associated key molecular targets, cytokines and signalling pathways. The therapeutic targets were identified particularly in relation to the turning points of the development of COVID-19, from mild symptoms to severe ARDS. Preclinical and clinical studies were reviewed for the effects of Chinese herbal medicines together with conventional therapies in reducing ARDS symptoms and addressing critical therapeutic targets associated with CS. Multiple herbal formulations, herbal extracts and single bioactive phytochemicals with or without conventional therapies demonstrated strong anti-CS effects through multiple mechanisms. However, evidence from larger, well-designed clinical trials is lacking and their detailed mechanisms of action are yet to be well elucidated. More research is warranted to further evaluate the therapeutic value of Chinese herbal medicine for CS in COVID-19-induced ARDS.
Collapse
Affiliation(s)
- Qingyuan Yu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
- Xiyuan Clinical Medical College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Rotina Kapini
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- School of Science, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Anthony Arsecularatne
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Wenting Song
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Chunguang Li
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Yang Liu
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| | - Junguo Ren
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Gerald Münch
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
- Pharmacology Unit, School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Jianxun Liu
- Beijing Key Laboratory of Pharmacology of Chinese Materia Region, Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (Q.Y.); (W.S.); (J.R.)
| | - Dennis Chang
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia; (X.Z.); (R.K.); (A.A.); (C.L.); (Y.L.); (G.M.)
| |
Collapse
|
8
|
Ye X, Li Y, Luo F, Xu Z, Kasimu K, Wang J, Xu P, Tan C, Yi H, Luo Y. Efficacy and safety of glucocorticoids in the treatment of COVID-19: a systematic review and meta-analysis of RCTs. Clin Exp Med 2024; 24:157. [PMID: 39003393 PMCID: PMC11246314 DOI: 10.1007/s10238-024-01405-0] [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: 04/17/2024] [Accepted: 06/14/2024] [Indexed: 07/15/2024]
Abstract
In the realm of acute respiratory infections, coronavirus disease-19 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), poses a global public health challenge. The application of corticosteroids (CSs) in COVID-19 remains a contentious topic among researchers. Accordingly, our team performed a comprehensive meta-analysis of randomized controlled trials (RCTs) to meticulously evaluate the safety and efficacy of CSs in hospitalized COVID-19 patients. To explore efficacy of CSs in the treatment of COVID-19 patients, we meticulously screened RCTs across key databases, including PubMed, Web of Science, Embase, Cochrane Library, ClinicalTrials.gov, as well as China's CNKI and Wanfang Data. We focused on assessing the 28 days mortality rates. We evaluated the data heterogeneity using the Chi-square test and I2 values, setting significance at 0.1 and 50%. Data from 21 RCTs involving 5721 participants were analyzed. The analysis did not demonstrate a significant association between CSs intervention and the 28 days mortality risk in hospitalized COVID-19 patients (relative risk [RR] = 0.93; 95% confidence interval [95% CI]: 0.84-1.03; P = 0.15). However, subgroup analysis revealed a significant reduction in 28 days mortality among patients with moderate-to-severe COVID-19 (RR at 0.85; 95% CI: 0.76-0.95; P = 0.004). Specifically, short-term CS administration (≤ 3 days) was associated with a substantial improvement in clinical outcomes (RR = 0.24; 95% CI: 0.09-0.63; P = 0.004), as was longer-term use (≥ 8 days) (RR = 0.88; 95% CI: 0.77-0.99; P = 0.04). Additionally, in patients with moderate-to-severe COVID-19, the administration of dexamethasone increased the number of 28 days ventilator-free days (Mean Difference = 1.92; 95% CI: 0.44-3.40; P = 0.01). Methylprednisolone also demonstrated significant benefits in improving clinical outcomes (RR = 0.24; 95% CI: 0.09-0.63; P = 0.004). Our meta-analysis demonstrated that although there is no significant difference in 28 days mortality rates among hospitalized COVID-19 patients, the use of CSs may be beneficial in improving clinical outcomes in moderate or severe COVID-19 patients. There was no significant increase in the occurrence of adverse events associated with the use of CSs. Our meta-analysis provides evidence that while CSs may not be suitable for all COVID-19 patients, they could be effective and safe in severely ill COVID-19 patients. Consequently, it is recommended to administer CSs for personalized treatments in COVID-19 cases to improve the clinical outcomes while minimizing adverse events.
Collapse
Affiliation(s)
- Xiangrong Ye
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
- Institute of Respiratory Diseases of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Ye Li
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
- Institute of Respiratory Diseases of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Feng Luo
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
- Institute of Respiratory Diseases of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Zhibin Xu
- Department of Organ Transplantation, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kaidirina Kasimu
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
- Institute of Respiratory Diseases of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Juan Wang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
- Institute of Respiratory Diseases of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Peihang Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chunjiang Tan
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
- Institute of Respiratory Diseases of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
| | - Hui Yi
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
- Institute of Respiratory Diseases of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
| | - Yifeng Luo
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
- Institute of Respiratory Diseases of Sun Yat-sen University, No. 58, Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
9
|
Monneret G, Voirin N, Richard JC, Cour M, Rimmelé T, Garnier L, Yonis H, Coudereau R, Gossez M, Malcus C, Wallet F, Delignette MC, Dailler F, Buisson M, Argaud L, Lukaszewicz AC, Venet F. Monitoring monocyte HLA-DR expression and CD4 + T lymphocyte count in dexamethasone-treated severe COVID-19 patients. Ann Intensive Care 2024; 14:76. [PMID: 38762684 PMCID: PMC11102415 DOI: 10.1186/s13613-024-01310-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/12/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND A 10-day dexamethasone regimen has emerged as the internationally adopted standard-of-care for severe COVID-19 patients. However, the immune response triggered by SARS-CoV-2 infection remains a complex and dynamic phenomenon, leading to various immune profiles and trajectories. The immune status of severe COVID-19 patients following complete dexamethasone treatment has yet to be thoroughly documented. RESULTS To analyze monocyte HLA-DR expression (mHLA-DR) and CD4 + T lymphocyte count (CD4) in critically ill COVID-19 patients after a dexamethasone course and evaluate their association with 28-day ICU mortality, adult COVID-19 patients (n = 176) with an ICU length of stay of at least 10 days and under dexamethasone treatment were included. Associations between each biomarker value (or in combination) measured at day 10 after ICU admission and 28-day mortality in ICU were evaluated. At day 10, the majority of patients presented decreased values of both parameters. A significant association between low mHLA-DR and 28-day mortality was observed. This association remained significant in a multivariate analysis including age, comorbidities or pre-existing immunosuppression (adjusted Hazard ratio (aHR) = 2.86 [1.30-6.32], p = 0.009). Similar results were obtained with decreased CD4 + T cell count (aHR = 2.10 [1.09-4.04], p = 0.027). When combining these biomarkers, patients with both decreased mHLA-DR and low CD4 presented with an independent and significant elevated risk of 28-day mortality (i.e., 60%, aHR = 4.83 (1.72-13.57), p = 0.001). CONCLUSIONS By using standardized immunomonitoring tools available in clinical practice, it is possible to identify a subgroup of patients at high risk of mortality at the end of a 10-day dexamethasone treatment. This emphasizes the significance of integrating immune monitoring into the surveillance of intensive care patients in order to guide further immumodulation approaches.
Collapse
Affiliation(s)
- Guillaume Monneret
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France.
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France.
| | | | - Jean-Christophe Richard
- Medical Intensive Care Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Martin Cour
- Medical Intensive Care Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | - Thomas Rimmelé
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
- Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | - Lorna Garnier
- Immunology Laboratory, Hospices Civils de Lyon, Lyon-Sud University Hospital, 69495, Pierre Bénite, France
| | - Hodane Yonis
- Medical Intensive Care Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Remy Coudereau
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
| | - Morgane Gossez
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude, Bernard-Lyon 1, Lyon, France
| | - Christophe Malcus
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
| | - Florent Wallet
- Intensive Care Department, Hospices Civils de Lyon, Lyon-Sud University Hospital, 69495, Pierre-Bénite, France
| | - Marie-Charlotte Delignette
- Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Frederic Dailler
- Neurological Anesthesiology and Intensive Care Department, Hospices Civils de Lyon, Pierre Wertheimer Hospital, Lyon, France
| | - Marielle Buisson
- Centre d'Investigation Clinique de Lyon (CIC 1407 Inserm), Hospices Civils de Lyon, Lyon, France
| | - Laurent Argaud
- Medical Intensive Care Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Anne-Claire Lukaszewicz
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
- Medical Intensive Care Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | - Fabienne Venet
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude, Bernard-Lyon 1, Lyon, France
| |
Collapse
|
10
|
Zhou R, Johnson KE, Rousseau JF, Rathouz PJ. Comparative effectiveness of dexamethasone in treatment of hospitalized COVID-19 patients in the United States during the first year of the pandemic: Findings from the National COVID Cohort Collaborative (N3C) data repository. PLoS One 2024; 19:e0294892. [PMID: 38512832 PMCID: PMC10956822 DOI: 10.1371/journal.pone.0294892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 11/11/2023] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Dexamethasone was approved for use in hospitalized COVID-19 patients early in the pandemic based on the RECOVERY trial, but evidence is still needed to support its real-world effectiveness in heterogeneous populations of patients with a wide range of comorbidities. METHODS COVID-19 inpatients represented within the National COVID Cohort Collaborative (N3C) Data Enclave, prior to vaccine availability, were studied. Primary outcome was in-hospital death; secondary outcome was combined in-hospital death and severe outcome defined by use of ECMO or mechanical ventilation. Missing data were imputed with single imputation. Dexamethasone-treated patients were propensity score (PS) matched to non-dexamethasone-treated controls, stratified by remdesivir treatment and based on demographics, baseline laboratory values, comorbidities, and amount of missing data before imputation. Treatment benefit was quantified using logistic regression. Further sensitivity analyses were performed using clinical adjusters in matched groups and in strata defined by quartiles of PS. RESULTS Dexamethasone treatment was associated with reduced risk of in-hospital mortality for n = 1,263 treated, matched 1:3 to untreated, patients not receiving remdesivir (OR = 0.77, 95% CI: 0.62 to 0.95, p = 0.017), and for n = 804 treated, matched 1:1 to untreated, patients receiving remdesivir (OR = 0.74, 95% CI: 0.53 to 1.02, p = 0.054). Treatment showed secondary outcome benefit. In sensitivity analyses, treatment effect generally remained similar with some heterogeneity of benefit across quartiles of PS, possibly reflecting concentration of benefit among the more severely affected. CONCLUSIONS We add evidence that dexamethasone provides benefit with respect to mortality and severe outcomes in a diverse, national hospitalized sample, prior to vaccine availability.
Collapse
Affiliation(s)
- Richard Zhou
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
| | - Kaitlyn E. Johnson
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, United States of America
- The Pandemic Prevention Institute, The Rockefeller Foundation, New York, New York, United States of America
| | - Justin F. Rousseau
- Dell Medical School at the University of Texas at Austin, Austin, Texas, United States of America
| | - Paul J. Rathouz
- Dell Medical School at the University of Texas at Austin, Austin, Texas, United States of America
| | | |
Collapse
|
11
|
Jacq A, Auvray C, Blot M, Bouhemad B, Casenaz A, Lamarthée B, Legendre M, Quenot JP, Zanetta G, Tinel C. Adequacy to immunosuppression management guidelines in kidney transplant recipients with severe COVID-19 pneumonia: a practice survey. FRONTIERS IN TRANSPLANTATION 2024; 3:1305152. [PMID: 38993755 PMCID: PMC11235282 DOI: 10.3389/frtra.2024.1305152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 03/01/2024] [Indexed: 07/13/2024]
Abstract
Introduction Coronavirus disease 2019 (COVID-19) poses an important risk of morbidity and of mortality, in patients after solid organ transplantation. Recommendations have been issued by various transplantation societies at the national and European level to manage the immunosuppressive (IS) regimen upon admission to intensive care unit (ICU). Method The aim of this study was to evaluate the adequacy of IS regimen minimization strategy in kidney transplant recipients hospitalized in an ICU for severe COVID-19, in relation to the issued recommendations. Results The immunosuppressive therapy was minimized in all patients, with respectively 63% and 59% of the patients meeting the local and european recommendations upon admission. During ICU stay, IS was further tapered leading to 85% (local) and 78% (european) adequacy, relative to the guidelines. The most frequent deviation was the lack of complete withdrawal of mycophenolic acid (22%). Nevertheless, the adequacy/inadequacy status was not associated to the ICU- or one-year-mortality. Discussion In this single-center cohort, the only variable associated with a reduction in mortality was vaccination, emphasizing that the key issue is immunization prior to infection, not restoration of immunity during ICU stay.
Collapse
Affiliation(s)
- Amélie Jacq
- Department of Nephrology and Kidney Transplantation, Dijon University Hospital, Dijon, France
| | | | - Mathieu Blot
- Department of Infectious Diseases, Dijon University Hospital, Dijon, France
| | - Belaïd Bouhemad
- Anesthesia and Intensive Care Department, Dijon University Hospital, Dijon, France
| | - Alice Casenaz
- Department of Virology, Dijon University Hospital, Dijon, France
| | - Baptiste Lamarthée
- TAI-IT Department, Inserm UMR Right, Université de Franche Comté, EFS BFC, Besançon, France
| | - Mathieu Legendre
- Department of Nephrology and Kidney Transplantation, Dijon University Hospital, Dijon, France
| | - Jean-Pierre Quenot
- Medical Intensive Care Department, Dijon University Hospital, Dijon, France
| | - Gilbert Zanetta
- Department of Nephrology and Kidney Transplantation, Dijon University Hospital, Dijon, France
| | - Claire Tinel
- Department of Nephrology and Kidney Transplantation, Dijon University Hospital, Dijon, France
- TAI-IT Department, Inserm UMR Right, Université de Franche Comté, EFS BFC, Besançon, France
| |
Collapse
|
12
|
Varghese R, Digholkar G, Karsiya J, Salvi S, Shah J, Kumar D, Sharma R. PDE5 inhibitors: breaking new grounds in the treatment of COVID-19. Drug Metab Pers Ther 2023; 38:295-307. [PMID: 38167268 DOI: 10.1515/dmpt-2023-0011] [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: 03/29/2023] [Accepted: 07/30/2023] [Indexed: 01/05/2024]
Abstract
INTRODUCTION Despite the ever-increasing occurrences of the coronavirus disease (COVID-19) cases around the world, very few medications have been validated in the clinical trials to combat COVID-19. Although several vaccines have been developed in the past quarter, the time elapsed between deployment and administration remains a major impediment. CONTENT Repurposing of pre-approved drugs, such as phosphodiesterase 5 (PDE5) inhibitors, could be a game-changer while lessening the burden on the current healthcare system. Repurposing and developing phosphodiesterase 5 (PDE5) inhibitors could extrapolate their utility to combat the SARS-CoV-2 infection, and potentially aid in the management of the symptoms associated with its newer variants such as BF.7, BQ.1, BQ.1.1, XBB.1.5, and XBB.1.16. SUMMARY Administration of PDE5 inhibitors via the oral and intravenous route demonstrates other potential off-label benefits, including anti-apoptotic, anti-inflammatory, antioxidant, and immunomodulatory effects, by intercepting several pathways. These effects can not only be of clinical importance in mild-to-moderate, but also moderate-to-severe SARS-CoV-2 infections. This article explores the various mechanisms by which PDE5 inhibitors alleviates the symptoms associated with COVID-19 as well as well as highlights recent studies and findings. OUTLOOK These benefits of PDE5 inhibitors make it a potential drug in the physicians' armamentarium in alleviating symptoms associated with SARS-CoV-2 infection. However, adequate clinical studies must be instituted to eliminate any untoward adverse events.
Collapse
Affiliation(s)
- Ryan Varghese
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, India
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Gargi Digholkar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, India
| | - Jainam Karsiya
- River Route Creative Group LLP, Mumbai, Maharashtra, India
| | - Sahil Salvi
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, India
| | - Jeenam Shah
- Department of Pulmonology, Saifee Hospital, Girgaon, Mumbai, Maharashtra, India
| | - Dileep Kumar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, India
- Department of Entomology, University of California, Davis, CA, USA
- UC Davis Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
13
|
Minnema J, Tap L, van der Bol JM, van Deudekom FJA, Faes MC, Jansen SWM, van der Linden CMJ, Lucke JA, Mooijaart SP, van Munster B, Noordam R, van Raaij BFM, Ruiter R, Smits RAL, Willems HC, Mattace-Raso FUS, Polinder-Bos HA. Delirium in older patients with COVID-19: Prevalence, risk factors and clinical outcomes across the first three waves of the pandemic. Int J Geriatr Psychiatry 2023; 38:e6024. [PMID: 37909117 DOI: 10.1002/gps.6024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 10/17/2023] [Indexed: 11/02/2023]
Abstract
OBJECTIVES Delirium is a serious condition, which poses treatment challenges during hospitalisation for COVID-19. Improvements in testing, vaccination and treatment might have changed patient characteristics and outcomes through the pandemic. We evaluated whether the prevalence and risk factors for delirium, and the association of delirium with in-hospital mortality changed through the pandemic. METHODS This study was part of the COVID-OLD study in 19 Dutch hospitals including patients ≥70 years in the first (spring 2020), second (autumn 2020) and third wave (autumn 2021). Multivariable logistic regression models were used to study risk factors for delirium, and in-hospital mortality. Differences in effect sizes between waves were studied by including interaction terms between wave and risk factor in logistic regression models. RESULTS 1540, 884 and 370 patients were included in the first, second and third wave, respectively. Prevalence of delirium in the third wave (12.7%) was significantly lower compared to the first (22.5%) and second wave (23.5%). In multivariable-adjusted analyses, pre-existing memory problems was a consistent risk factor for delirium across waves. Previous delirium was a risk factor for delirium in the first wave (OR 4.02), but not in the second (OR 1.61) and third wave (OR 2.59, p-value interaction-term 0.028). In multivariable-adjusted analyses, delirium was not associated with in-hospital mortality in all waves. CONCLUSION Delirium prevalence declined in the third wave, which might be the result of vaccination and improved treatment strategies. Risk factors for delirium remained consistent across waves, although some attenuation was seen in the second wave.
Collapse
Affiliation(s)
- Julia Minnema
- Division of Geriatric Medicine, Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Lisanne Tap
- Division of Geriatric Medicine, Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | | | | - Miriam C Faes
- Department of Geriatrics, Amphia Hospital, Breda, The Netherlands
| | - Steffy W M Jansen
- Department of Geriatrics, Catharina Hospital, Eindhoven, the Netherlands
| | | | - Jacinta A Lucke
- Department of Emergency Medicine, Spaarne Gasthuis, Haarlem, The Netherlands
| | - Simon P Mooijaart
- Department of Internal Medicine, Section of Geriatrics and Gerontology, Leiden University Medical Centre, Leiden, The Netherlands
- LUMC Center of Medicine for Older People, Leiden University Medical Centre, Leiden, The Netherlands
| | - Barbara van Munster
- Department of Internal Medicine and Geriatrics, University Medical Centre Groningen, Groningen, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Geriatrics and Gerontology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Bas F M van Raaij
- Department of Internal Medicine, Section of Geriatrics and Gerontology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Rikje Ruiter
- Department of Internal Medicine, Maasstad Ziekenhuis, Rotterdam, The Netherlands
| | - Rosalinde A L Smits
- Department of Internal Medicine, Section of Geriatrics and Gerontology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Hanna C Willems
- Department of Internal Medicine and Geriatrics, AmsterdamUMC, Amsterdam, The Netherlands
| | - Francesco U S Mattace-Raso
- Division of Geriatric Medicine, Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Harmke A Polinder-Bos
- Division of Geriatric Medicine, Department of Internal Medicine, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
14
|
Nguyen TM, Craig DB, Tran D, Nguyen T, Draghici S. A novel approach for predicting upstream regulators (PURE) that affect gene expression. Sci Rep 2023; 13:18571. [PMID: 37903768 PMCID: PMC10616115 DOI: 10.1038/s41598-023-41374-0] [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: 02/06/2023] [Accepted: 08/25/2023] [Indexed: 11/01/2023] Open
Abstract
External factors such as exposure to a chemical, drug, or toxicant (CDT), or conversely, the lack of certain chemicals can cause many diseases. The ability to identify such causal CDTs based on changes in the gene expression profile is extremely important in many studies. Furthermore, the ability to correctly infer CDTs that can revert the gene expression changes induced by a given disease phenotype is a crucial step in drug repurposing. We present an approach for Predicting Upstream REgulators (PURE) designed to tackle this challenge. PURE can correctly infer a CDT from the measured expression changes in a given phenotype, as well as correctly identify drugs that could revert disease-induced gene expression changes. We compared the proposed approach with four classical approaches as well as with the causal analysis used in Ingenuity Pathway Analysis (IPA) on 16 data sets (1 rat, 5 mouse, and 10 human data sets), involving 8 chemicals or drugs. We assessed the results based on the ability to correctly identify the CDT as indicated by its rank. We also considered the number of false positives, i.e. CDTs other than the correct CDT that were reported to be significant by each method. The proposed approach performed best in 11 out of the 16 experiments, reporting the correct CDT at the very top 7 times. IPA was the second best, reporting the correct CDT at the top 5 times, but was unable to identify the correct CDT at all in 5 out of the 16 experiments. The validation results showed that our approach, PURE, outperformed some of the most popular methods in the field. PURE could effectively infer the true CDTs responsible for the observed gene expression changes and could also be useful in drug repurposing applications.
Collapse
Affiliation(s)
- Tuan-Minh Nguyen
- Department of Computer Science, Wayne State University, Detroit, 48202, USA
| | - Douglas B Craig
- Department of Computer Science, Wayne State University, Detroit, 48202, USA
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Duc Tran
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Tin Nguyen
- Department of Computer Science and Software Engineering, Auburn University, Auburn, 36849, USA
| | - Sorin Draghici
- Department of Computer Science, Wayne State University, Detroit, 48202, USA.
- Advaita Bioinformatics, Ann Arbor, MI, 48105, USA.
| |
Collapse
|
15
|
Kim MH, Shin HJ, Kim J, Jo S, Kim EK, Park YS, Kyong T. Novel Risks of Unfavorable Corticosteroid Response in Patients with Mild-to-Moderate COVID-19 Identified Using Artificial Intelligence-Assisted Analysis of Chest Radiographs. J Clin Med 2023; 12:5852. [PMID: 37762792 PMCID: PMC10532025 DOI: 10.3390/jcm12185852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/25/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
The prediction of corticosteroid responses in coronavirus disease 2019 (COVID-19) patients is crucial in clinical practice, and exploring the role of artificial intelligence (AI)-assisted analysis of chest radiographs (CXR) is warranted. This retrospective case-control study involving mild-to-moderate COVID-19 patients treated with corticosteroids was conducted from 4 September 2021, to 30 August 2022. The primary endpoint of the study was corticosteroid responsiveness, defined as the advancement of two or more of the eight-categories-ordinal scale. Serial abnormality scores for consolidation and pleural effusion on CXR were obtained using a commercial AI-based software based on days from the onset of symptoms. Amongst the 258 participants included in the analysis, 147 (57%) were male. Multivariable logistic regression analysis revealed that high pleural effusion score at 6-9 days from onset of symptoms (adjusted odds ratio of (aOR): 1.022, 95% confidence interval (CI): 1.003-1.042, p = 0.020) and consolidation scores up to 9 days from onset of symptoms (0-2 days: aOR: 1.025, 95% CI: 1.006-1.045, p = 0.010; 3-5 days: aOR: 1.03 95% CI: 1.011-1.051, p = 0.002; 6-9 days: aOR; 1.052, 95% CI: 1.015-1.089, p = 0.005) were associated with an unfavorable corticosteroid response. AI-generated scores could help intervene in the use of corticosteroids in COVID-19 patients who would not benefit from them.
Collapse
Affiliation(s)
- Min Hyung Kim
- Department of Internal Medicine, Division of Infectious Disease, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si 16995, Republic of Korea; (M.H.K.); (Y.S.P.)
| | - Hyun Joo Shin
- Department of Radiology, Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si 16995, Republic of Korea; (H.J.S.); (E.-K.K.)
- Center for Digital Health, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si 16995, Republic of Korea
| | - Jaewoong Kim
- Department of Hospital Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si 16995, Republic of Korea; (J.K.); (S.J.)
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sunhee Jo
- Department of Hospital Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si 16995, Republic of Korea; (J.K.); (S.J.)
| | - Eun-Kyung Kim
- Department of Radiology, Research Institute of Radiological Science and Center for Clinical Imaging Data Science, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si 16995, Republic of Korea; (H.J.S.); (E.-K.K.)
- Center for Digital Health, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si 16995, Republic of Korea
| | - Yoon Soo Park
- Department of Internal Medicine, Division of Infectious Disease, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si 16995, Republic of Korea; (M.H.K.); (Y.S.P.)
| | - Taeyoung Kyong
- Department of Hospital Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si 16995, Republic of Korea; (J.K.); (S.J.)
| |
Collapse
|
16
|
Zulfiqar S, Gasser RB, Ghodsian S, Almukhtar M, Holland C, Rostami A. Strongyloides coinfection in COVID-19 patients treated with corticosteroids: A systematic review. Rev Med Virol 2023; 33:e2469. [PMID: 37353858 DOI: 10.1002/rmv.2469] [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: 03/30/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/25/2023]
Abstract
The COVID-19 pandemic linked to the virus SARS-CoV-2, which began in China, affected ∼765 million people as of 30 April 2023. The widespread use of corticosteroids for the symptomatic treatment of COVID-19 could lead to the reactivation of infections of opportunistic pathogens, including Strongyloides. We sought to determine the clinical symptoms and demographic characteristics of SARS-CoV-2-Strongyloides co-infection, particularly in patients with severe disease and being treated with immunosuppressive drugs. To do this, we undertook a systematic review of the literature, and searched public accessible scientific databases-the Web of Science, Scopus, PubMed/Medline and Embase -for eligible studies (1 December 2019 to 30 August 2022). The review protocol is registered in PROSPERO (CRD42022377062). Descriptive statistical analyses were used to present the clinical and laboratory parameters of the co-infection; for this, we calculated prevalence using the following formula: positive cases/total number of cases × 100. Of a total of 593 studies identified, 17 studies reporting 26 co-infected patients met the criteria for inclusion in this review. The median age of these patients was 55.14 years. Most of cases (53.8%) were treated with dexamethasone, followed by methylprednisolone (26.9%). Eighteen of 26 patients were immigrants living in European countries or the USA; most of these immigrants originated from Latin America (58%) and South-East Asia (11%). The commonest symptoms of co-infection were abdominal pain (50%), fever (46.1%), dyspnoea (30.7%) and cough (30.7%), and frequently reported laboratory findings were high absolute eosinophil count (38.4%), high white blood cell count (30.7%), high C-reactive protein (23.0%) and high neutrophil count (19.2%). Two of the 26 patients (7.7%) had fatal outcomes. Most of the SARS-CoV-2-Strongyloides coinfected cases were immigrants living in developed countries, emphasising the need for clinicians in these countries to be aware of clinical and laboratory parameters associated with such co-infections, as well as the key importance of rapid and accurate diagnostic tests for timely and effective diagnosis and patient management.
Collapse
Affiliation(s)
- Sana Zulfiqar
- School of Medicine, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
| | - Robin B Gasser
- Department of Veterinary Biosciences, Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Sahar Ghodsian
- Department of Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mustafa Almukhtar
- Takhar Family Medicine and Urgent Care, Sacramento, California, United States
| | - Celia Holland
- Department of Zoology, School of Natural Sciences, Trinity College Dublin, the University of Dublin, College Green Dublin, Ireland
| | - Ali Rostami
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
17
|
Varghese R, Digholkar G, Karsiya J, Salvi S, Shah J, Kumar D, Sharma R. PDE5 inhibitors: breaking new grounds in the treatment of COVID-19. Drug Metab Pers Ther 2023; 0:dmdi-2023-0011. [PMID: 37608528 DOI: 10.1515/dmdi-2023-0011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/30/2023] [Indexed: 08/24/2023]
Abstract
INTRODUCTION Despite the ever-increasing occurrences of the coronavirus disease (COVID-19) cases around the world, very few medications have been validated in the clinical trials to combat COVID-19. Although several vaccines have been developed in the past quarter, the time elapsed between deployment and administration remains a major impediment. CONTENT Repurposing of pre-approved drugs, such as phosphodiesterase 5 (PDE5) inhibitors, could be a game-changer while lessening the burden on the current healthcare system. Repurposing and developing phosphodiesterase 5 (PDE5) inhibitors could extrapolate their utility to combat the SARS-CoV-2 infection, and potentially aid in the management of the symptoms associated with its newer variants such as BF.7, BQ.1, BQ.1.1, XBB.1.5, and XBB.1.16. SUMMARY Administration of PDE5 inhibitors via the oral and intravenous route demonstrates other potential off-label benefits, including anti-apoptotic, anti-inflammatory, antioxidant, and immunomodulatory effects, by intercepting several pathways. These effects can not only be of clinical importance in mild-to-moderate, but also moderate-to-severe SARS-CoV-2 infections. This article explores the various mechanisms by which PDE5 inhibitors alleviates the symptoms associated with COVID-19 as well as well as highlights recent studies and findings. OUTLOOK These benefits of PDE5 inhibitors make it a potential drug in the physicians' armamentarium in alleviating symptoms associated with SARS-CoV-2 infection. However, adequate clinical studies must be instituted to eliminate any untoward adverse events.
Collapse
Affiliation(s)
- Ryan Varghese
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, India
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Gargi Digholkar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, India
| | - Jainam Karsiya
- River Route Creative Group LLP, Mumbai, Maharashtra, India
| | - Sahil Salvi
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, India
| | - Jeenam Shah
- Department of Pulmonology, Saifee Hospital, Girgaon, Mumbai, Maharashtra, India
| | - Dileep Kumar
- Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Pune, Maharashtra, India
- Department of Entomology, University of California, Davis, CA, USA
- UC Davis Comprehensive Cancer Center, University of California, Davis, CA, USA
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
18
|
Resende ADS, de Oliveira YLM, de Franca MNF, Magalhães LS, Correa CB, Fukutani KF, Lipscomb MW, de Moura TR. Obesity in Severe COVID-19 Patients Has a Distinct Innate Immune Phenotype. Biomedicines 2023; 11:2116. [PMID: 37626613 PMCID: PMC10452870 DOI: 10.3390/biomedicines11082116] [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: 06/23/2023] [Revised: 07/13/2023] [Accepted: 07/23/2023] [Indexed: 08/27/2023] Open
Abstract
Obesity alters the capacity of effective immune responses in infections. To further address this phenomenon in the context of COVID-19, this study investigated how the immunophenotype of leukocytes was altered in individuals with obesity in severe COVID-19. This cross-sectional study enrolled 27 ICU COVID-19 patients (67% women, 56.33 ± 19.55 years) that were assigned to obese (BMI ≥ 30 kg/m2, n = 9) or non-obese (BMI < 30kg/m2, n = 18) groups. Monocytes, NK, and both Low-Density (LD) and High-Density (HD) neutrophils were isolated from peripheral blood samples, and surface receptors' frequency and expression patterns were analyzed by flow cytometry. Clinical status and biochemical data were additionally evaluated. The frequency of monocytes was negatively correlated with BMI, while NK cells and HD neutrophils were positively associated (p < 0.05). Patients with obesity showed a significant reduction of monocytes, and these cells expressed high levels of PD-L1 (p < 0.05). A higher frequency of NK cells and increased expression of TREM-1+ on HD neutrophils were detected in obese patients (p < 0.05). The expression of receptors related to antigen-presentation, phagocytosis, chemotaxis, inflammation and suppression were strongly correlated with clinical markers only in obese patients (p < 0.05). Collectively, these outcomes revealed that obesity differentially affected, and largely depressed, innate immune response in severe COVID-19.
Collapse
Affiliation(s)
- Ayane de Sá Resende
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju 49060-100, Sergipe, Brazil; (Y.L.M.d.O.); (M.N.F.d.F.); (L.S.M.); (C.B.C.); (K.F.F.)
| | - Yrna Lorena Matos de Oliveira
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju 49060-100, Sergipe, Brazil; (Y.L.M.d.O.); (M.N.F.d.F.); (L.S.M.); (C.B.C.); (K.F.F.)
| | - Mariana Nobre Farias de Franca
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju 49060-100, Sergipe, Brazil; (Y.L.M.d.O.); (M.N.F.d.F.); (L.S.M.); (C.B.C.); (K.F.F.)
| | - Lucas Sousa Magalhães
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju 49060-100, Sergipe, Brazil; (Y.L.M.d.O.); (M.N.F.d.F.); (L.S.M.); (C.B.C.); (K.F.F.)
- Department of Parasitology and Pathology, ICBS, Federal University of Alagoas, Maceio 57072-900, Alagoas, Brazil
| | - Cristiane Bani Correa
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju 49060-100, Sergipe, Brazil; (Y.L.M.d.O.); (M.N.F.d.F.); (L.S.M.); (C.B.C.); (K.F.F.)
- Physiological Sciences Graduate Program, Federal University of Sergipe, São Cristovao 49100-000, Sergipe, Brazil
| | - Kiyoshi Ferreira Fukutani
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju 49060-100, Sergipe, Brazil; (Y.L.M.d.O.); (M.N.F.d.F.); (L.S.M.); (C.B.C.); (K.F.F.)
| | | | - Tatiana Rodrigues de Moura
- Health Sciences Graduate Program, Federal University of Sergipe, Aracaju 49060-100, Sergipe, Brazil; (Y.L.M.d.O.); (M.N.F.d.F.); (L.S.M.); (C.B.C.); (K.F.F.)
| |
Collapse
|
19
|
Tenda ED, Henrina J, Samosir J, Amalia R, Yulianti M, Pitoyo CW, Setiati S. Machine learning-based COVID-19 acute respiratory distress syndrome phenotyping and clinical outcomes: A systematic review. Heliyon 2023; 9:e17276. [PMID: 37366530 PMCID: PMC10275654 DOI: 10.1016/j.heliyon.2023.e17276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/28/2023] Open
Abstract
COVID-19-related acute respiratory distress syndrome (CARDS) has been suggested to differ from the typical ARDS. While distinct phenotypes of ARDS have been identified through latent class analysis (LCA), it is unclear whether such phenotypes exist for CARDS and how they affect clinical outcomes. To address this question, we conducted a systematic review of the current evidence.We searched several, including PubMed, EBSCO Host, and Web of Science, from inception to July 1, 2022. Our exposure and outcome of interest were different CARDS phenotypes identified and their associated outcomes, such as 28-day, 90-day, 180-day mortality, ventilator-free days, and other relevant outcomes.We identified four studies comprising a total of 1776 CARDS patients.Of the four studies, three used LCA to identify subphenotypes (SPs) of CARDS. One study based on longitudinal data identified two SPs, with SP2 associated with worse ventilation and mechanical parameters than SP1. The other two studies based on baseline data also identified two SPs, with SP2 and SP1 were associated with hyperinflammatory and hypoinflammatory CARDS, respectively. The fourth study identified three SPs primarily stratified by comorbidities using multifactorial analysis.All studies identified a subphenotype associated with poorer outcomes, including mortality, ventilator-free days, multiple-organ injury, and pulmonary embolism. Two studies reported differential responses to corticosteroids among the SPs, with improved mortality in the hyperinflammatory and worse in the hypoinflammatory SPs.Overall, our review highlights the importance of phenotyping in understanding CARDS and its impact on disease management and prognostication. However, a consensus approach to phenotyping is necessary to ensure consistency and comparability across studies. We recommend that randomized clinical trials stratified by phenotype should only be initiated after such consensus is reached. Short title COVID-19 ARDS subphenotypes and outcomes.
Collapse
Affiliation(s)
- Eric Daniel Tenda
- Division of Respirology and Critical Care, Department of Internal Medicine, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Joshua Henrina
- Division of Respirology and Critical Care, Department of Internal Medicine, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Jistrani Samosir
- Department of Internal Medicine, Dr.T.C. Hillers General Public Hospital, East Nusa Tenggara, Kabupaten Sikka, Indonesia
| | - Ridha Amalia
- University of Indonesia Hospital, Depok, Indonesia
| | - Mira Yulianti
- Division of Respirology and Critical Care, Department of Internal Medicine, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Ceva Wicaksono Pitoyo
- Division of Respirology and Critical Care, Department of Internal Medicine, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Siti Setiati
- Division of Geriatric Medicine, Department of Internal Medicine, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, Indonesia
- Clinical Epidemiology and Evidence-Based Medicine Unit, Faculty of Medicine, Cipto Mangunkusumo Hospital, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
20
|
Barabutis N. Activating transcription factor 6 in the endothelial context. Pulm Pharmacol Ther 2023; 80:102216. [PMID: 37121466 PMCID: PMC10155510 DOI: 10.1016/j.pupt.2023.102216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Affiliation(s)
- Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, 1800 Bienville Drive, Monroe, LA, 71201, USA.
| |
Collapse
|
21
|
Higher dose corticosteroids in patients admitted to hospital with COVID-19 who are hypoxic but not requiring ventilatory support (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet 2023. [PMID: 37060915 PMCID: PMC10156147 DOI: 10.1016/s0140-6736(23)00510-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Abstract
BACKGROUND Low-dose corticosteroids have been shown to reduce mortality for patients with COVID-19 requiring oxygen or ventilatory support (non-invasive mechanical ventilation, invasive mechanical ventilation, or extracorporeal membrane oxygenation). We evaluated the use of a higher dose of corticosteroids in this patient group. METHODS This randomised, controlled, open-label platform trial (Randomised Evaluation of COVID-19 Therapy [RECOVERY]) is assessing multiple possible treatments in patients hospitalised for COVID-19. Eligible and consenting adult patients with clinical evidence of hypoxia (ie, receiving oxygen or with oxygen saturation <92% on room air) were randomly allocated (1:1) to either usual care with higher dose corticosteroids (dexamethasone 20 mg once daily for 5 days followed by 10 mg dexamethasone once daily for 5 days or until discharge if sooner) or usual standard of care alone (which included dexamethasone 6 mg once daily for 10 days or until discharge if sooner). The primary outcome was 28-day mortality among all randomised participants. On May 11, 2022, the independent data monitoring committee recommended stopping recruitment of patients receiving no oxygen or simple oxygen only due to safety concerns. We report the results for these participants only. Recruitment of patients receiving ventilatory support is ongoing. The RECOVERY trial is registered with ISRCTN (50189673) and ClinicalTrials.gov (NCT04381936). FINDINGS Between May 25, 2021, and May 13, 2022, 1272 patients with COVID-19 and hypoxia receiving no oxygen (eight [1%]) or simple oxygen only (1264 [99%]) were randomly allocated to receive usual care plus higher dose corticosteroids (659 patients) versus usual care alone (613 patients, of whom 87% received low-dose corticosteroids during the follow-up period). Of those randomly assigned, 745 (59%) were in Asia, 512 (40%) in the UK, and 15 (1%) in Africa. 248 (19%) had diabetes and 769 (60%) were male. Overall, 123 (19%) of 659 patients allocated to higher dose corticosteroids versus 75 (12%) of 613 patients allocated to usual care died within 28 days (rate ratio 1·59 [95% CI 1·20-2·10]; p=0·0012). There was also an excess of pneumonia reported to be due to non-COVID infection (64 cases [10%] vs 37 cases [6%]; absolute difference 3·7% [95% CI 0·7-6·6]) and an increase in hyperglycaemia requiring increased insulin dose (142 [22%] vs 87 [14%]; absolute difference 7·4% [95% CI 3·2-11·5]). INTERPRETATION In patients hospitalised for COVID-19 with clinical hypoxia who required either no oxygen or simple oxygen only, higher dose corticosteroids significantly increased the risk of death compared with usual care, which included low-dose corticosteroids. The RECOVERY trial continues to assess the effects of higher dose corticosteroids in patients hospitalised with COVID-19 who require non-invasive ventilation, invasive mechanical ventilation, or extracorporeal membrane oxygenation. FUNDING UK Research and Innovation (Medical Research Council), National Institute of Health and Care Research, and Wellcome Trust.
Collapse
|
22
|
Heller M, Henrici C, Büttner J, Leube S, Treske I, Pospischil P, Doll M, Schanz I, Hallier A, Herrmann E, Schmidt M, Sarrazin C. SARS-CoV-2 neutralizing antibody therapies: an early retrospective cohort study of 26 hospitalized patients treated with bamlanivimab or casirivimab/imdevimab. Int J Infect Dis 2023; 129:260-265. [PMID: 36690138 PMCID: PMC9859643 DOI: 10.1016/j.ijid.2023.01.012] [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: 01/27/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVES In this early retrospective cohort study, a total of 26 patients with SARS-CoV-2 were treated with bamlanivimab or casirivimab/imdevimab, and the reduction of the viral load associated with the developed clinical symptoms was analyzed. METHODS Patients in the intervention groups received bamlanivimab or casirivimab/imdevimab. Patients without treatment served as control. Outcomes were assessed by clinical symptoms and change in log viral load from baseline based on the cycle threshold over a period of 18 days. RESULTS Median log viral load decline was higher in both intervention groups after 3 and 6 days compared to control. However, at later time points, the decline of the viral load was more distinct in the control group. Mild symptoms of COVID-19 were observed in 6.3% of the intervention groups and in no patient of the control. No patients treated with bamlanivimab, 18.8% treated with casirivimab/imdevimab, and 14.2% in the control group developed moderate symptoms. Severe symptoms were recorded only in the control group (14.2%), including one related death. CONCLUSION Treatment with monoclonal SARS-CoV-2 antibodies seems to accelerate decline of virus loads, especially in the first 6 days after administration, compared to control. This may be associated with a reduced likeliness of a severe course of COVID-19.
Collapse
Affiliation(s)
- Martin Heller
- Department of Gastroenterology and Infectiology, Medical Clinic, St. Josefs-Hospital Wiesbaden, Wiesbaden, Germany.
| | - Clara Henrici
- Department of Gastroenterology and Infectiology, Medical Clinic, St. Josefs-Hospital Wiesbaden, Wiesbaden, Germany
| | - Judith Büttner
- Department of Gastroenterology and Infectiology, Medical Clinic, St. Josefs-Hospital Wiesbaden, Wiesbaden, Germany
| | - Sebastian Leube
- Department of Cardiology, Medical Clinic, St. Josefs-Hospital Wiesbaden, Wiesbaden, Germany
| | - Isabelle Treske
- Department of Cardiology, Medical Clinic, St. Josefs-Hospital Wiesbaden, Wiesbaden, Germany
| | - Petra Pospischil
- Department of Gastroenterology and Infectiology, Medical Clinic, St. Josefs-Hospital Wiesbaden, Wiesbaden, Germany
| | - Michael Doll
- Department of Gastroenterology and Infectiology, Medical Clinic, St. Josefs-Hospital Wiesbaden, Wiesbaden, Germany
| | - Ilka Schanz
- Department of Interdisciplinary Intensive Care, Medical Clinic, St. Josefs-Hospital Wiesbaden, Wiesbaden, Germany
| | - Agnes Hallier
- Department of Emergency Medicine, St. Josefs-Hospital Wiesbaden, Wiesbaden, Germany
| | - Eva Herrmann
- Institute for Biostatistics and Mathematical Modeling, Universitätsklinikum der Goethe-Universität Frankfurt, Frankfurt am Main, Germany
| | - Michael Schmidt
- Blutspendedienst, DRK Baden-Württemberg - Hessen Gesellschaft mit beschränkter Haftung, Frankfurt Germany
| | - Christoph Sarrazin
- Department of Gastroenterology and Infectiology, Medical Clinic, St. Josefs-Hospital Wiesbaden, Wiesbaden, Germany
| |
Collapse
|
23
|
Segú-Vergés C, Artigas L, Coma M, Peck RW. Artificial intelligence assessment of the potential of tocilizumab along with corticosteroids therapy for the management of COVID-19 evoked acute respiratory distress syndrome. PLoS One 2023; 18:e0280677. [PMID: 36791125 PMCID: PMC9931125 DOI: 10.1371/journal.pone.0280677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/05/2023] [Indexed: 02/16/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS), associated with high mortality rate, affects up to 67% of hospitalized COVID-19 patients. Early evidence indicated that the pathogenesis of COVID-19 evoked ARDS is, at least partially, mediated by hyperinflammatory cytokine storm in which interleukin 6 (IL-6) plays an essential role. The corticosteroid dexamethasone is an effective treatment for severe COVID-19 related ARDS. However, trials of other immunomodulatory therapies, including anti-IL6 agents such as tocilizumab and sarilumab, have shown limited evidence of benefit as monotherapy. But recently published large trials have reported added benefit of tocilizumab in combination with dexamethasone in severe COVID-19 related ARDS. In silico tools can be useful to shed light on the mechanisms evoked by SARS-CoV-2 infection and of the potential therapeutic approaches. Therapeutic performance mapping system (TPMS), based on systems biology and artificial intelligence, integrate available biological, pharmacological and medical knowledge to create mathematical models of the disease. This technology was used to identify the pharmacological mechanism of dexamethasone, with or without tocilizumab, in the management of COVID-19 evoked ARDS. The results showed that while dexamethasone would be addressing a wider range of pathological processes with low intensity, tocilizumab might provide a more direct and intense effect upon the cytokine storm. Based on this in silico study, we conclude that the use of tocilizumab alongside dexamethasone is predicted to induce a synergistic effect in dampening inflammation and subsequent pathological processes, supporting the beneficial effect of the combined therapy in critically ill patients. Future research will allow identifying the ideal subpopulation of patients that would benefit better from this combined treatment.
Collapse
Affiliation(s)
- Cristina Segú-Vergés
- Anaxomics Biotech, Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | - Richard W. Peck
- Pharma Research & Development (pRED), F. Hoffman-La Roche Ltd., Basel, Switzerland
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
24
|
Pereira PC, de Lima CJ, Fernandes AB, Zângaro RA, Villaverde AB. Cardiopulmonary and hematological effects of infrared LED photobiomodulation in the treatment of SARS-COV2. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 238:112619. [PMID: 36495670 PMCID: PMC9721157 DOI: 10.1016/j.jphotobiol.2022.112619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND COVID-19 disease is caused by SARS-CoV-2 which can trigger acute respiratory syndrome, which presents with dense alveolar and interstitial infiltrates and pulmonary edema, causing severe hypoxemia and significant alteration to pulmonary mechanics with reduced pulmonary compliance. The photobiomodulation technique alters cellular and molecular metabolism, showing promising results regarding the reduction of acute pulmonary inflammation. OBJECTIVE To compare the photomodulation technique using near-infrared LED to conventional respiratory physiotherapy treatment in patients with COVID-19 in reversing acute conditions, reducing hospitalization time, and decreasing the need for oxygen therapy. METHODOLOGY The cohort was comprised of 30 patients undergoing COVID-19 treatment who were divided and allocated into two equal groups randomly: the LED group (LED), treated with infrared LED at 940 nm and conventional therapy, and the control group (CON), who received conventional treatment (antibiotic therapy for preventing superimposed bacterial infections, and physiotherapy) with LED irradiation off. Phototherapy used a vest with an array of 300 LEDs (940 nm) mounted on a 36 cm × 58 cm area and positioned in the patient's anterior thoracic and abdominal regions. The total power was 6 W, with 15 min irradiation time. Cardiopulmonary functions and blood count were monitored before and after treatment. The patients were treated daily for 7 days. Statistical analysis was conducted using a two-tailed unpaired Student's t-test at a significance level of α = 0.05. RESULTS Post-treatment, the LED group showed a reduction in hospital discharge time and a statistically significant improvement for the following cardiopulmonary functions: Partial Oxygen Saturation, Tidal Volume, Maximum Inspiratory, and Expiratory Pressures, Respiratory Frequency, Heart Rate, and Systolic Blood Pressure (p < 0.05). Regarding blood count, it was observed that post-treatment, the LED group presented with significant differences in the count of leukocytes, neutrophils, and lymphocytes. CONCLUSION Photobiomodulation therapy can be used as a complement to conventional treatment of COVID-19, promoting the improvement of cardiopulmonary functions, and minimization of respiratory symptoms.
Collapse
Affiliation(s)
- Pâmela Camila Pereira
- Anhembi Morumbi University (UAM), Institute of Biomedical Engineering, Estrada Dr. Altino Bondensan 500, Distrito de Eugênio de Melo, CEP: 12.247-016 São José dos Campos, SP, Brazil,University Center of Itajubá – (FEPI), Rua Dr. Antônio Braga Filho 687, Bairro Varginha, CEP: 37501-002 Itajubá, MG, Brazil
| | - Carlos José de Lima
- Anhembi Morumbi University (UAM), Institute of Biomedical Engineering, Estrada Dr. Altino Bondensan 500, Distrito de Eugênio de Melo, CEP: 12.247-016 São José dos Campos, SP, Brazil,Center of Innovation, Technology and Education – (CITE), Estrada Dr. Altino Bondensan 500, Distrito de Eugênio de Melo, CEP: 12.247-016 São José dos Campos, SP, Brazil
| | - Adriana Barrinha Fernandes
- Anhembi Morumbi University (UAM), Institute of Biomedical Engineering, Estrada Dr. Altino Bondensan 500, Distrito de Eugênio de Melo, CEP: 12.247-016 São José dos Campos, SP, Brazil,Center of Innovation, Technology and Education – (CITE), Estrada Dr. Altino Bondensan 500, Distrito de Eugênio de Melo, CEP: 12.247-016 São José dos Campos, SP, Brazil
| | - Renato Amaro Zângaro
- Anhembi Morumbi University (UAM), Institute of Biomedical Engineering, Estrada Dr. Altino Bondensan 500, Distrito de Eugênio de Melo, CEP: 12.247-016 São José dos Campos, SP, Brazil,Center of Innovation, Technology and Education – (CITE), Estrada Dr. Altino Bondensan 500, Distrito de Eugênio de Melo, CEP: 12.247-016 São José dos Campos, SP, Brazil
| | - Antonio Balbin Villaverde
- Anhembi Morumbi University (UAM), Institute of Biomedical Engineering, Estrada Dr. Altino Bondensan 500, Distrito de Eugênio de Melo, CEP: 12.247-016 São José dos Campos, SP, Brazil,Center of Innovation, Technology and Education – (CITE), Estrada Dr. Altino Bondensan 500, Distrito de Eugênio de Melo, CEP: 12.247-016 São José dos Campos, SP, Brazil,Corresponding author at: Center of Innovation, Technology and Education – CITE, Estrada Dr. Altino Bondensan 500, Distrito de Eugênio de Melo, CEP: 12.247-016 São José dos Campos, SP, Brazil
| |
Collapse
|
25
|
A simple in-host model for COVID-19 with treatments: model prediction and calibration. J Math Biol 2023; 86:20. [PMID: 36625956 PMCID: PMC9838461 DOI: 10.1007/s00285-022-01849-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/14/2022] [Accepted: 12/01/2022] [Indexed: 01/11/2023]
Abstract
In this paper, we provide a simple ODEs model with a generic nonlinear incidence rate function and incorporate two treatments, blocking the virus binding and inhibiting the virus replication to investigate the impact of calibration on model predictions for the SARS-CoV-2 infection dynamics. We derive conditions of the infection eradication for the long-term dynamics using the basic reproduction number, and complement the characterization of the dynamics at short-time using the resilience and reactivity of the virus-free equilibrium are considered to inform on the average time of recovery and sensitivity to perturbations in the initial virus free stage. Then, we calibrate the treatment model to clinical datasets for viral load in mild and severe cases and immune cells in severe cases. Based on the analysis, the model calibrated to these different datasets predicts distinct scenarios: eradication with a non reactive virus-free equilibrium, eradication with a reactive virus-free equilibrium, and failure of infection eradication. Moreover, severe cases generate richer dynamics and different outcomes with the same treatment. Calibration to different datasets can lead to diverse model predictions, but combining long- and short-term dynamics indicators allows the categorization of model predictions and determination of infection severity.
Collapse
|
26
|
Arcellana AE, Lim KW, Arcegono M, Jimeno C. Critical Illness-Related Corticosteroid Insufficiency (CIRCI) Among Patients with COVID-19 at a Tertiary Hospital: Clinical Characteristics and Outcomes. J ASEAN Fed Endocr Soc 2022; 38:90-99. [PMID: 37252409 PMCID: PMC10213165 DOI: 10.15605/jafes.038.01.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/11/2022] [Indexed: 02/12/2024] Open
Abstract
OBJECTIVES Among critically ill patients, there is usually impairment of the hypothalamic-pituitary-adrenal axis, leading to a condition known as critical illness-related corticosteroid insufficiency (CIRCI). This investigation aims to determine the incidence of and characterize CIRCI among patients with COVID-19 as well as to analyze the outcomes of these critically ill patients. METHODOLOGY This is a single-center, retrospective cohort study that investigated the occurrence of CIRCI among critically ill patients infected with COVID-19. RESULTS In this cohort, there were 145 COVID-19-positive patients with refractory shock, which reflects that 22.94% of the COVID-19 admissions have probable CIRCI.Patients who were given corticosteroids were found to have statistically significant longer median days on a ventilator (p=0.001). However, those on the corticosteroid arm were at higher risk of morbidity and mortality and a greater proportion had organ dysfunction. Multivariable logistic regression analysis revealed that SOFA score was a significant predictor of mortality in CIRCI (p=0.013). CONCLUSION CIRCI has a unique presentation among patients with COVID-19 because of the presence of a high level of inflammation in this life-threatening infection. It is possibly a harbinger of a markedly increased risk of mortality in these patients.
Collapse
Affiliation(s)
- Anna Elvira Arcellana
- Division of Endocrinology, Diabetes and Metabolism, University of the Philippines-Philippine General Hospital
| | - Kenneth Wilson Lim
- Division of Pulmonary Medicine, University of the Philippines-Philippine General Hospital
| | - Marlon Arcegono
- Division of Infectious Diseases, University of the Philippines-Philippine General Hospital
| | - Cecilia Jimeno
- Division of Endocrinology, Diabetes and Metabolism, University of the Philippines-Philippine General Hospital
- Department of Pharmacology and Toxicology, University of the Philippines College of Medicine
| |
Collapse
|
27
|
Blanca D, Nicolosi S, Bandera A, Blasi F, Mantero M, Hu C, de Amicis MM, Lucchi T, Schinco G, Peyvandi F, Gualtierotti R, Fracanzani AL, Lombardi R, Canetta C, Montano N, Beretta L. Comparison between the first and second COVID-19 waves in Internal Medicine wards in Milan, Italy: a retrospective observational study. Intern Emerg Med 2022; 17:2219-2228. [PMID: 35970982 PMCID: PMC9377666 DOI: 10.1007/s11739-022-03052-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/09/2022] [Indexed: 01/08/2023]
Abstract
COVID-19 spread in two pandemic waves in Italy between 2020 and 2021. The aim of this study is to compare the first with the second COVID-19 wave, analyzing modifiable and non-modifiable factors and how these factors affected mortality in patients hospitalized in Internal Medicine wards. Consecutive patients with SARS-CoV-2 infection and dyspnea requiring O2 supplementation were included. The severity of lung involvement was categorized according to the patients' oxygen need. Six hundred and ten SARS-CoV-2 hospitalized patients satisfied the inclusion criteria. The overall estimated 4-week mortality was similar in the two pandemic waves. Several variables were associated with mortality after univariate analysis, but they lacked the significance after multivariable adjustment. Steroids did not exert any protective effect when analyzed in time-dependent models in the whole sample; however, steroids seemed to exert a protective effect in more severe patients. When analyzing the progression to different states of O2 supplementation during hospital stay, mortality was almost exclusively associated with the use of high-flow O2 or CPAP. The analysis of the transition from one state to the other by Cox-Markov models confirmed that age and the severity of lung involvement at admission, along with fever, were relevant factor for mortality or progression.
Collapse
Affiliation(s)
- Deborah Blanca
- Division of Internal Medicine, Immunology and Allergology, Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
| | - Selene Nicolosi
- Division of Internal Medicine, Immunology and Allergology, Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
| | - Alessandra Bandera
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
- Department of Infectious Diseases, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Blasi
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
- Respiratory Unit Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Marco Mantero
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
- Respiratory Unit Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Cinzia Hu
- Department of Internal Medicine, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Tiziano Lucchi
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Giuseppina Schinco
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Flora Peyvandi
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Roberta Gualtierotti
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Anna Ludovica Fracanzani
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
- Unit of Medicine and Metabolic Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Rosa Lombardi
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
- Unit of Medicine and Metabolic Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ciro Canetta
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
- High Care Internal Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Nicola Montano
- Division of Internal Medicine, Immunology and Allergology, Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
| | - Lorenzo Beretta
- Division of Internal Medicine, Immunology and Allergology, Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - on behalf of the Covid Network
- Division of Internal Medicine, Immunology and Allergology, Center for Systemic Autoimmune Diseases, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università Degli Studi di Milano, Milan, Italy
- Department of Infectious Diseases, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Respiratory Unit Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Department of Internal Medicine, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
- Geriatric Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Angelo Bianchi Bonomi Hemophilia and Thrombosis Center, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Unit of Medicine and Metabolic Disease, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- High Care Internal Medicine Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
28
|
Leligdowicz A, Harhay MO, Calfee CS. Immune Modulation in Sepsis, ARDS, and Covid-19 - The Road Traveled and the Road Ahead. NEJM EVIDENCE 2022; 1:EVIDra2200118. [PMID: 38319856 DOI: 10.1056/evidra2200118] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Immune Modulation in Sepsis, ARDS, and Covid-19Leligdowicz et al. consider the history and future of immunomodulating therapies in sepsis and ARDS, including ARDS due to Covid-19, and remark on the larger challenge of clinical research on therapies for syndromes with profound clinical and biologic heterogeneity.
Collapse
Affiliation(s)
- Aleksandra Leligdowicz
- Department of Medicine, Division of Critical Care Medicine, Western University, London, ON, Canada
- Robarts Research Institute, Western University, London, ON, Canada
| | - Michael O Harhay
- Clinical Trials Methods and Outcomes Lab, Palliative and Advanced Illness Research (PAIR) Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Carolyn S Calfee
- Department of Medicine, Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, San Francisco
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco
| |
Collapse
|
29
|
Rossi E, Mutti L, Morrione A, Giordano A. Neuro-Immune Interactions in Severe COVID-19 Infection. Pathogens 2022; 11:1256. [PMID: 36365007 PMCID: PMC9699641 DOI: 10.3390/pathogens11111256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 is a new coronavirus that has affected the world since 2019. Interstitial pneumonia is the most common clinical presentation, but additional symptoms have been reported, including neurological manifestations. Severe forms of infection, especially in elderly patients, present as an excessive inflammatory response called "cytokine storm", which can lead to acute respiratory distress syndrome (ARDS), multiorgan failure and death. Little is known about the relationship between symptoms and clinical outcomes or the characteristics of virus-host interactions. The aim of this narrative review is to highlight possible links between neurological involvement and respiratory damage mediated by pathological inflammatory pathways in SARS-CoV-2 infection. We will focus on neuro-immune interactions and age-related immunity decline and discuss some pathological mechanisms that contribute to negative outcomes in COVID-19 patients. Furthermore, we will describe available therapeutic strategies and their effects on COVID-19 neurological symptoms.
Collapse
Affiliation(s)
- Elena Rossi
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
- Italian Group for Research and Therapy for Mesothelioma (GIMe), 27058 Voghera, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy
| | - Andrea Morrione
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Antonio Giordano
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
30
|
Zhou R, Johnson KE, Rousseau JF, Rathouz PJ. Comparative Effectiveness of Dexamethasone in Treatment of Hospitalized COVID-19 Patients during the First Year of the Pandemic: The N3C Data Repository. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.10.22.22281373. [PMID: 36324806 PMCID: PMC9628188 DOI: 10.1101/2022.10.22.22281373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Dexamethasone, a widely available glucocorticoid, was approved for use in hospitalized COVID-19 patients early in the pandemic based on the RECOVERY trial; however, evidence is still needed to support its real-world effectiveness in patients with a wide range of comorbidities and in diverse care settings. Objectives To conduct a comparative effectiveness analysis of dexamethasone use with and without remdesivir in hospitalized COVID-19 patients using electronic health record data. Methods We conducted a retrospective real-world effectiveness analysis using the harmonized, highly granular electronic health record data of the National COVID Cohort Collaborative (N3C) Data Enclave. Analysis was restricted to COVID-19 patients in an inpatient setting, prior to vaccine availability. Primary outcome was in-hospital death; secondary outcome was combined in-hospital death and severe outcome as defined by use of ECMO or mechanical ventilation during stay. Missing data were imputed with single imputation. Matching of dexamethasone-treated patients to non-dexamethasone-treated controls was accomplished using propensity score (PS) matching, stratified by remdesivir treatment and based on demographics, baseline laboratory values, and comorbidities. Treatment benefit was quantified using logistic regression. Further sensitivity analyses were performed using clinical adjusters in matched groups and in strata defined by quartiles of PS. Results Regression analysis revealed a statistically significant association between dexamethasone use and reduced risk of in-hospital mortality for those not receiving remdesivir (OR=0.77, 95% CI:0.62 to 0.95, p=0.017), and a borderline statistically significant risk for those receiving remdesivir (OR=0.74, 95% CI: 0.53 to 1.02, p=0.054). Treatment also showed secondary outcome benefit. In sensitivity analyses, treatment effect size generally remained similar with some heterogeneity of benefit across strata of PS. Conclusions We add evidence that dexamethasone provides benefit with respect to mortality and severe outcomes in a diverse, national hospitalized sample, prior to vaccine availability.
Collapse
Affiliation(s)
- Richard Zhou
- Department of Biomedical Engineering, University of Texas at Austin
| | - Kaitlyn E. Johnson
- Department of Integrative Biology, The University of Texas at Austin
- The Pandemic Prevention Institute, The Rockefeller Foundation
| | | | | | | |
Collapse
|
31
|
Sadeghi S, Arezoomandi N, Ardestani MM, Ardestani ME, Ghiasi F, Farajzadegan Z. Efficacy and Safety Comparison of Two Different Doses of Dexamethasone in Hospitalized Patients with COVID-19: A Randomized Clinical Trial. J Res Pharm Pract 2022; 11:136-143. [PMID: 37969616 PMCID: PMC10642588 DOI: 10.4103/jrpp.jrpp_42_22] [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: 07/06/2022] [Accepted: 11/01/2022] [Indexed: 11/17/2023] Open
Abstract
Objective The current study aims to investigate high- versus low-dose dexamethasone administration to control the disease with minor complications. Methods The current multicentric randomized clinical trial was conducted on 119 patients with COVID-19 pneumonia and assigned into two groups of low-dose (8 mg daily intravenous dose for at least 7 days or until discharge) (n = 61) versus high-dose dexamethasone (24 mg for 3 days followed by daily 8 mg for the at least 4 days later or until discharge) (n = 58) during 2020-2021. Oxygen saturation, dyspnea severity based on the Borg scale, and laboratory indices were assessed at 3, 5, and 7 days of corticosteroid therapy. Patients were compared regarding the length of hospitalization, intensive care unit (ICU) admission requirement, and noninvasive or invasive ventilation. The other investigations included corticosteroid-related adverse effects and mortality rates within a month after the medications. Findings Oxygen saturation, Borg scale, and C-reactive protein levels were significantly altered by the time in both the groups (P < 0.05). In contrast, the trend of improvements in Borg scale (P = 0.007) and lactate dehydrogenase levels (P = 0.034) were superior in high-dose treated cases. Drug-related adverse (P = 0.809), mortality rate (P = 0.612), hospitalization duration (P = 0.312), ICU admission requirement (P = 0.483), and noninvasive (P = 0.396) and invasive ventilation (P = 0.420) did not differ between the groups. Conclusion According to this study, low- versus high-dose dexamethasone therapy did not affect the outcomes, so low-dose dexamethasone is recommended for COVID-19 pneumonia to achieve optimal results and prevent potential adverse events.
Collapse
Affiliation(s)
- Somayeh Sadeghi
- Department of Internal Medicine, Acquired Immunodeficiency Research Center, Isfahan University of Medical, Isfahan, Iran
| | - Nima Arezoomandi
- Department of Internal Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | | | | | - Farzin Ghiasi
- Department of Internal Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Ziba Farajzadegan
- Department of Community and Family Medicine, Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
32
|
Fredericks AM, Jentzsch MS, Cioffi WG, Cohen M, Fairbrother WG, Gandhi SJ, Harrington EO, Nau GJ, Reichner JS, Ventetuolo CE, Levy MM, Ayala A, Monaghan SF. Deep RNA sequencing of intensive care unit patients with COVID-19. Sci Rep 2022; 12:15755. [PMID: 36130991 PMCID: PMC9491252 DOI: 10.1038/s41598-022-20139-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/09/2022] [Indexed: 02/04/2023] Open
Abstract
COVID-19 has impacted millions of patients across the world. Molecular testing occurring now identifies the presence of the virus at the sampling site: nasopharynx, nares, or oral cavity. RNA sequencing has the potential to establish both the presence of the virus and define the host's response in COVID-19. Single center, prospective study of patients with COVID-19 admitted to the intensive care unit where deep RNA sequencing (> 100 million reads) of peripheral blood with computational biology analysis was done. All patients had positive SARS-CoV-2 PCR. Clinical data was prospectively collected. We enrolled fifteen patients at a single hospital. Patients were critically ill with a mortality of 47% and 67% were on a ventilator. All the patients had the SARS-CoV-2 RNA identified in the blood in addition to RNA from other viruses, bacteria, and archaea. The expression of many immune modulating genes, including PD-L1 and PD-L2, were significantly different in patients who died from COVID-19. Some proteins were influenced by alternative transcription and splicing events, as seen in HLA-C, HLA-E, NRP1 and NRP2. Entropy calculated from alternative RNA splicing and transcription start/end predicted mortality in these patients. Current upper respiratory tract testing for COVID-19 only determines if the virus is present. Deep RNA sequencing with appropriate computational biology may provide important prognostic information and point to therapeutic foci to be precisely targeted in future studies.
Collapse
Affiliation(s)
- Alger M Fredericks
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University/Rhode Island Hospital, 593 Eddy Street, Middle House 211, Providence, RI, 02903, USA
| | - Maximilian S Jentzsch
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University/Rhode Island Hospital, 593 Eddy Street, Middle House 211, Providence, RI, 02903, USA
| | - William G Cioffi
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University/Rhode Island Hospital, 593 Eddy Street, Middle House 211, Providence, RI, 02903, USA
| | - Maya Cohen
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, USA
| | | | | | | | - Gerard J Nau
- Division of Infectious Disease, Department of Medicine, Alpert Medical School of Brown University /Rhode Island Hospital, Providence, USA
| | - Jonathan S Reichner
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University/Rhode Island Hospital, 593 Eddy Street, Middle House 211, Providence, RI, 02903, USA
| | - Corey E Ventetuolo
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, USA
| | - Mitchell M Levy
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, USA
| | - Alfred Ayala
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University/Rhode Island Hospital, 593 Eddy Street, Middle House 211, Providence, RI, 02903, USA
| | - Sean F Monaghan
- Division of Surgical Research, Department of Surgery, Alpert Medical School of Brown University/Rhode Island Hospital, 593 Eddy Street, Middle House 211, Providence, RI, 02903, USA.
| |
Collapse
|
33
|
Hardy E, Fernandez-Patron C. Could Endogenous Glucocorticoids Influence SARS-CoV-2 Infectivity? Cells 2022; 11:cells11192955. [PMID: 36230917 PMCID: PMC9562004 DOI: 10.3390/cells11192955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/10/2022] [Accepted: 09/20/2022] [Indexed: 12/15/2022] Open
Abstract
Endogenous glucocorticoids and their synthetic analogues, such as dexamethasone, stimulate receptor-mediated signal transduction mechanisms on target cells. Some of these mechanisms result in beneficial outcomes whereas others are deleterious in the settings of pathogen infections and immunological disorders. Here, we review recent studies by several groups, including our group, showing that glucocorticoids can directly interact with protein components on SARS-CoV-2, the causative agent of COVID-19. We postulate an antiviral defence mechanism by which endogenous glucocorticoids (e.g., cortisol produced in response to SARS-CoV-2 infection) can bind to multiple sites on SARS-CoV-2 surface protein, Spike, inducing conformational alterations in Spike subunit 1 (S1) that inhibit SARS-CoV-2 interaction with the host SARS-CoV-2 receptor, ACE2. We suggest that glucocorticoids-mediated inhibition of S1 interaction with ACE2 may, consequently, affect SARS-CoV-2 infectivity. Further, glucocorticoids interactions with Spike could protect against a broad spectrum of coronaviruses and their variants that utilize Spike for infection of the host. These notions may be useful for the design of new antivirals for coronavirus diseases.
Collapse
Affiliation(s)
- Eugenio Hardy
- Center of Molecular Immunology, P.O. Box 16040, Havana 11600, Cuba
- Correspondence: (E.H.); (C.F.-P.)
| | - Carlos Fernandez-Patron
- Department of Biochemistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB T6G 2H7, Canada
- Correspondence: (E.H.); (C.F.-P.)
| |
Collapse
|
34
|
Finch K, Sharma D, Wagner S. Lenalidomide induced pneumonitis. J Oncol Pharm Pract 2022:10781552221124533. [PMID: 36071644 DOI: 10.1177/10781552221124533] [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: 11/17/2022]
Abstract
INTRODUCTION Iatrogenic lung injury is a very rare, albeit serious complication with antineoplastic therapy, including immunomodulatory drugs. Pneumonitis typically presents clinically with symptoms such as cough, dyspnea, fever, and hypoxemia. Radiographic evaluation often demonstrates diffuse, patchy infiltrates and ground-glass opacities. CASE REPORT We present a case in which therapy from an immunomodulatory drug, lenalidomide, elicited a pneumonitis in the form of a 9 cm lung mass. An exhaustive workup was completed to rule out viral, bacterial, and fungal infections as well as malignant causes. Lenalidomide-induced lung injury was suspected. MANAGEMENT AND OUTCOME Lenalidomide was discontinued and corticosteroid therapy was initiated. This resulted in a complete clinical and radiographic resolution of symptoms. DISCUSSION Several case reports of pneumonitis have been associated with immunomodulatory drug therapy, and while most of these exhibit diffuse ground-glass opacities radiographically, our patient presented with a 9 cm lung mass. Our findings stress the importance of a thorough medication review while ruling out other potential causes of lung injury.
Collapse
Affiliation(s)
- Kelsey Finch
- Lung Institute, 14479Columbus Regional Health, Columbus, IN, USA
| | - Deepankar Sharma
- Lung Institute, 14479Columbus Regional Health, Columbus, IN, USA
| | - Stephanie Wagner
- Lung Institute, 14479Columbus Regional Health, Columbus, IN, USA
| |
Collapse
|
35
|
Murakami K, Sano H, Tode N, Tsukita Y, Sato K, Narita D, Kimura N, Matsumoto S, Ono Y, Iwasaki C, Sugiyama H, Suzuki M, Kakuto S, Konno S, Kanamori H, Baba H, Oshima K, Takei K, Tokuda K, Tamada T, Sugiura H. Clinical features of COVID-19 patients with rebound phenomenon after corticosteroid therapy. BMJ Open Respir Res 2022. [PMCID: PMC9445231 DOI: 10.1136/bmjresp-2022-001332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Rational Corticosteroid therapy plays a key role in the treatment of COVID-19 patients with respiratory failure. However, a rebound phenomenon after steroid cessation rarely occurs. Here, we investigated the clinical features of patients with rebound after steroid therapy. Methods In total, 84 patients with COVID-19 treated with corticosteroids were enrolled and analysed retrospectively. A rebound was defined as when a patient’s respiratory status deteriorated after the cessation of corticosteroid therapy, without secondary bacterial infection. Results Subjects in the rebound group were more likely to having severe respiratory failure than those in the non-rebound group. While the duration of steroid therapy was longer in the rebound group (8 days vs 10 days, p=0.0009), the dosage of steroid and the timing of the start or termination of steroid therapy did not show any differences between the two groups (p=0.17 and 0.68, respectively). The values of soluble interleukin-2 receptor (sIL-2R) at the baseline and the values of C reactive protein (CRP) or lactate dehydrogenase (LDH) at the end of steroid therapy were significantly higher in the rebound group (937 vs 1336 U/mL; p=0.002, 0.63 vs 3.96 mg/dL; p=0.01 and 278 vs 451 IU/mL; p=0.01, respectively). No patient in the rebound group suffered from thromboses, and the causes of death were exacerbation of COVID-19, ventilator-associated pneumonia or sepsis. The prediction model using baseline features for the rebound phenomenon included four variables of age >68 years, required supplemental oxygen >5 L/min, lymphocyte counts <792 /µL and sIL-2R >1146 U/mL. The discrimination ability of this model was 0.906 (0.755–0.968). Conclusion These findings suggest that severe respiratory failure has a higher risk for the rebound phenomenon after the cessation of corticosteroids, and the values of sIL-2R, LDH and CRP are useful to assess the probability of developing rebound. A multivariate model was developed to predict rebound risk, which showed acceptable discrimination ability.
Collapse
Affiliation(s)
- Koji Murakami
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hirohito Sano
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, Japan
| | - Naoki Tode
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoko Tsukita
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Sato
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Daisuke Narita
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Nozomu Kimura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shuichiro Matsumoto
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoshinao Ono
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chikashi Iwasaki
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hatsumi Sugiyama
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Manami Suzuki
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sho Kakuto
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shuichi Konno
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hajime Kanamori
- Department of Infectious Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Baba
- Department of Infectious Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kengo Oshima
- Department of Infectious Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kentarou Takei
- Department of Infectious Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Koichi Tokuda
- Department of Infectious Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tsutomu Tamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hisatoshi Sugiura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
36
|
Karra N, Fernandes J, Swindle EJ, Morgan H. Integrating an aerosolized drug delivery device with conventional static cultures and a dynamic airway barrier microphysiological system. BIOMICROFLUIDICS 2022; 16:054102. [PMID: 36118260 PMCID: PMC9473724 DOI: 10.1063/5.0100019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
Organ on a chip or microphysiological systems (MPSs) aim to resolve current challenges surrounding drug discovery and development resulting from an unrepresentative static cell culture or animal models that are traditionally used by generating a more physiologically relevant environment. Many different airway MPSs have been developed that mimic alveolar or bronchial interfaces, but few methods for aerosol drug delivery at the air-liquid interface exist. This work demonstrates a compact Surface Acoustic Wave (SAW) drug delivery device that generates an aerosol of respirable size for delivery of compounds directly onto polarized or differentiated epithelial cell cultures within an airway barrier MPS and conventional static inserts. As proof of principle, the SAW drug delivery device was used to nebulize viral dsRNA analog poly I:C and steroids fluticasone and dexamethasone without disrupting their biological function.
Collapse
Affiliation(s)
- Nikita Karra
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, United Kingdom
| | - Joao Fernandes
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, United Kingdom
| | | | - Hywel Morgan
- Author to whom correspondence should be addressed:
| |
Collapse
|
37
|
Laudanski K, Okeke T, Siddiq K, Hajj J, Restrepo M, Gullipalli D, Song WC. A disturbed balance between blood complement protective factors (FH, ApoE) and common pathway effectors (C5a, TCC) in acute COVID-19 and during convalesce. Sci Rep 2022; 12:13658. [PMID: 35953544 PMCID: PMC9366819 DOI: 10.1038/s41598-022-17011-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 07/18/2022] [Indexed: 11/09/2022] Open
Abstract
A complement effect on homeostasis during infection is determined by both cytotoxic (activate complement component 5 (C5a) terminal cytotoxic complex (TCC)), and cytoprotective elements (complement factor H (FH), as well as apolipoprotein E (ApoE)). Here, we investigated the gap in knowledge in their blood milieu during SARS-CoV-2 infection with respect to the viral burden, level of tissue necrosis, and immunological response. 101 patients hospitalized with a PCR-confirmed diagnosis of COVID-19 had blood collected at H1 (48 h), H2 (3-4 Days), H3 (5-7 days), H4 (more than 7 days up to 93 days). Pre-existing conditions, treatment, the incidence of cerebrovascular events (CVA), a history of deep venous thrombosis (DVT) and pulmonary embolism (PE), and mortality was collected using electronic medical records. Plasma C5a, TCC, FH, and ApoE were considered as a complement milieu. Tissue necrosis (HMGB1, RAGE), non-specific inflammatory responses (IL-6, C-reactive protein), overall viral burden (SARS-CoV-2 spike protein), and specific immune responses (IgG, IgA, IgM directed αS- & N-proteins) were assessed simultaneously. C5a remained elevated across all time points, with the peak at 5-7 days. Studied elements of complement coalesced around three clusters: #0 (↑↑↑C5a, ↑↑TCC, ↓↓ApoE), #1 ↑C5a, ↑TCC, ↑↑↑FH); #2 (↑C5a, ↑TCC, ↑FH, ↑↑↑ApoE). The decline in FH and ApoE was a predictor of death, while TCC and C5a correlated with patient length of stay, APACHE, and CRP. Increased levels of C5a (Δ = 122.64; p = 0.0294; data not shown) and diminished levels of FH (Δ = 836,969; p = 0.0285; data not shown) co-existed with CVA incidence. C5a correlated storngly with blood RAGE and HMGB1, but not with viral load and immunological responsiveness. Remdesivir positively affected FH preservation, while convalescent plasma treatment elevated C5a levels. Three clusters of complement activation demonstrated a various milieu of ApoE & FH vs C5a & TCC in COVID-19 patients. Complement activation is linked to increased necrosis markers but not to viral burden or immune system response.
Collapse
Affiliation(s)
- Krzysztof Laudanski
- Department of Anesthesiology and Critical Care, The University of Pennsylvania, JMB 127, 3620 Hamilton Walk, Philadelphia, PA, 19146, USA.
- Leonard Davis Institute for Health Economics, The University of Pennsylvania Colonial Penn Center, 3641 Locust Walk, Philadelphia, PA, 19104, USA.
| | - Tony Okeke
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Kumal Siddiq
- College of Arts and Sciences, Drexel University, Philadelphia, PA, USA
| | - Jihane Hajj
- School of Nursing, Widener College, Chester, PA, USA
| | - Mariana Restrepo
- College of Arts and Sciences, The University of Pennsylvania, Philadelphia, PA, USA
| | - Damodar Gullipalli
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA, USA
| | - Wen-Chao Song
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
38
|
Ochodo EA, Owino E, Nyagol B, Fox T, McCaul M, Kredo T, Cohen K, Rupali P. Molnupiravir for treating COVID-19. Hippokratia 2022. [DOI: 10.1002/14651858.cd015381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Eleanor A Ochodo
- Centre for Global Health Research; Kenya Medical Research Institute; Kisumu Kenya
- Centre for Evidence-based Health Care, Division of Epidemiology and Biostatistics, Faculty of Medicine and Health Sciences; Stellenbosch University; Cape Town South Africa
| | - Eddy Owino
- Centre for Global Health Research; Kenya Medical Research Institute; Kisumu Kenya
| | - Bruce Nyagol
- Centre for Global Health Research; Kenya Medical Research Institute; Kisumu Kenya
| | - Tilly Fox
- Department of Clinical Sciences; Liverpool School of Tropical Medicine; Liverpool UK
| | - Michael McCaul
- Centre for Evidence-based Health Care, Division of Epidemiology and Biostatistics, Faculty of Medicine and Health Sciences; Stellenbosch University; Cape Town South Africa
| | - Tamara Kredo
- Cochrane South Africa; South African Medical Research Council; Cape Town South Africa
| | - Karen Cohen
- Division of Clinical Pharmacology; University of Cape Town; Cape Town South Africa
| | - Priscilla Rupali
- Department of Infectious Diseases; Christian Medical College Vellore; Tamil Nadu India
| |
Collapse
|
39
|
El-Saber Batiha G, Al-Gareeb AI, Saad HM, Al-kuraishy HM. COVID-19 and corticosteroids: a narrative review. Inflammopharmacology 2022; 30:1189-1205. [PMID: 35562628 PMCID: PMC9106274 DOI: 10.1007/s10787-022-00987-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 03/30/2022] [Indexed: 02/06/2023]
Abstract
It has been reported that corticosteroid therapy was effective in the management of severe acute respiratory syndrome (SARS) and the Middle East Respiratory Syndrome (MERS), and recently in coronavirus disease 2019 (COVID-19). Corticosteroids are potent anti-inflammatory drugs that mitigate the risk of acute respiratory distress syndrome (ARDS) in COVID-19 and other viral pneumonia, despite a reduction of viral clearance; corticosteroids inhibit the development of cytokine storm and multi-organ damage. The risk-benefit ratio should be assessed for critical COVID-19 patients. In conclusion, corticosteroid therapy is an effective way in the management of COVID-19, it reduces the risk of complications primarily acute lung injury and the development of ARDS. Besides, corticosteroid therapy mainly dexamethasone and methylprednisolone are effective in reducing the severity of COVID-19 and associated comorbidities such as chronic obstructive pulmonary diseases (COPD), rheumatoid arthritis, and inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 AlBeheira Egypt
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyiah University, Baghdad, Iraq
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, 51744 Matrouh Egypt
| | - Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyiah University, Baghdad, Iraq
| |
Collapse
|
40
|
Chopra M, Joshi A, Dey S, Kapoor R, Nair RK, Bhalla S, Nilakantan A. Effect of treatment regimens in severe COVID pneumonia at an Indian tertiary care hospital: An observational, real-world study. Med J Armed Forces India 2022; 78:469-474. [PMID: 35919732 PMCID: PMC9334214 DOI: 10.1016/j.mjafi.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 05/16/2022] [Indexed: 12/15/2022] Open
Abstract
Background Corticosteroids have attracted attention as a treatment option for severe Coronavirus disease (COVID-19). However, published data on steroid therapy is debatable, and real-world data is lacking. This study evaluated the effect of treatment regimens, especially Pulse steroid therapy (Injection Methyl Prednisolone 250 mg iv once a day for three days) in severe-COVID-19 pneumonia at an Indian tertiary care hospital. Methods This observational cross-sectional study included severe COVID-19 pneumonia patients aged >18 years, requiring assisted ventilation. As part of the hospital protocol, patients received either pulse steroid therapy, remdesivir or tocilizumab in addition to the recommended steroid doses i.e., injection of dexamethasone 6 mg iv once a day. The association of factors and treatment regimens to patient outcomes was evaluated. Results Data of eighty-three patients were assessed, majority being above 60 years (n = 30, 36.14%) and males (n = 45/83, 54.21%). The commonest comorbidities were hypertension (n = 26), diabetes (n = 23) and obesity (n = 19), fifty-five patients (66.26%) reported at least one comorbidity. Sixty-one patients (73.49%) had received pulse steroid regimen, forty-eight patients (57.83%) were administered remdesivir-based regimen while twelve patients (14.46%) had received tocilizumab treatment. 54.1% patients managed with pulse steroid regimens were discharged after treatment, statistically similar to remdesivir-managed subgroup (62.5%, p > 0.05). On sub-group analysis, pulse steroids showed better outcomes in young males with no comorbidities. No comorbidity had significant relationship with patient outcomes (p > 0.05). Conclusion Pulse steroid therapy is an effective therapy in management of patients with severe COVID-19 pneumonia in a real-world setting, with better outcomes in young males without comorbidities. Pulse steroids can be considered a viable option for severe-COVID-19 pneumonia management.
Collapse
|
41
|
Zhou F, Deng J, Heybati K, Zuo QK, Ali S, Hou W, Wong CY, Ramaraju HB, Chang O, Dhivagaran T, Silver Z. Efficacy and safety of corticosteroid regimens for the treatment of hospitalized COVID-19 patients: a meta-analysis. Future Virol 2022; 17:463-489. [PMID: 35814934 PMCID: PMC9249165 DOI: 10.2217/fvl-2021-0244] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 04/08/2022] [Indexed: 12/15/2022]
Abstract
Aim To evaluate the efficacy and safety of corticosteroids for treating hospitalized COVID-19 patients. Materials & methods Efficacy outcomes included time to negative SARS-CoV-2 tests, length of stay, duration and incidence of intensive unit care stay, incidence of mortality and duration and incidence of mechanical ventilation. Safety outcomes included the incidence of adverse events and severe adverse events, incidence of hyperglycemia and incidence of nosocomial infections. Results Ninety-five randomized controlled trials (RCTs) and observational studies (n = 42,205) were included. Corticosteroids were associated with increased length of stay (based on RCT only), increased time to negative tests, decreased length of mechanical ventilation and increased odds of hyperglycemia. Conclusion Corticosteroids should be considered in patients requiring mechanical ventilation, and glycemic monitoring may be needed when administering corticosteroids.
Collapse
Affiliation(s)
- Fangwen Zhou
- Faculty of Health Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Jiawen Deng
- Faculty of Health Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Kiyan Heybati
- Mayo Clinic Alix School of Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905, USA
| | - Qi Kang Zuo
- Department of Anesthesiology, Rutgers, New Jersey Medical School, 185 S Orange Ave, Newark, NJ 07103, USA
- Faculty of Science, McGill University, 845 Sherbrooke St W, Montreal, QC, H3A 0G5, Canada
| | - Saif Ali
- Faculty of Health Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Wenteng Hou
- Faculty of Health Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Chi Yi Wong
- Faculty of Health Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | | | - Oswin Chang
- Faculty of Health Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Thanansayan Dhivagaran
- Faculty of Health Sciences, McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
- Integrated Biomedical Engineering & Health Sciences Program (iBioMed), McMaster University, 1280 Main St W, Hamilton, ON, L8S 4L8, Canada
| | - Zachary Silver
- Faculty of Science, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| |
Collapse
|
42
|
Wong EKC, Watt J, Zou H, Chandraraj A, Zhang AW, Brookes J, Verduyn A, Berall A, Norman R, Piggott KL, Izukawa T, Straus SE, Liu B. Characteristics, treatment and delirium incidence of older adults hospitalized with COVID-19: a multicentre retrospective cohort study. CMAJ Open 2022; 10:E692-E701. [PMID: 35882392 PMCID: PMC9334012 DOI: 10.9778/cmajo.20210176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The COVID-19 pandemic has affected older adults disproportionately, and delirium is a concerning consequence; however, the relationship between delirium and corticosteroid use is uncertain. The objective of the present study was to describe patient characteristics, treatments and outcomes among older adults hospitalized with COVID-19, with a focus on dexamethasone use and delirium incidence. METHODS We completed this retrospective cohort study at 7 sites (including acute care, rehabilitation and long-term care settings) in Toronto, Ontario, Canada. We included adults aged 65 years or older, consecutively hospitalized with confirmed SARS-CoV-2 infection, between Mar. 11, 2020, and Apr. 30, 2021. We abstracted patient characteristics and outcomes from charts and analyzed them descriptively. We used a logistic regression model to determine the association between dexamethasone use and delirium incidence. RESULTS During the study period, 927 patients were admitted to the acute care hospitals with COVID-19. Patients' median age was 79.0 years (interquartile range [IQR] 72.0-87.0), and 417 (45.0%) were female. Most patients were frail (61.9%), based on a Clinical Frailty Scale score of 5 or greater. The prevalence of delirium was 53.6%, and the incidence was 33.1%. Use of restraints was documented in 20.4% of patients. In rehabilitation and long-term care settings (n = 115), patients' median age was 86.0 years (IQR 78.5-91.0), 72 (62.6%) were female and delirium occurred in 17 patients (14.8%). In patients admitted to acute care during wave 2 of the pandemic (Aug. 1, 2020, to Feb. 20, 2021), dexamethasone use had a nonsignificant association with delirium incidence (adjusted odds ratio 1.38, 95% confidence interval 0.77-2.50). Overall, in-hospital death occurred in 262 (28.4%) patients in acute care settings and 28 (24.3%) patients in rehabilitation or long-term care settings. INTERPRETATION In-hospital death, delirium and use of restraints were common in older adults admitted to hospital with COVID-19. Further research should be directed to improving the quality of care for this population with known vulnerabilities during continued waves of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Eric Kai-Chung Wong
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Jennifer Watt
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Hanyan Zou
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Arthana Chandraraj
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Alissa Wenyue Zhang
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Jahnel Brookes
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Ashley Verduyn
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Anna Berall
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Richard Norman
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Katrina Lynn Piggott
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Terumi Izukawa
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Sharon E Straus
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont
| | - Barbara Liu
- Li Ka Shing Knowledge Institute (Wong, Watt, Chandraraj, Straus), and Division of Geriatric Medicine (Wong, Watt, Straus), Department of Medicine, St. Michael's Hospital, Unity Health Toronto; Division of Geriatric Medicine (Wong, Watt, Norman, Piggott, Izukawa, Straus, Liu), Department of Medicine, University of Toronto; Division of Geriatric Medicine (Zou, Norman), Department of Medicine, Sinai Health and University Health Network; Division of Geriatric Medicine (Zhang, Piggott), Department of Medicine, Sunnybrook Health Sciences Centre; Kunin-Lunenfeld Centre for Applied Research & Evaluation (Brookes, Berall, Izukawa), Rotman Research Institute, Baycrest Health Sciences Centre; Providence Healthcare and Houses of Providence (Verduyn), Unity Health Toronto; Division of Geriatric Medicine (Izukawa), Department of Medicine, Baycrest Health Sciences Centre, Toronto, Ont.
| |
Collapse
|
43
|
Sarker H, Panigrahi R, Hardy E, Glover JNM, Elahi S, Fernandez-Patron C. Glucocorticoids Bind to SARS-CoV-2 S1 at Multiple Sites Causing Cooperative Inhibition of SARS-CoV-2 S1 Interaction With ACE2. Front Immunol 2022; 13:906687. [PMID: 35784352 PMCID: PMC9242398 DOI: 10.3389/fimmu.2022.906687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/12/2022] [Indexed: 12/15/2022] Open
Abstract
Dexamethasone may reduce mortality in COVID-19 patients. Whether dexamethasone or endogenous glucocorticoids, such as cortisol, biochemically interact with SARS-CoV-2 spike 1 protein (S1), or its cellular receptor ACE2, is unknown. Using molecular dynamics (MD) simulations and binding energy calculations, we identified 162 druggable pockets in various conformational states of S1 and all possible binding pockets for cortisol and dexamethasone. Through biochemical binding studies, we confirmed that cortisol and dexamethasone bind to S1. Limited proteolysis and mass spectrometry analyses validated several MD identified binding pockets for cortisol and dexamethasone on S1. Interaction assays indicated that cortisol and dexamethasone separately and cooperatively disrupt S1 interaction with ACE2, through direct binding to S1, without affecting ACE2 catalytic activity. Cortisol disrupted the binding of the mutant S1 Beta variant (E484K, K417N, N501Y) to ACE2. Delta and Omicron variants are mutated in or near identified cortisol-binding pockets in S1, which may affect cortisol binding to them. In the presence of cortisol, we find increased inhibition of S1 binding to ACE2 by an anti-SARS-CoV-2 S1 human chimeric monoclonal antibody against the receptor binding domain. Whether glucocorticoid/S1 direct interaction is an innate defence mechanism that may have contributed to mild or asymptomatic SARS-CoV-2 infection deserves further investigation.
Collapse
Affiliation(s)
- Hassan Sarker
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Rashmi Panigrahi
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | | | - J. N. Mark Glover
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shokrollah Elahi
- Department of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Carlos Fernandez-Patron
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Carlos Fernandez-Patron,
| |
Collapse
|
44
|
Aşut Ö, Çalı Ş. Therapeutic Interventions Implemented During the First Year of the COVID-19 Pandemic: A Systematic Review of Evidence. CYPRUS JOURNAL OF MEDICAL SCIENCES 2022. [DOI: 10.4274/cjms.2021.2021-168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
45
|
Pathogenesis of pneumonia and acute lung injury. Clin Sci (Lond) 2022; 136:747-769. [PMID: 35621124 DOI: 10.1042/cs20210879] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 12/15/2022]
Abstract
Pneumonia and its sequelae, acute lung injury, present unique challenges for pulmonary and critical care healthcare professionals, and these challenges have recently garnered global attention due to the ongoing Sars-CoV-2 pandemic. One limitation to translational investigation of acute lung injury, including its most severe manifestation (acute respiratory distress syndrome, ARDS) has been heterogeneity resulting from the clinical and physiologic diagnosis that represents a wide variety of etiologies. Recent efforts have improved our understanding and approach to heterogeneity by defining sub-phenotypes of ARDS although significant gaps in knowledge remain. Improving our mechanistic understanding of acute lung injury and its most common cause, infectious pneumonia, can advance our approach to precision targeted clinical interventions. Here, we review the pathogenesis of pneumonia and acute lung injury, including how respiratory infections and lung injury disrupt lung homoeostasis, and provide an overview of respiratory microbial pathogenesis, the lung microbiome, and interventions that have been demonstrated to improve outcomes-or not-in human clinical trials.
Collapse
|
46
|
Wang Y, Wu M, Li Y, Yuen HH, He ML. The effects of SARS-CoV-2 infection on modulating innate immunity and strategies of combating inflammatory response for COVID-19 therapy. J Biomed Sci 2022; 29:27. [PMID: 35505345 PMCID: PMC9063252 DOI: 10.1186/s12929-022-00811-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/27/2022] [Indexed: 12/15/2022] Open
Abstract
The global pandemic of COVID-19 has caused huge causality and unquantifiable loss of social wealth. The innate immune response is the first line of defense against SARS-CoV-2 infection. However, strong inflammatory response associated with dysregulation of innate immunity causes severe acute respiratory syndrome (SARS) and death. In this review, we update the current knowledge on how SARS-CoV-2 modulates the host innate immune response for its evasion from host defense and its corresponding pathogenesis caused by cytokine storm. We emphasize Type I interferon response and the strategies of evading innate immune defense used by SARS-CoV-2. We also extensively discuss the cells and their function involved in the innate immune response and inflammatory response, as well as the promises and challenges of drugs targeting excessive inflammation for antiviral treatment. This review would help us to figure out the current challenge questions of SARS-CoV-2 infection on innate immunity and directions for future studies.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Mandi Wu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Yichen Li
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Ho Him Yuen
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China. .,CityU Shenzhen Research Institute, Nanshan, Shenzhen, China.
| |
Collapse
|
47
|
Ngai HW, Kim DH, Hammad M, Gutova M, Aboody K, Cox CD. Stem Cell-based therapies for COVID-19-related acute respiratory distress syndrome. J Cell Mol Med 2022; 26:2483-2504. [PMID: 35426198 PMCID: PMC9077311 DOI: 10.1111/jcmm.17265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
As the number of confirmed cases and resulting death toll of the COVID-19 pandemic continue to increase around the globe - especially with the emergence of new mutations of the SARS-CoV-2 virus in addition to the known alpha, beta, gamma, delta and omicron variants - tremendous efforts continue to be dedicated to the development of interventive therapeutics to mitigate infective symptoms or post-viral sequelae in individuals for which vaccines are not accessible, viable or effective in the prevention of illness. Many of these investigations aim to target the associated acute respiratory distress syndrome, or ARDS, which induces damage to lung epithelia and other physiologic systems and is associated with progression in severe cases. Recently, stem cell-based therapies have demonstrated preliminary efficacy against ARDS based on a number of preclinical and preliminary human safety studies, and based on promising outcomes are now being evaluated in phase II clinical trials for ARDS. A number of candidate stem cell therapies have been found to exhibit low immunogenicity, coupled with inherent tropism to injury sites. In recent studies, these have demonstrated the ability to modulate suppression of pro-inflammatory cytokine signals such as those characterizing COVID-19-associated ARDS. Present translational studies are aiming to optimize the safety, efficacy and delivery to fully validate stem cell-based strategies targeting COVID-19 associated ARDS for viable clinical application.
Collapse
Affiliation(s)
- Hoi Wa Ngai
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Dae Hong Kim
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Mohamed Hammad
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Margarita Gutova
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Karen Aboody
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | | |
Collapse
|
48
|
Menchén DA, Vázquez JB, Allende JMB, García GH. [Viral pneumonia. COVID-19 pneumonia]. Medicine (Baltimore) 2022; 13:3224-3234. [PMID: 35582693 PMCID: PMC9097969 DOI: 10.1016/j.med.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Viruses are becoming more and more important as etiological agents of pneumonia, mainly due to improvements in diagnostic techniques. At present, they account for approximately one-third of community-acquired pneumonia cases. They tend to occur in children and elderly patients, causing anything from mild cases to severe cases which require intubation and intensive care. The main causative agents include rhinovirus, influenza virus A or B, metapneumovirus, respiratory syncytial virus, parainfluenza virus, coronavirus, or adenovirus. Infection produced by SARS-CoV-2, the cause of the current COVID-19 pandemic, which has had devastating consequences, is proof of the vital importance of viral pneumonia. This is the main subject of this update. Drawing on the abundant information available, which has been continuously evolving since the beginning of the pandemic, this section will review the main characteristics of the virus, its pathophysiology, and its clinical manifestations as well as the main diagnostic and treatment methods.
Collapse
Affiliation(s)
- D Alonso Menchén
- Servicio de Medicina Interna. Hospital Universitario Príncipe de Asturias. Universidad de Alcalá. Alcalá de Henares. Madrid. España
| | - J Balsa Vázquez
- Servicio de Medicina Interna. Hospital Universitario Príncipe de Asturias. Universidad de Alcalá. Alcalá de Henares. Madrid. España
| | - J M Barbero Allende
- Servicio de Medicina Interna. Hospital Universitario Príncipe de Asturias. Universidad de Alcalá. Alcalá de Henares. Madrid. España
| | - G Hernández García
- Servicio de Medicina Interna. Hospital Universitario Príncipe de Asturias. Universidad de Alcalá. Alcalá de Henares. Madrid. España
| |
Collapse
|
49
|
Consolaro E, Suter F, Rubis N, Pedroni S, Moroni C, Pastò E, Paganini MV, Pravettoni G, Cantarelli U, Perico N, Perna A, Peracchi T, Ruggenenti P, Remuzzi G. A Home-Treatment Algorithm Based on Anti-inflammatory Drugs to Prevent Hospitalization of Patients With Early COVID-19: A Matched-Cohort Study (COVER 2). Front Med (Lausanne) 2022; 9:785785. [PMID: 35530041 PMCID: PMC9073076 DOI: 10.3389/fmed.2022.785785] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/18/2022] [Indexed: 12/15/2022] Open
Abstract
Background and AimWhile considerable success has been achieved in the management of patients hospitalized with severe coronavirus disease 2019 (COVID-19), far less progress has been made with early outpatient treatment. We assessed whether the implementation of a home treatment algorithm—designed based on a pathophysiologic and pharmacologic rationale—and including non-steroidal anti-inflammatory drugs, especially relatively selective cyclooxygenase-2 inhibitors and, when needed, corticosteroids, anticoagulants, oxygen therapy and antibiotics—at the very onset of mild COVID-19 symptoms could effectively reduce hospital admissions.MethodsThis fully academic, matched-cohort study evaluated outcomes in 108 consecutive consenting patients with mild COVID-19, managed at home by their family doctors between January 2021 and May 2021, according to the proposed treatment algorithm and in 108 age-, sex-, and comorbidities-matched patients on other therapeutic schedules (ClinicalTrials.gov: NCT04854824). The primary outcome was COVID-19-related hospitalization. Analyses were by intention-to-treat.ResultsOne (0.9%) patient in the “recommended” cohort and 12 (11.1%) in the “control” cohort were admitted to hospital (P = 0.0136). The proposed algorithm reduced the cumulative length of hospital stays by 85% (from 141 to 19 days) as well as related costs (from €60.316 to €9.058). Only 9.8 patients needed to be treated with the recommended algorithm to prevent one hospitalization event. The rate of resolution of major symptoms was numerically—but not significantly—higher in the “recommended” than in the “control” cohort (97.2 vs. 93.5%, respectively; P = 0.322). Other symptoms lingered in a smaller proportion of patients in the “recommended” than in the “control” cohort (20.4 vs. 63.9%, respectively; P < 0.001), and for a shorter period.ConclusionThe adoption of the proposed outpatient treatment algorithm during the early, mild phase of COVID-19 reduced the incidence of subsequent hospitalization and related costs.
Collapse
Affiliation(s)
- Elena Consolaro
- Agenzia di Tutela della Salute (ATS) Insubria, Varese, Italy
| | - Fredy Suter
- Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Nadia Rubis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
- *Correspondence: Nadia Rubis
| | | | - Chiara Moroni
- Agenzia di Tutela della Salute (ATS) Insubria, Varese, Italy
| | - Elena Pastò
- Agenzia di Tutela della Salute (ATS) Insubria, Varese, Italy
| | | | | | | | - Norberto Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Annalisa Perna
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Tobia Peracchi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Piero Ruggenenti
- Azienda Socio-Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
50
|
Lidou-Renault V, Baudouin E, Courtois-Amiot P, Bianco C, Esnault H, Rouet A, Baque M, Tomeo C, Rainone A, Thietart S, Veber R, Ayache C, Pepin M, Lafuente-Lafuente C, Duron E, Cailleaux PE, Haguenauer D, Lemarié N, Paillaud E, Raynaud-Simon A, Thomas C, Boddaert J, Zerah L, Vallet H. Corticosteroid therapy in COVID-19 associated with in-hospital mortality in geriatric patients: a propensity matched cohort study. J Gerontol A Biol Sci Med Sci 2022; 77:1352-1360. [PMID: 35395678 PMCID: PMC9129112 DOI: 10.1093/gerona/glac084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Indexed: 12/15/2022] Open
Abstract
Background Few data are available on the prognosis of older patients who received corticosteroids for COVID-19. We aimed to compare the in-hospital mortality of geriatric patients hospitalized for COVID-19 who received corticosteroids or not. Methods We conducted a multicentric retrospective cohort study in 15 acute COVID-19 geriatric wards in the Paris area from March to April 2020 and November 2020 to May 2021. We included all consecutive patients aged 70 years and older who were hospitalized with confirmed COVID-19 in these wards. Propensity score and multivariate analyses were used. Results Of the 1 579 patients included (535 received corticosteroids), the median age was 86 (interquartile range 81–91) years, 56% of patients were female, the median Charlson Comorbidity Index (CCI) was 2.6 (interquartile range 1–4), and 64% of patients were frail (Clinical Frailty Score 5–9). The propensity score analysis paired 984 patients (492 with and without corticosteroids). The in-hospital mortality was 32.3% in the matched cohort. On multivariate analysis, the probability of in-hospital mortality was increased with corticosteroid use (odds ratio [OR] = 2.61 [95% confidence interval (CI) 1.63–4.20]). Other factors associated with in-hospital mortality were age (OR = 1.04 [1.01–1.07], CCI (OR = 1.18 [1.07–1.29], activities of daily living (OR = 0.85 [0.75–0.95], oxygen saturation < 90% on room air (OR = 2.15 [1.45–3.17], C-reactive protein level (OR = 2.06 [1.69–2.51], and lowest lymphocyte count (OR = 0.49 [0.38–0.63]). Among the 535 patients who received corticosteroids, 68.3% had at least one corticosteroid side effect, including delirium (32.9%), secondary infections (32.7%), and decompensated diabetes (14.4%). Conclusions In this multicentric matched-cohort study of geriatric patients hospitalized for COVID-19, the use of corticosteroids was significantly associated with in-hospital mortality.
Collapse
Affiliation(s)
| | | | - Pauline Courtois-Amiot
- Assistance Publique-Hôpitaux de Paris (APHP), Université de Paris, Hôpital Bichat, Department of Geriatric Medicine, Paris, France
| | - Celine Bianco
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Saint Antoine, Department of Geriatric Medicine, Paris, France
| | - Hélène Esnault
- Assistance Publique-Hôpitaux de Paris (APHP), Université de Paris, Hôpital Bichat, Department of Geriatric Medicine, Paris, France
| | - Audrey Rouet
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Tenon, Department of Geriatric Medicine, Paris, France
| | - Margaux Baque
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Saint Antoine, Department of Geriatric Medicine, Paris, France
- Sorbonne Université, INSERM UMR1135, Centre d’immunologie et des Maladies Infectieuses, Paris, France
| | - Charlotte Tomeo
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Rothschild, Department of Geriatric Medicine, Paris, France
| | - Antonio Rainone
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Charles Foix, Department of Geriatric Medicine, Ivry Sur Seine, France
| | - Sara Thietart
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Pitié Salpêtrière, Department of Geriatric Medicine, Paris, France
| | - Romain Veber
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Rothschild, Department of Geriatric Medicine, Paris, France
| | - Clementine Ayache
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Rothschild, Department of Geriatric Medicine, Paris, France
| | - Marion Pepin
- Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Ambroise Paré, Department of Geriatric Medicine, Boulogne, Billancourt, France
- Université de Versailles Saint-Quentin en Yvelines, Université Paris-Saclay, INSERM, CESP, Clinical Epidemiology, Villejuif, France
| | - Carmelo Lafuente-Lafuente
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Charles Foix, Department of Geriatric Medicine, Ivry Sur Seine, France
| | - Emmanuelle Duron
- Assistance Publique-Hôpitaux de Paris (APHP), University hospital of Paris-Saclay, Department of Geriatric Medicine, Paul Brousse Hospital, Villejuif, France
- Université Paris-Saclay, INSERM 1178, CESP, Équipe MOODS, Le Kremlin-Bicêtre, France
| | - Pierre-Emmanuel Cailleaux
- Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Louis Mourier, Department of Geriatric Medicine, Colombes, France
| | - Didier Haguenauer
- Assistance Publique-Hôpitaux de Paris (APHP), Hôpital Louis Mourier, Department of Geriatric Medicine, Colombes, France
| | - Nadège Lemarié
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Tenon, Department of Geriatric Medicine, Paris, France
| | - Elena Paillaud
- Assistance Publique-Hôpitaux de Paris (APHP), Université de Paris, Paris Cancer Institute CARPEM, Department of Geriatric Medicine, Hôpital Européen Georges Pompidou, Paris, France
| | - Agathe Raynaud-Simon
- Assistance Publique-Hôpitaux de Paris (APHP), Université de Paris, Hôpital Bichat, Department of Geriatric Medicine, Paris, France
| | - Caroline Thomas
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Saint Antoine, Department of Geriatric Medicine, Paris, France
| | - Jacques Boddaert
- Sorbonne Université, INSERM UMR1135, Centre d’immunologie et des Maladies Infectieuses, Paris, France
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Pitié Salpêtrière, Department of Geriatric Medicine, Paris, France
| | - Lorène Zerah
- Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Pitié Salpêtrière, Department of Geriatric Medicine, Paris, France
- Sorbonne Université, INSERM, Institut Pierre Louis d’Épidémiologie et de Santé Publique, IPLESP, Paris, France
| | - Hélène Vallet
- Address correspondence to: Hélène Vallet, MD, PhD, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne Université, Hôpital Saint Antoine, Department of Geriatric Medicine, 184 rue du Faubourg Saint Antoine, Paris 75012, France. E-mail:
| |
Collapse
|