1
|
Hamidi F, Hamishehkar H, Azari Markid PP, Sarbakhsh P. Identifying factors related to mortality of hospitalized COVID-19 patients using machine learning methods. Heliyon 2024; 10:e35561. [PMID: 39170355 PMCID: PMC11337730 DOI: 10.1016/j.heliyon.2024.e35561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
Background The COVID-19 pandemic has had a profound impact globally, presenting significant social and economic challenges. This study aims to explore the factors affecting mortality among hospitalized COVID-19 patients and construct a machine learning-based model to predict the risk of mortality. Methods The study examined COVID-19 patients admitted to Imam Reza Hospital in Tabriz, Iran, between March 2020 and November 2021. The Elastic Net method was employed to identify and rank features associated with mortality risk. Subsequently, an artificial neural network (ANN) model was developed based on these features to predict mortality risk. The performance of the model was evaluated by receiver operating characteristic (ROC) curve analysis. Results The study included 706 patients with 96 features, out of them 26 features were identified as crucial predictors of mortality. The ANN model, utilizing 20 of these features, achieved an area under the ROC curve (AUC) of 98.8 %, effectively stratifying patients by mortality risk. Conclusion The developed model offers accurate and precipitous mortality risk predictions for COVID-19 patients, enhancing the responsiveness of healthcare systems to high-risk individuals.
Collapse
Affiliation(s)
- Farzaneh Hamidi
- Department of Biostatistics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hadi Hamishehkar
- Clinical Research Development Unit of Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Parvin Sarbakhsh
- Health and Environment Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
2
|
Structure and activity of human TMPRSS2 protease implicated in SARS-CoV-2 activation. Nat Chem Biol 2022; 18:963-971. [PMID: 35676539 DOI: 10.1038/s41589-022-01059-7] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 05/09/2022] [Indexed: 02/07/2023]
Abstract
Transmembrane protease, serine 2 (TMPRSS2) has been identified as key host cell factor for viral entry and pathogenesis of SARS-CoV-2. Specifically, TMPRSS2 proteolytically processes the SARS-CoV-2 Spike (S) protein, enabling virus-host membrane fusion and infection of the airways. We present here a recombinant production strategy for enzymatically active TMPRSS2 and characterization of its matured proteolytic activity, as well as its 1.95 Å X-ray cocrystal structure with the synthetic protease inhibitor nafamostat. Our study provides a structural basis for the potent but nonspecific inhibition by nafamostat and identifies distinguishing features of the TMPRSS2 substrate binding pocket that explain specificity. TMPRSS2 cleaved SARS-CoV-2 S protein at multiple sites, including the canonical S1/S2 cleavage site. We ranked the potency of clinical protease inhibitors with half-maximal inhibitory concentrations ranging from 1.4 nM to 120 µM and determined inhibitor mechanisms of action, providing the groundwork for drug development efforts to selectively inhibit TMPRSS2.
Collapse
|
3
|
Sakamuru S, Huang R, Xia M. Use of Tox21 Screening Data to Evaluate the COVID-19 Drug Candidates for Their Potential Toxic Effects and Related Pathways. Front Pharmacol 2022; 13:935399. [PMID: 35910344 PMCID: PMC9333127 DOI: 10.3389/fphar.2022.935399] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/16/2022] [Indexed: 12/15/2022] Open
Abstract
Currently, various potential therapeutic agents for coronavirus disease-2019 (COVID-19), a global pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), are being investigated worldwide mainly through the drug repurposing approach. Several anti-viral, anti-bacterial, anti-malarial, and anti-inflammatory drugs were employed in randomized trials and observational studies for developing new therapeutics for COVID-19. Although an increasing number of repurposed drugs have shown anti-SARS-CoV-2 activities in vitro, so far only remdesivir has been approved by the US FDA to treat COVID-19, and several other drugs approved for Emergency Use Authorization, including sotrovimab, tocilizumab, baricitinib, paxlovid, molnupiravir, and other potential strategies to develop safe and effective therapeutics for SARS-CoV-2 infection are still underway. Many drugs employed as anti-viral may exert unwanted side effects (i.e., toxicity) via unknown mechanisms. To quickly assess these drugs for their potential toxicological effects and mechanisms, we used the Tox21 in vitro assay datasets generated from screening ∼10,000 compounds consisting of approved drugs and environmental chemicals against multiple cellular targets and pathways. Here we summarize the toxicological profiles of small molecule drugs that are currently under clinical trials for the treatment of COVID-19 based on their in vitro activities against various targets and cellular signaling pathways.
Collapse
|
4
|
Electrodeposition of Cobalt Oxides on Carbon Nanotubes for Sensitive Bromhexine Sensing. Molecules 2022; 27:molecules27134078. [PMID: 35807327 PMCID: PMC9268198 DOI: 10.3390/molecules27134078] [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: 05/25/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 11/25/2022] Open
Abstract
We develop an electrochemical sensor for the determination of bromhexine hydrochloride (BHC), a widely use mucolytic drug. The sensor is prepared by electrodeposition of cobalt oxides (CoOx) on a glassy carbon electrode modified with carboxylated single-walled carbon nanotubes (SWCNT). A synergistic effect between CoOx and SWCNT is observed, leading to a significant improvement in the BHC electrooxidation current. Based on cyclic voltammetry studies at varying scan rates, we conclude that the electrochemical oxidation of BHC is under mixed diffusion–adsorption control. The proposed sensor allows the amperometric determination of BHC in a linear range of 10–500 µM with a low applied voltage of 0.75 V. The designed sensor provides reproducible measurements, is not affected by common interfering substances, and shows excellent performance for the analysis of BHC in pharmaceutical preparations.
Collapse
|
5
|
Gorący A, Rosik J, Szostak B, Ustianowski Ł, Ustianowska K, Gorący J. Human Cell Organelles in SARS-CoV-2 Infection: An Up-to-Date Overview. Viruses 2022; 14:v14051092. [PMID: 35632833 PMCID: PMC9144443 DOI: 10.3390/v14051092] [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: 03/31/2022] [Revised: 05/10/2022] [Accepted: 05/17/2022] [Indexed: 12/10/2022] Open
Abstract
Since the end of 2019, the whole world has been struggling with the life-threatening pandemic amongst all age groups and geographic areas caused by Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). The Coronavirus Disease 2019 (COVID-19) pandemic, which has led to more than 468 million cases and over 6 million deaths reported worldwide (as of 20 March 2022), is one of the greatest threats to human health in history. Meanwhile, the lack of specific and irresistible treatment modalities provoked concentrated efforts in scientists around the world. Various mechanisms of cell entry and cellular dysfunction were initially proclaimed. Especially, mitochondria and cell membrane are crucial for the course of infection. The SARS-CoV-2 invasion depends on angiotensin converting enzyme 2 (ACE2), transmembrane serine protease 2 (TMPRSS2), and cluster of differentiation 147 (CD147), expressed on host cells. Moreover, in this narrative review, we aim to discuss other cell organelles targeted by SARS-CoV-2. Lastly, we briefly summarize the studies on various drugs.
Collapse
Affiliation(s)
- Anna Gorący
- Independent Laboratory of Invasive Cardiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (A.G.); (J.G.)
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University, 70-204 Szczecin, Poland
| | - Jakub Rosik
- Independent Laboratory of Invasive Cardiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (A.G.); (J.G.)
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (B.S.); (Ł.U.); (K.U.)
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Correspondence:
| | - Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (B.S.); (Ł.U.); (K.U.)
| | - Łukasz Ustianowski
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (B.S.); (Ł.U.); (K.U.)
| | - Klaudia Ustianowska
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (B.S.); (Ł.U.); (K.U.)
| | - Jarosław Gorący
- Independent Laboratory of Invasive Cardiology, Pomeranian Medical University, 70-204 Szczecin, Poland; (A.G.); (J.G.)
| |
Collapse
|
6
|
Das K, Patil A, Goren A, Cockerell CJ, Goldust M. Androgens and COVID-19. J Cosmet Dermatol 2022; 21:3176-3180. [PMID: 35576054 PMCID: PMC9348029 DOI: 10.1111/jocd.15090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022]
Abstract
Background The humans have been disproportionately affected by the coronavirus disease (COVID‐19) pandemic. The novel coronavirus or the severe acute respiratory syndrome coronavirus 2 (SARS‐COV2) causing coronavirus disease (COVID‐19) has spread across the globe. Androgens have been suggested to have a role in COVID‐19 pathogenesis. Objective The objective of this review article is to study the link between androgens and COVID‐19. Methodology PubMed and Google Scholar search was performed to retrieve literature related to the topic. Review articles, clinical trials, retrospective studies, observational studies, and case–control studies were considered for the review. Results Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infected men are more inclined to be hospitalized for intensive care unit (ICU) as compared with women. This difference in the ICU admissions provides some clue for possible influence of androgens in the severity of COVID‐19. The contribution of androgen and androgen receptor in COVID‐19 disease and its severity, as well as the numerous medications targeting androgen and its receptor for lowering COVID‐19 disease severity, are discussed in this review. Available literature suggests the role of androgen in the pathogenesis and severity of COVID‐19. Sensitivity for androgen may be an important factor in regulating the severity of COVID‐19 disease. Conclusion There is a scope for the development of COVID‐19 treatments based on androgen suppression. Clinical trials may furnish pivotal data and add more evidence‐based options for the management of COVID‐19.
Collapse
Affiliation(s)
- Kinnor Das
- Consultant Dermatolgist, Apollo clinic, Silchar, Assam, India
| | - Anant Patil
- Department of Pharmacology, Dr. DY Patil Medical College, Navi Mumbai, India
| | - Andy Goren
- University of Rome G. Marconi, Rome, Italy
| | - Clay J Cockerell
- Departments of Dermatology and Pathology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Cockerell Dermatopathology, Dallas, Texas, USA
| | - Mohamad Goldust
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
7
|
Bahadoram S, Keikhaei B, Bahadoram M, Mahmoudian-Sani MR, Hassanzadeh S, Saeedi-Boroujeni A, Alikhani K. [Bromhexine is a potential drug for COVID-19; From hypothesis to clinical trials]. Vopr Virusol 2022; 67:126-132. [PMID: 35521985 DOI: 10.36233/0507-4088-106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Indexed: 11/05/2022]
Abstract
COVID-19 (novel coronavirus disease 2019), caused by the SARS-CoV-2 virus, has various clinical manifestations and several pathogenic pathways. Although several therapeutic options have been used to control COVID-19, none of these medications have been proven to be a definitive cure. Transmembrane serine protease 2 (TMPRSS2) is a protease that has a key role in the entry of SARS-CoV-2 into host cells. Following the binding of the viral spike (S) protein to the angiotensin-converting enzyme 2 (ACE2) receptors of the host cells, TMPRSS2 processes and activates the S protein on the epithelial cells. As a result, the membranes of the virus and host cell fuse. Bromhexine is a specific TMPRSS2 inhibitor that potentially inhibits the infectivity cycle of SARS-CoV-2. Moreover, several clinical trials are evaluating the efficacy of bromhexine in COVID-19 patients. The findings of these studies have shown that bromhexine is effective in improving the clinical outcomes of COVID-19 and has prophylactic effects by inhibiting TMPRSS2 and viral penetration into the host cells. Bromhexine alone cannot cure all of the symptoms of SARS-CoV-2 infection. However, it could be an effective addition to control and prevent the disease progression along with other drugs that are used to treat COVID-19. Further studies are required to investigate the efficacy of bromhexine in COVID-19.
Collapse
Affiliation(s)
- S Bahadoram
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences
| | - B Keikhaei
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences
| | - M Bahadoram
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences
| | - M-R Mahmoudian-Sani
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences
| | - S Hassanzadeh
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences
| | - A Saeedi-Boroujeni
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences; Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences; Abadan University of Medical Sciences;ImmunologyToday, Universal Scientific Education and Research Network (USERN)
| | - K Alikhani
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences
| |
Collapse
|
8
|
Mantzourani C, Vasilakaki S, Gerogianni VE, Kokotos G. The discovery and development of transmembrane serine protease 2 (TMPRSS2) inhibitors as candidate drugs for the treatment of COVID-19. Expert Opin Drug Discov 2022; 17:231-246. [PMID: 35072549 PMCID: PMC8862169 DOI: 10.1080/17460441.2022.2029843] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has caused the devastating pandemic named coronavirus disease 2019 (COVID-19). Unfortunately, the discovery of antiviral agents to combat COVID-19 is still an unmet need. Transmembrane serine protease 2 (TMPRSS2) is an important mediator in viral infection and thus, TMPRRS2 inhibitors may be attractive agents for COVID-19 treatment. AREAS COVERED This review article discusses the role of TMPRSS2 in SARS-CoV-2 cell entry and summarizes the inhibitors of TMPRSS2 and their potential anti-SARS activity. Two known TMPRSS2 inhibitors, namely camostat and nafamostat, approved drugs for the treatment of pancreatitis, are under clinical trials as potential drugs against COVID-19. EXPERT OPINION Due to the lack of the crystal structure of TMPRSS2, homology models have been developed to study the interactions of known inhibitors, including repurposed drugs, with the enzyme. However, novel TMPRSS2 inhibitors have been identified through high-throughput screening, and appropriate assays studying their in vitro activity have been set up. The discovery of TMPRSS2's crystal structure will facilitate the rational design of novel inhibitors and in vivo studies and clinical trials will give a clear answer if TMPRSS2 inhibitors could be a new weapon against COVID-19.
Collapse
Affiliation(s)
- Christiana Mantzourani
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Vasilakaki
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Athens, Greece
| | - Velisaria-Eleni Gerogianni
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Athens, Greece
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
9
|
Bromhexine Hydrochloride Prophylaxis of COVID-19 for Medical Personnel: A Randomized Open-Label Study. Interdiscip Perspect Infect Dis 2022; 2022:4693121. [PMID: 35103059 PMCID: PMC8799951 DOI: 10.1155/2022/4693121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/13/2022] [Indexed: 12/23/2022] Open
Abstract
Background. Bromhexine hydrochloride has been suggested as a TMPRSS2 protease blocker that precludes the penetration of SARS-CoV-2 into cells. We aimed to assess the preventive potential of regular bromhexine hydrochloride intake for COVID-19 risk reduction in medical staff actively involved in the evaluation and treatment of patients with confirmed or suspected SARS-CoV-2 infection. Methods. In a single-centre randomized open-label study, medical staff managing patients with suspected and confirmed COVID-19 were enrolled and followed up for 8 weeks. The study began at the initiation of COVID-19 management in the clinic. The study was prematurely terminated after the enrollment of 50 participants without a history of SARS-CoV-2 infection: 25 were assigned to bromhexine hydrochloride treatment (8 mg 3 times per day), and 25 were controls. The composite primary endpoint was a positive nasopharyngeal swab polymerase chain reaction (PCR) test for SARS-CoV-2 or signs of clinical infection within 28 days and at week 8. Secondary endpoints included time from the first contact with a person with COVID-19 to the appearance of respiratory infection symptoms; the number of days before a first positive SARS-CoV-2 test; the number of asymptomatic participants with a positive nasopharyngeal swab test; the number of symptomatic COVID-19 cases; and adverse events. Results. The rate of the combined primary endpoint did not differ significantly between the active treatment group (2/25 [8%]) and control group (7/25 [28%]);
. A fewer number of participants developed symptomatic COVID-19 in the treatment group compared to controls (0/25 vs. 5/25;
). Conclusion. Although the study was underpowered, it showed that Bromhexine hydrochloride prophylaxis was associated with a reduced rate of symptomatic COVID-19. The prophylactic treatment was not associated with a lower combined primary endpoint rate, a positive swab PCR test, or COVID-19 (ClinicalTrials.gov number, NCT04405999).
Collapse
|
10
|
Wettstein L, Kirchhoff F, Münch J. The Transmembrane Protease TMPRSS2 as a Therapeutic Target for COVID-19 Treatment. Int J Mol Sci 2022; 23:1351. [PMID: 35163273 PMCID: PMC8836196 DOI: 10.3390/ijms23031351] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 01/25/2023] Open
Abstract
TMPRSS2 is a type II transmembrane protease with broad expression in epithelial cells of the respiratory and gastrointestinal tract, the prostate, and other organs. Although the physiological role of TMPRSS2 remains largely elusive, several endogenous substrates have been identified. TMPRSS2 serves as a major cofactor in SARS-CoV-2 entry, and primes glycoproteins of other respiratory viruses as well. Consequently, inhibiting TMPRSS2 activity is a promising strategy to block viral infection. In this review, we provide an overview of the role of TMPRSS2 in the entry processes of different respiratory viruses. We then review the different classes of TMPRSS2 inhibitors and their clinical development, with a focus on COVID-19 treatment.
Collapse
Affiliation(s)
| | | | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (L.W.); (F.K.)
| |
Collapse
|
11
|
Law MF, Ho R, Law KWT, Cheung CKM. Gastrointestinal and hepatic side effects of potential treatment for COVID-19 and vaccination in patients with chronic liver diseases. World J Hepatol 2021; 13:1850-1874. [PMID: 35069994 PMCID: PMC8727202 DOI: 10.4254/wjh.v13.i12.1850] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/20/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) is a global pandemic. Many clinical trials have been performed to investigate potential treatments or vaccines for this disease to reduce the high morbidity and mortality. The drugs of higher interest include umifenovir, bromhexine, remdesivir, lopinavir/ritonavir, steroid, tocilizumab, interferon alpha or beta, ribavirin, fivapiravir, nitazoxanide, ivermectin, molnupiravir, hydroxychloroquine/chloroquine alone or in combination with azithromycin, and baricitinib. Gastrointestinal (GI) symptoms and liver dysfunction are frequently seen in patients with COVID-19, which can make it difficult to differentiate disease manifestations from treatment adverse effects. GI symptoms of COVID-19 include anorexia, dyspepsia, nausea, vomiting, diarrhea and abdominal pain. Liver injury can be a result of systemic inflammation or cytokine storm, or due to the adverse drug effects in patients who have been receiving different treatments. Regular monitoring of liver function should be performed. COVID-19 vaccines have been rapidly developed with different technologies including mRNA, viral vectors, inactivated viruses, recombinant DNA, protein subunits and live attenuated viruses. Patients with chronic liver disease or inflammatory bowel disease and liver transplant recipients are encouraged to receive vaccination as the benefits outweigh the risks. Vaccination against COVID-19 is also recommended to family members and healthcare professionals caring for these patients to reduce exposure to the severe acute respiratory syndrome coronavirus 2 virus.
Collapse
Affiliation(s)
- Man Fai Law
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
| | - Rita Ho
- Department of Medicine, North District Hospital, Hong Kong, China
| | | | - Carmen Ka Man Cheung
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
12
|
Honarmand K, Penn J, Agarwal A, Siemieniuk R, Brignardello-Petersen R, Bartoszko JJ, Zeraatkar D, Agoritsas T, Burns K, Fernando SM, Foroutan F, Ge L, Lamontagne F, Jimenez-Mora MA, Murthy S, Yepes-Nuñez JJ, Vandvik PO, Ye Z, Rochwerg B. Clinical trials in COVID-19 management & prevention: A meta-epidemiological study examining methodological quality. J Clin Epidemiol 2021; 139:68-79. [PMID: 34274489 PMCID: PMC8280397 DOI: 10.1016/j.jclinepi.2021.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/16/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To describe the characteristics of Covid-19 randomized clinical trials (RCTs) and examine the association between trial characteristics and the likelihood of finding a significant effect. STUDY DESIGN We conducted a systematic review to identify RCTs (up to October 21, 2020) evaluating drugs or blood products to treat or prevent Covid-19. We extracted trial characteristics (number of centers, funding sources, and sample size) and assessed risk of bias (RoB) using the Cochrane RoB 2.0 tool. We performed logistic regressions to evaluate the association between RoB due to randomization, single vs. multicentre, funding source, and sample size, and finding a statistically significant effect. RESULTS We included 91 RCTs (n = 46,802); 40 (44%) were single-center, 23 (25.3%) enrolled <50 patients, 28 (30.8%) received industry funding, and 75 (82.4%) had high or probably high RoB. Thirty-eight trials (41.8%) reported a statistically significant effect. RoB due to randomization and being a single-center trial were associated with increased odds of finding a statistically significant effect. CONCLUSIONS There is high variability in RoB among Covid-19 trials. Researchers, funders, and knowledge-users should be cognizant of the impact of RoB due to randomization and single-center trial status in designing, evaluating, and interpreting the results of RCTs. REGISTRATION CRD42020192095.
Collapse
Affiliation(s)
- Kimia Honarmand
- Division of Critical Care, Department of Medicine, Western University, 1151 Richmond Street London, Ontario, N6A 3K7, Canada.
| | - Jeremy Penn
- Faculty of Health Sciences, McMaster University, 1280 Main St. West, Hamilton, Ontario, L8S 4L8, Canada
| | - Arnav Agarwal
- Department of Health Research Methods, Evidence and Impact, McMaster University, 1280 Main St. West, Hamilton, Ontario, L8S 4L8, Canada; Department of Medicine, University of Toronto, 27 King's College Circle, Toronto, Ontario, M5S 1A1, Canada
| | - Reed Siemieniuk
- Department of Health Research Methods, Evidence and Impact, McMaster University, 1280 Main St. West, Hamilton, Ontario, L8S 4L8, Canada
| | - Romina Brignardello-Petersen
- Department of Health Research Methods, Evidence and Impact, McMaster University, 1280 Main St. West, Hamilton, Ontario, L8S 4L8, Canada
| | - Jessica J Bartoszko
- Department of Health Research Methods, Evidence and Impact, McMaster University, 1280 Main St. West, Hamilton, Ontario, L8S 4L8, Canada
| | - Dena Zeraatkar
- Department of Health Research Methods, Evidence and Impact, McMaster University, 1280 Main St. West, Hamilton, Ontario, L8S 4L8, Canada; Department of Biomedical Informatics, Harvard Medical School, Boston, 25 Shattuck Street, Boston, MA, 02115, USA
| | - Thomas Agoritsas
- Department of Health Research Methods, Evidence and Impact, McMaster University, 1280 Main St. West, Hamilton, Ontario, L8S 4L8, Canada; Division General Internal Medicine, University Hospitals of Geneva, Rue Gabrielle-Perret-Gentil 4 1205, Geneva, Switzerland
| | - Karen Burns
- Department of Health Research Methods, Evidence and Impact, McMaster University, 1280 Main St. West, Hamilton, Ontario, L8S 4L8, Canada; Unity Health Toronto, St. Michael's Hospital, Li Ka Shing Knowledge Institute, 30 Bond St, Toronto, Ontario, M5B 1W8, Canada
| | - Shannon M Fernando
- Division of Critical Care, Department of Medicine, University of Ottawa, 75 Laurier Ave. E, Ottawa, Ontario, K1N 6N5, Canada
| | - Farid Foroutan
- Ted Rogers Centre for Heart Research, University Health Network, Toronto General Hospital, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Long Ge
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, 222 Tianshui S Rd, Chengguan District, Lanzhou, Gansu, China
| | - Francois Lamontagne
- Department of Medicine and Centre de recherche du CHU de Sherbrooke, 12e Avenue N Porte 6, Sherbrooke, Quebec, J1H 5N4, Canada
| | - Mario A Jimenez-Mora
- School of Medicine, Universidad de los Andes, Cra. 1 #18a-12, Bogotá D.C, Colombia
| | - Srinivas Murthy
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Juan Jose Yepes-Nuñez
- School of Medicine, Universidad de los Andes, Cra. 1 #18a-12, Bogotá D.C, Colombia; Pulmonology Service, Internal Medicine Section, Fundación Santa Fe de Bogotá University Hospital, Cra. 7b (#)12390, Bogotá D.C, Colombia
| | - Per O Vandvik
- Department of Health and Society, Faculty of Medicine, University of Oslo, Problemveien 7, 0315, Oslo, Norway
| | - Zhikang Ye
- Department of Health Research Methods, Evidence and Impact, McMaster University, 1280 Main St. West, Hamilton, Ontario, L8S 4L8, Canada
| | - Bram Rochwerg
- Department of Health Research Methods, Evidence and Impact, McMaster University, 1280 Main St. West, Hamilton, Ontario, L8S 4L8, Canada; Department of Medicine, McMaster University, 1280 Main St. West, Hamilton, Ontario, L8S 4L8, Canada
| |
Collapse
|
13
|
Gillot C, Favresse J, Mullier F, Lecompte T, Dogné JM, Douxfils J. NETosis and the Immune System in COVID-19: Mechanisms and Potential Treatments. Front Pharmacol 2021; 12:708302. [PMID: 34421600 PMCID: PMC8376580 DOI: 10.3389/fphar.2021.708302] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
NETosis is a form of neutrophil death leading to the release of extracellular chromatin and the assembling of proteins, including antiviral proteins, primed by an initial pathogenic stimulus. Under certain specific conditions, neutrophils can exhibit a double-edged activity. This event has been implicated in COVID-19 among other conditions. Neutrophil extracellular traps (NETs) are involved in the pathogenesis of COVID-19 by promoting a pro-inflammatory and a procoagulant state leading to multiorgan failure. This particular form of host defense promoted by neutrophils is closely related to the well-known cytokine storm observed in severe COVID-19 patients. These two elements therefore represent possible targets for treatment of severe SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Constant Gillot
- Department of Pharmacy, University of Namur, Namur Research Institute for Life Sciences, Namur Thrombosis and Hemostasis Center, Namur, Belgium
| | - Julien Favresse
- Department of Pharmacy, University of Namur, Namur Research Institute for Life Sciences, Namur Thrombosis and Hemostasis Center, Namur, Belgium
- Department of Laboratory Medicine, Clinique St-Luc Bouge, Namur, Belgium
| | - François Mullier
- Laboratory Hematology, Université Catholique de Louvain, CHU UCL Namur, Namur Research Institute for Life Sciences (NARILIS), Namur Thrombosis and Haemostasis Centre (NTHC), Yvoir, Belgium
| | - Thomas Lecompte
- Division of Angiology and Haemostasis, University Hospitals of Geneva, Geneva, Switzerland
| | - Jean-Michel Dogné
- Department of Pharmacy, University of Namur, Namur Research Institute for Life Sciences, Namur Thrombosis and Hemostasis Center, Namur, Belgium
| | - Jonathan Douxfils
- Department of Pharmacy, University of Namur, Namur Research Institute for Life Sciences, Namur Thrombosis and Hemostasis Center, Namur, Belgium
- Qualiblood s.a., Namur, Belgium
| |
Collapse
|
14
|
Ben-Zuk N, Dechtman ID, Henn I, Weiss L, Afriat A, Krasner E, Gal Y. Potential Prophylactic Treatments for COVID-19. Viruses 2021; 13:1292. [PMID: 34372498 PMCID: PMC8310088 DOI: 10.3390/v13071292] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 01/08/2023] Open
Abstract
The World Health Organization declared the SARS-CoV-2 outbreak a Public Health Emergency of International Concern at the end of January 2020 and a pandemic two months later. The virus primarily spreads between humans via respiratory droplets, and is the causative agent of Coronavirus Disease 2019 (COVID-19), which can vary in severity, from asymptomatic or mild disease (the vast majority of the cases) to respiratory failure, multi-organ failure, and death. Recently, several vaccines were approved for emergency use against SARS-CoV-2. However, their worldwide availability is acutely limited, and therefore, SARS-CoV-2 is still expected to cause significant morbidity and mortality in the upcoming year. Hence, additional countermeasures are needed, particularly pharmaceutical drugs that are widely accessible, safe, scalable, and affordable. In this comprehensive review, we target the prophylactic arena, focusing on small-molecule candidates. In order to consolidate a potential list of such medications, which were categorized as either antivirals, repurposed drugs, or miscellaneous, a thorough screening for relevant clinical trials was conducted. A brief molecular and/or clinical background is provided for each potential drug, rationalizing its prophylactic use as an antiviral or inflammatory modulator. Drug safety profiles are discussed, and current medical indications and research status regarding their relevance to COVID-19 are shortly reviewed. In the near future, a significant body of information regarding the effectiveness of drugs being clinically studied for COVID-19 is expected to accumulate, in addition to information regarding the efficacy of prophylactic treatments.
Collapse
Affiliation(s)
- Noam Ben-Zuk
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel-Aviv 61909, Israel; (N.B.-Z.); (I.H.); (L.W.)
| | - Ido-David Dechtman
- The Israel Defense Force Medical Corps, Tel Hashomer, Military Post 02149, Israel;
- Pulmonology Department, Edith Wolfson Medical Center, 62 Halochamim Street, Holon 5822012, Israel
| | - Itai Henn
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel-Aviv 61909, Israel; (N.B.-Z.); (I.H.); (L.W.)
| | - Libby Weiss
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel-Aviv 61909, Israel; (N.B.-Z.); (I.H.); (L.W.)
| | - Amichay Afriat
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel;
| | - Esther Krasner
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel-Aviv 61909, Israel; (N.B.-Z.); (I.H.); (L.W.)
| | - Yoav Gal
- Chemical, Biological, Radiological and Nuclear Defense Division, Ministry of Defense, HaKirya, Tel-Aviv 61909, Israel; (N.B.-Z.); (I.H.); (L.W.)
- Israel Institute for Biological Research, Ness-Ziona 76100, Israel
| |
Collapse
|
15
|
Sgrignani J, Cavalli A. Computational Identification of a Putative Allosteric Binding Pocket in TMPRSS2. Front Mol Biosci 2021; 8:666626. [PMID: 33996911 PMCID: PMC8119889 DOI: 10.3389/fmolb.2021.666626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/01/2021] [Indexed: 12/23/2022] Open
Abstract
Camostat, nafamostat, and bromhexine are inhibitors of the transmembrane serine protease TMPRSS2. The inhibition of TMPRSS2 has been shown to prevent the viral infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other viruses. However, while camostat and nafamostat inhibit TMPRSS2 by forming a covalent adduct, the mode of action of bromhexine remains unclear. TMPRSS2 is autocatalytically activated from its inactive form, zymogen, through a proteolytic cleavage that promotes the binding of Ile256 to a putative allosteric pocket (A-pocket). Computer simulations, reported here, indicate that Ile256 binding induces a conformational change in the catalytic site, thus providing the atomistic rationale to the activation process of the enzyme. Furthermore, computational docking and molecular dynamics simulations indicate that bromhexine competes with the N-terminal Ile256 for the same binding site, making it a potential allosteric inhibitor. Taken together, these findings provide the atomistic basis for the development of more selective and potent TMPRSS2 inhibitors.
Collapse
Affiliation(s)
- Jacopo Sgrignani
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
16
|
Eco-friendly UPLC-MS/MS analysis of possible add-on therapy for COVID-19 in human plasma: Insights of greenness assessment. Microchem J 2021; 166:106234. [PMID: 33824542 PMCID: PMC8016544 DOI: 10.1016/j.microc.2021.106234] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/23/2021] [Accepted: 03/29/2021] [Indexed: 12/23/2022]
Abstract
Facing the pandemic COVID-19 is of highest priority for all researchers nowadays. Recent statistics indicate that the majority of the cases are home-treated. Two drugs of interest, Guaifenesin and Bromohexine HCl, are among the add-on therapy for treatment of COVID-19 mild cases, which has raised the need for their simultaneous determination. The analysis of the two drugs of interest was described using ultra-performance liquid chromatography-tandem mass spectrometric (UPLC-MS/MS) in plasma of healthy human volunteers using tetryzoline HCl as an internal standard (IS) after liquid-liquid extraction. The applied chromatographic conditions were Kinetex C18 (100 Å, 2.6 µm X 50 mm X 4.6 mm) column and a mixture of methanol: water (95: 5, v/v) as a mobile phase at flow rate 1 mL/min. The positive ionization mode was used for detecting the ions, by observing the pairs of transition m/z 199 < 125 for GUF, m/z 377 < 114 for BRM and m/z 201 < 131 for IS. The linearity range was from 50 to 1500 ng/mL for GUF and 0.5-50 µg/mL for BRM. Limit of detection (LOD) was found to be 35.16 and 0.43 ng/ml for GUF and BRM, respectively. The method was validated according to FDA guidance. The proposed method was assessed to be more eco-friendly versus the reported method using the greenness assessment tools: National Environmental Methods Index (NEMI), Assessment of Green Profile (AGP), Green Analytical Procedure Index (GAPI) and Eco-Scale. The proposed method was applied for the application of a pilot pharmacokinetic study.
Collapse
|
17
|
Welte T, Ambrose LJ, Sibbring GC, Sheikh S, Müllerová H, Sabir I. Current evidence for COVID-19 therapies: a systematic literature review. Eur Respir Rev 2021; 30:30/159/200384. [PMID: 33731328 PMCID: PMC9489065 DOI: 10.1183/16000617.0384-2020] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/10/2021] [Indexed: 01/09/2023] Open
Abstract
Effective therapeutic interventions for the treatment and prevention of coronavirus disease 2019 (COVID-19) are urgently needed. A systematic review was conducted to identify clinical trials of pharmacological interventions for COVID-19 published between 1 December 2019 and 14 October 2020. Data regarding efficacy of interventions, in terms of mortality, hospitalisation and need for ventilation, were extracted from identified studies and synthesised qualitatively. In total, 42 clinical trials were included. Interventions assessed included antiviral, mucolytic, antimalarial, anti-inflammatory and immunomodulatory therapies. Some reductions in mortality, hospitalisation and need for ventilation were seen with interferons and remdesivir, particularly when administered early, and with the mucolytic drug, bromhexine. Most studies of lopinavir/ritonavir and hydroxychloroquine did not show significant efficacy over standard care/placebo. Dexamethasone significantly reduced mortality, hospitalisation and need for ventilation versus standard care, particularly in patients with severe disease. Evidence for other classes of interventions was limited. Many trials had a moderate-to-high risk of bias, particularly in terms of blinding; most were short-term and some included low patient numbers.This review highlights the need for well-designed clinical trials of therapeutic interventions for COVID-19 to increase the quality of available evidence. It also emphasises the importance of tailoring interventions to disease stage and severity for maximum efficacy.
Collapse
Affiliation(s)
- Tobias Welte
- Dept of Pulmonary and Infectious Diseases, Hannover University School of Medicine, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Juul S, Nielsen EE, Feinberg J, Siddiqui F, Jørgensen CK, Barot E, Holgersson J, Nielsen N, Bentzer P, Veroniki AA, Thabane L, Bu F, Klingenberg S, Gluud C, Jakobsen JC. Interventions for treatment of COVID-19: Second edition of a living systematic review with meta-analyses and trial sequential analyses (The LIVING Project). PLoS One 2021; 16:e0248132. [PMID: 33705495 PMCID: PMC7954033 DOI: 10.1371/journal.pone.0248132] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/22/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND COVID-19 is a rapidly spreading disease that has caused extensive burden to individuals, families, countries, and the world. Effective treatments of COVID-19 are urgently needed. This is the second edition of a living systematic review of randomized clinical trials assessing the effects of all treatment interventions for participants in all age groups with COVID-19. METHODS AND FINDINGS We planned to conduct aggregate data meta-analyses, trial sequential analyses, network meta-analysis, and individual patient data meta-analyses. Our systematic review was based on PRISMA and Cochrane guidelines, and our eight-step procedure for better validation of clinical significance of meta-analysis results. We performed both fixed-effect and random-effects meta-analyses. Primary outcomes were all-cause mortality and serious adverse events. Secondary outcomes were admission to intensive care, mechanical ventilation, renal replacement therapy, quality of life, and non-serious adverse events. According to the number of outcome comparisons, we adjusted our threshold for significance to p = 0.033. We used GRADE to assess the certainty of evidence. We searched relevant databases and websites for published and unpublished trials until November 2, 2020. Two reviewers independently extracted data and assessed trial methodology. We included 82 randomized clinical trials enrolling a total of 40,249 participants. 81 out of 82 trials were at overall high risk of bias. Meta-analyses showed no evidence of a difference between corticosteroids versus control on all-cause mortality (risk ratio [RR] 0.89; 95% confidence interval [CI] 0.79 to 1.00; p = 0.05; I2 = 23.1%; eight trials; very low certainty), on serious adverse events (RR 0.89; 95% CI 0.80 to 0.99; p = 0.04; I2 = 39.1%; eight trials; very low certainty), and on mechanical ventilation (RR 0.86; 95% CI 0.55 to 1.33; p = 0.49; I2 = 55.3%; two trials; very low certainty). The fixed-effect meta-analyses showed indications of beneficial effects. Trial sequential analyses showed that the required information size for all three analyses was not reached. Meta-analysis (RR 0.93; 95% CI 0.82 to 1.07; p = 0.31; I2 = 0%; four trials; moderate certainty) and trial sequential analysis (boundary for futility crossed) showed that we could reject that remdesivir versus control reduced the risk of death by 20%. Meta-analysis (RR 0.82; 95% CI 0.68 to 1.00; p = 0.05; I2 = 38.9%; four trials; very low certainty) and trial sequential analysis (required information size not reached) showed no evidence of difference between remdesivir versus control on serious adverse events. Fixed-effect meta-analysis showed indications of a beneficial effect of remdesivir on serious adverse events. Meta-analysis (RR 0.40; 95% CI 0.19 to 0.87; p = 0.02; I2 = 0%; two trials; very low certainty) showed evidence of a beneficial effect of intravenous immunoglobulin versus control on all-cause mortality, but trial sequential analysis (required information size not reached) showed that the result was severely underpowered to confirm or reject realistic intervention effects. Meta-analysis (RR 0.63; 95% CI 0.35 to 1.14; p = 0.12; I2 = 77.4%; five trials; very low certainty) and trial sequential analysis (required information size not reached) showed no evidence of a difference between tocilizumab versus control on serious adverse events. Fixed-effect meta-analysis showed indications of a beneficial effect of tocilizumab on serious adverse events. Meta-analysis (RR 0.70; 95% CI 0.51 to 0.96; p = 0.02; I2 = 0%; three trials; very low certainty) showed evidence of a beneficial effect of tocilizumab versus control on mechanical ventilation, but trial sequential analysis (required information size not reached) showed that the result was severely underpowered to confirm of reject realistic intervention effects. Meta-analysis (RR 0.32; 95% CI 0.15 to 0.69; p < 0.00; I2 = 0%; two trials; very low certainty) showed evidence of a beneficial effect of bromhexine versus standard care on non-serious adverse events, but trial sequential analysis (required information size not reached) showed that the result was severely underpowered to confirm or reject realistic intervention effects. Meta-analyses and trial sequential analyses (boundary for futility crossed) showed that we could reject that hydroxychloroquine versus control reduced the risk of death and serious adverse events by 20%. Meta-analyses and trial sequential analyses (boundary for futility crossed) showed that we could reject that lopinavir-ritonavir versus control reduced the risk of death, serious adverse events, and mechanical ventilation by 20%. All remaining outcome comparisons showed that we did not have enough information to confirm or reject realistic intervention effects. Nine single trials showed statistically significant results on our outcomes, but were underpowered to confirm or reject realistic intervention effects. Due to lack of data, it was not relevant to perform network meta-analysis or possible to perform individual patient data meta-analyses. CONCLUSIONS No evidence-based treatment for COVID-19 currently exists. Very low certainty evidence indicates that corticosteroids might reduce the risk of death, serious adverse events, and mechanical ventilation; that remdesivir might reduce the risk of serious adverse events; that intravenous immunoglobin might reduce the risk of death and serious adverse events; that tocilizumab might reduce the risk of serious adverse events and mechanical ventilation; and that bromhexine might reduce the risk of non-serious adverse events. More trials with low risks of bias and random errors are urgently needed. This review will continuously inform best practice in treatment and clinical research of COVID-19. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42020178787.
Collapse
Affiliation(s)
- Sophie Juul
- Copenhagen Trial Unit–Centre for Clinical Intervention Research,
Rigshospitalet, Copenhagen University Hospital, Copenhagen,
Denmark
| | - Emil Eik Nielsen
- Copenhagen Trial Unit–Centre for Clinical Intervention Research,
Rigshospitalet, Copenhagen University Hospital, Copenhagen,
Denmark
- Department of Internal Medicine–Cardiology Section, Holbæk Hospital,
Holbæk, Denmark
| | - Joshua Feinberg
- Copenhagen Trial Unit–Centre for Clinical Intervention Research,
Rigshospitalet, Copenhagen University Hospital, Copenhagen,
Denmark
| | - Faiza Siddiqui
- Copenhagen Trial Unit–Centre for Clinical Intervention Research,
Rigshospitalet, Copenhagen University Hospital, Copenhagen,
Denmark
| | - Caroline Kamp Jørgensen
- Copenhagen Trial Unit–Centre for Clinical Intervention Research,
Rigshospitalet, Copenhagen University Hospital, Copenhagen,
Denmark
| | - Emily Barot
- Copenhagen Trial Unit–Centre for Clinical Intervention Research,
Rigshospitalet, Copenhagen University Hospital, Copenhagen,
Denmark
| | - Johan Holgersson
- Department of Clinical Sciences Lund, Anesthesia & Intensive Care,
Helsingborg Hospital, Lund University, Lund, Sweden
| | - Niklas Nielsen
- Department of Clinical Sciences Lund, Anesthesia & Intensive Care,
Helsingborg Hospital, Lund University, Lund, Sweden
| | - Peter Bentzer
- Department of Clinical Sciences Lund, Anesthesia & Intensive Care,
Helsingborg Hospital, Lund University, Lund, Sweden
| | - Areti Angeliki Veroniki
- Department of Primary Education, School of Education, University of
Ioannina, Ioannina, Greece
- Knowledge Translation Program, Li Ka Shing Knowledge Institute, St.
Michael’s Hospital, Toronto, Ontario, Canada
| | - Lehana Thabane
- Department of Health Research Methods, Evidence, and Impact, McMaster
University, Hamilton, ON, Canada
| | - Fanlong Bu
- Centre for Evidence-based Chinese Medicine, Beijing University of Chinese
Medicine, Beijing, China
| | - Sarah Klingenberg
- Copenhagen Trial Unit–Centre for Clinical Intervention Research,
Rigshospitalet, Copenhagen University Hospital, Copenhagen,
Denmark
| | - Christian Gluud
- Copenhagen Trial Unit–Centre for Clinical Intervention Research,
Rigshospitalet, Copenhagen University Hospital, Copenhagen,
Denmark
| | - Janus Christian Jakobsen
- Copenhagen Trial Unit–Centre for Clinical Intervention Research,
Rigshospitalet, Copenhagen University Hospital, Copenhagen,
Denmark
- Faculty of Health Sciences, University of Southern Denmark, Odense,
Denmark
| |
Collapse
|
19
|
El Amri C. Serine Protease Inhibitors to Treat Lung Inflammatory Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:215-226. [PMID: 34019272 DOI: 10.1007/978-3-030-68748-9_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lung is a vital organ that ensures breathing function. It provides the essential interface of air filtering providing oxygen to the whole body and eliminating carbon dioxide in the blood; because of its exposure to the external environment, it is fall prey to many exogenous elements, such as pathogens, especially viral infections or environmental toxins and chemicals. These exogenous actors in addition to intrinsic disorders lead to important inflammatory responses that compromise lung tissue and normal functioning. Serine proteases regulating inflammation responses are versatile enzymes, usually involved in pro-inflammatory cytokines or other molecular mediator's production and activation of immune cells. In this chapter, an overview on major serine proteases in airway inflammation as therapeutic targets and their clinically relevant inhibitors is provided. Recent updates on serine protease inhibitors in the context of the COVID-19 pandemic are summarized.
Collapse
Affiliation(s)
- Chahrazade El Amri
- Sorbonne Université, Faculty of Sciences and Engineering, IBPS, UMR 8256 CNRS-UPMC, ERL INSERM U1164, Biological Adaptation and Ageing, Paris, France.
| |
Collapse
|