101
|
Sari A, Ekinci O, Saraçoğlu KT, Balık R, Aslan M, Balık Y, Önal C, Aslan M, Cevher S, Parmaksız A, Vatansever Ş, Çicek MC, Ayan ÖS, Şensöz Çelik G, Toprak A, Yılmaz M, Yurt E, Bakan N, Tekin S, Adıyeke E. A Comparison of the Effects of Dexamethasone and Methylprednisolone, Used on Level-3 Intensive Care COVID-19 Patients, on Mortality: A Multi-Center Retrospective Study. J Korean Med Sci 2023; 38:e232. [PMID: 37489719 PMCID: PMC10366414 DOI: 10.3346/jkms.2023.38.e232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/22/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is often a mild disease, usually manifesting with respiratory complaints, and is sometimes mortal due to multiple organ failure. Hyperinflammation is a known COVID-19 component and is associated with organ dysfunction, disease severity and mortality. Controlling hyperinflammatory response is crucial in determining treatment direction. An important agent in providing this control is corticosteroids. This study aimed to determine whether dexamethasone and methylprednisolone, doses, administration time and duration in COVID-19 treatment are associated with improved treatment outcomes. METHODS This retrospective multicenter study was conducted with participation of 6 healthcare centers which collected data by retrospectively examining files of 1,340 patients admitted to intensive care unit due to COVID-19 between March 2020 and September 2021, diagnosed with polymerase chain reaction (+) and/or clinically and radiologically. RESULTS Mortality in the pulse methylprednisolone group was statistically significantly higher than that in the other 3 groups. Mortality was higher in older patients with comorbidities such as hypertension, diabetes mellitus, chronic kidney failure, coronary artery disease, and dementia. Pulse and mini-pulse steroid doses were less effective than standard methylprednisolone and dexamethasone doses, pulse steroid doses being associated with high mortality. Standard-dose methylprednisolone and dexamethasone led to similar effects, but standard dose methylprednisolone was more effective in severe patients who required mechanical ventilation (MV). Infection development was related to steroid treatment duration, not cumulative steroid dose. CONCLUSION Corticosteroids are shown to be beneficial in critical COVID-19, but the role of early corticosteroids in mild COVID-19 patients remains unclear. The anti-inflammatory effects of corticosteroids may have a positive effect by reducing mortality in severe COVID-19 patients. Although dexamethasone was first used for this purpose, methylprednisolone was found to be as effective at standard doses. Methylprednisolone administered at standard doses was associated with greater PaO2/FiO2 ratios than dexamethasone, especially in the severe group requiring MV. High dose pulse steroid doses are closely associated with mortality and standard methylprednisolone dose is recommended.
Collapse
Affiliation(s)
- Ahmet Sari
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Haydarpasa Numune Training and Research Hospital, Istanbul, Turkey.
| | - Osman Ekinci
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Haydarpasa Numune Training and Research Hospital, Istanbul, Turkey
| | - Kemal Tolga Saraçoğlu
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Kartal Dr. Lutfi Kırdar Training and Research Hospital, Istanbul, Turkey
| | - Recep Balık
- Department of İnfectious Diseases and Clinical Microbiology, Health Sciences University Haydarpasa Numune Training and Research Hospital, Istanbul, Turkey
| | - Mesut Aslan
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Haydarpasa Numune Training and Research Hospital, Istanbul, Turkey
| | - Yelda Balık
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Haydarpasa Numune Training and Research Hospital, Istanbul, Turkey
| | - Ceren Önal
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Kartal Dr. Lutfi Kırdar Training and Research Hospital, Istanbul, Turkey
| | - Murat Aslan
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Bakırköy Dr. Sadi Konuk Training and Research Hospital, İstanbul, Turkey
| | - Semra Cevher
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Bakırköy Dr. Sadi Konuk Training and Research Hospital, İstanbul, Turkey
| | - Aylin Parmaksız
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Bakırköy Dr. Sadi Konuk Training and Research Hospital, İstanbul, Turkey
| | - Şeule Vatansever
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Prof. Dr. Cemil Taşcıoğlu City Hospital, İstanbul, Turkey
| | - Münire Canan Çicek
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Prof. Dr. Cemil Taşcıoğlu City Hospital, İstanbul, Turkey
| | - Özge Sayın Ayan
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Prof. Dr. Cemil Taşcıoğlu City Hospital, İstanbul, Turkey
| | - Gaye Şensöz Çelik
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Prof. Dr. Cemil Taşcıoğlu City Hospital, İstanbul, Turkey
| | - Açelya Toprak
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Prof. Dr. Cemil Taşcıoğlu City Hospital, İstanbul, Turkey
| | - Mehmet Yılmaz
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Derince Training and Research Hospital, Kocaeli, Turkey
| | - Emine Yurt
- Department of Anesthesiology and Reanimation/Intensive Care, Health Sciences University Derince Training and Research Hospital, Kocaeli, Turkey
| | - Nurten Bakan
- Department of Anesthesiology and Reanimation/Intensive Care, Sancaktepe Şehit Prof Dr Ilhan Varank Training and Research Hospital, İstanbul, Turkey
| | - Selda Tekin
- Department of Anesthesiology and Reanimation/Intensive Care, Sancaktepe Şehit Prof Dr Ilhan Varank Training and Research Hospital, İstanbul, Turkey
| | - Esra Adıyeke
- Department of Anesthesiology and Reanimation/Intensive Care, Sancaktepe Şehit Prof Dr Ilhan Varank Training and Research Hospital, İstanbul, Turkey
| |
Collapse
|
102
|
Abduljaleel Z, Melebari S, Athar M, Dehlawi S, Udhaya Kumar S, Aziz SA, Dannoun AI, Malik SM, Thasleem J, George Priya Doss C. SARS-CoV-2 vaccine breakthrough infections (VBI) by Omicron variant (B.1.1.529) and consequences in structural and functional impact. Cell Signal 2023:110798. [PMID: 37423342 DOI: 10.1016/j.cellsig.2023.110798] [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: 05/04/2023] [Revised: 06/18/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023]
Abstract
This study investigated the efficacy of existing vaccines against hospitalization and infection due to the Omicron variant of COVID-19, particularly for those who received two doses of Moderna or Pfizer vaccines and one dose of Johnson & Johnson vaccine or who were vaccinated more than five months before. A total of 36 variants in Omicron's spike protein, targeted by all three vaccinations, have made antibodies less effective at neutralizing the virus. The genotyping of the SARS-CoV-2 viral sequence revealed clinically significant variants such as E484K in three genetic mutations (T95I, D614G, and del142-144). A woman showed two of these mutations, indicating a potential risk of infection after successful immunization, as recently reported by Hacisuleyman (2021). We examine the effects of mutations on domains (NID, RBM, and SD2) found at the interfaces of the spike domains Omicron B.1.1529, Delta/B.1.1529, Alpha/B.1.1.7, VUM B.1.526, B.1.575.2, and B.1.1214 (formerly VOI Iota). We tested the affinity of Omicron for ACE2 and found that the wild- and mutant-spike proteins were using atomistic molecular dynamics simulations. According to the binding free energies calculated during mutagenesis, the ACE2 bound Omicron spikes more strongly than the wild strain SARS-CoV-2. T95I, D614G, and E484K are three substitutions that significantly contribute to RBD, corresponding to ACE2 binding energies and a doubling of the electrostatic potential of Omicron spike proteins. The Omicron appears to bind to ACE2 with greater affinity, increasing its infectivity and transmissibility. The spike virus was designed to strengthen antibody immune evasion through binding while boosting receptor binding by enhancing IgG and IgM antibodies that stimulate human β-cell, as opposed to the wild strain, which has more vital stimulation of both antibodies.
Collapse
Affiliation(s)
- Zainularifeen Abduljaleel
- Science and Technology Unit, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia; Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia.
| | - Sami Melebari
- Department of Molecular Biology, The Regional Laboratory, Ministry of Health (MOH), Makkah, Saudi Arabia
| | - Mohammed Athar
- Science and Technology Unit, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia; Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
| | - Saied Dehlawi
- Department of Molecular Biology, The Regional Laboratory, Ministry of Health (MOH), Makkah, Saudi Arabia
| | - S Udhaya Kumar
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Syed A Aziz
- Department of Pathology and Lab Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Anas Ibrahim Dannoun
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia
| | - Shaheer M Malik
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Jasheela Thasleem
- Jamal Mohamed College, Bharathidasan University, 7, Race Course Road, Kaja Nagar, Tiruchirappalli, Tamil Nadu 620020, India
| | - C George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| |
Collapse
|
103
|
Granholm A, Schjørring OL, Jensen AKG, Kaas-Hansen BS, Munch MW, Klitgaard TL, Crescioli E, Kjaer MBN, Strøm T, Lange T, Perner A, Rasmussen BS, Møller MH. Association between days alive without life support/out of hospital and health-related quality of life. Acta Anaesthesiol Scand 2023; 67:762-771. [PMID: 36915265 DOI: 10.1111/aas.14231] [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: 10/18/2022] [Revised: 02/28/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Trials in critically ill patients increasingly focus on days alive without life support (DAWOLS) or days alive out of hospital (DAOOH) and health-related quality of life (HRQoL). DAWOLS and DAOOH convey more information than mortality and are simpler and faster to collect than HRQoL. However, whether these outcomes are associated with HRQoL is uncertain. We thus aimed to assess the associations between DAWOLS and DAOOH and long-term HRQoL. METHODS Secondary analysis of the COVID STEROID 2 trial including adults with COVID-19 and severe hypoxaemia and the Handling Oxygenation Targets in the Intensive Care Unit (HOT-ICU) trial including adult intensive care unit patients with acute hypoxaemic respiratory failure. Associations between DAWOLS and DAOOH at day 28 and 90 and long-term HRQoL (after 6 or 12 months) using the EuroQol 5-dimension 5-level survey (EQ VAS and EQ-5D-5L index values) were assessed using flexible models and evaluated using measures of fit and prediction adequacy in both datasets (comprising internal performance and external validation), non-parametric correlation coefficients and graphical presentations. RESULTS We found no strong associations between DAWOLS or DAOOH and HRQoL in survivors at HRQoL-follow-up (615 and 1476 patients, respectively). There was substantial variability in outcomes, and predictions from the best fitted models were poor both internally and externally in the other trial dataset, which also showed inadequate calibration. Moderate associations were found when including non-survivors, although predictions remained uncertain and calibration inadequate. CONCLUSION DAWOLS and DAOOH were poorly associated with HRQoL in adult survivors of severe or critical illness included in the COVID STEROID 2 and HOT-ICU trials.
Collapse
Affiliation(s)
- Anders Granholm
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Collaboration for Research in Intensive Care (CRIC), Copenhagen, Denmark
| | - Olav Lilleholt Schjørring
- Collaboration for Research in Intensive Care (CRIC), Copenhagen, Denmark
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Aksel Karl Georg Jensen
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Benjamin Skov Kaas-Hansen
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Marie Warrer Munch
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Collaboration for Research in Intensive Care (CRIC), Copenhagen, Denmark
| | - Thomas Lass Klitgaard
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
| | - Elena Crescioli
- Collaboration for Research in Intensive Care (CRIC), Copenhagen, Denmark
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Maj-Brit Nørregaard Kjaer
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Collaboration for Research in Intensive Care (CRIC), Copenhagen, Denmark
| | - Thomas Strøm
- Department of Anaesthesia and Critical Care Medicine, Odense University Hospital, Odense, Denmark
- Department of Anaesthesia and Critical Care Medicine, Hospital Sønderjylland, University Hospital of Southern Denmark, Odense, Denmark
| | - Theis Lange
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Anders Perner
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Collaboration for Research in Intensive Care (CRIC), Copenhagen, Denmark
| | - Bodil Steen Rasmussen
- Collaboration for Research in Intensive Care (CRIC), Copenhagen, Denmark
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Morten Hylander Møller
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
- Collaboration for Research in Intensive Care (CRIC), Copenhagen, Denmark
| |
Collapse
|
104
|
Salton F, Confalonieri P, Meduri GU, Mondini L, Trotta L, Barbieri M, Bozzi C, Torregiani C, Lerda S, Bellan M, Confalonieri M, Ruaro B, Tavano S, Pozzan R. Theory and Practice of Glucocorticoids in COVID-19: Getting to the Heart of the Matter-A Critical Review and Viewpoints. Pharmaceuticals (Basel) 2023; 16:924. [PMID: 37513836 PMCID: PMC10385094 DOI: 10.3390/ph16070924] [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: 05/03/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Prolonged, low-dose glucocorticoids (GCs) have shown the highest efficacy among pharmacological and non-pharmacological treatments for COVID-19. Despite the World Health Organization's recommendation against their use at the beginning of the pandemic, GCs at a dose equivalent to dexamethasone 6 mg/day for 10 days are now indicated in all COVID-19 cases who require respiratory support. However, the efficacy of the intervention depends on the timing of initiation, the dose, and other individual factors. Indeed, patients treated with similar GC protocols often experience different outcomes, which do not always correlate with the presence of comorbidities or with the severity of respiratory involvement at baseline. This prompted us to critically review the literature on the rationale, pharmacological principles, and clinical evidence that should guide GC treatment. Based on these data, the best treatment protocol probably involves an initial bolus dose to saturate the glucocorticoid receptors, followed by a continuous infusion to maintain constant plasma levels, and eventually a slow tapering to interruption. Methylprednisolone has shown the highest efficacy among different GC molecules, most likely thanks to its higher ability to penetrate the lung. Decreased tissue sensitivity to glucocorticoids is thought to be the main mechanism accounting for the lower response to the treatment in some individuals. We do not have a readily available test to identify GC resistance; therefore, to address inter-individual variability, future research should aim at investigating clinical, physiological, and laboratory markers to guide a personalized GC treatment approach.
Collapse
Affiliation(s)
- Francesco Salton
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Paola Confalonieri
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Gianfranco Umberto Meduri
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Lucrezia Mondini
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Liliana Trotta
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Mariangela Barbieri
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Chiara Bozzi
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Chiara Torregiani
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Selene Lerda
- Business School, University of Milano, 20149 Milano, Italy
| | - Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- Center for Autoimmune and Allergic Disease (CAAD), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- A.O.U. Maggiore della Carità, 28100 Novara, Italy
| | - Marco Confalonieri
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Barbara Ruaro
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Stefano Tavano
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Riccardo Pozzan
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| |
Collapse
|
105
|
Granholm A, Kaas-Hansen BS, Lange T, Munch MW, Harhay MO, Zampieri FG, Perner A, Møller MH, Jensen AKG. Use of days alive without life support and similar count outcomes in randomised clinical trials - an overview and comparison of methodological choices and analysis methods. BMC Med Res Methodol 2023; 23:139. [PMID: 37316785 DOI: 10.1186/s12874-023-01963-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/03/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Days alive without life support (DAWOLS) and similar outcomes that seek to summarise mortality and non-mortality experiences are increasingly used in critical care research. The use of these outcomes is challenged by different definitions and non-normal outcome distributions that complicate statistical analysis decisions. METHODS We scrutinized the central methodological considerations when using DAWOLS and similar outcomes and provide a description and overview of the pros and cons of various statistical methods for analysis supplemented with a comparison of these methods using data from the COVID STEROID 2 randomised clinical trial. We focused on readily available regression models of increasing complexity (linear, hurdle-negative binomial, zero-one-inflated beta, and cumulative logistic regression models) that allow comparison of multiple treatment arms, adjustment for covariates and interaction terms to assess treatment effect heterogeneity. RESULTS In general, the simpler models adequately estimated group means despite not fitting the data well enough to mimic the input data. The more complex models better fitted and thus better replicated the input data, although this came with increased complexity and uncertainty of estimates. While the more complex models can model separate components of the outcome distributions (i.e., the probability of having zero DAWOLS), this complexity means that the specification of interpretable priors in a Bayesian setting is difficult. Finally, we present multiple examples of how these outcomes may be visualised to aid assessment and interpretation. CONCLUSIONS This summary of central methodological considerations when using, defining, and analysing DAWOLS and similar outcomes may help researchers choose the definition and analysis method that best fits their planned studies. TRIAL REGISTRATION COVID STEROID 2 trial, ClinicalTrials.gov: NCT04509973, ctri.nic.in: CTRI/2020/10/028731.
Collapse
Affiliation(s)
- Anders Granholm
- Department of Intensive Care 4131, Copenhagen University Hospital - Rigshospitalet, DK-2100, Copenhagen, Denmark.
| | - Benjamin Skov Kaas-Hansen
- Department of Intensive Care 4131, Copenhagen University Hospital - Rigshospitalet, DK-2100, Copenhagen, Denmark
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Theis Lange
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Marie Warrer Munch
- Department of Intensive Care 4131, Copenhagen University Hospital - Rigshospitalet, DK-2100, Copenhagen, Denmark
| | - Michael O Harhay
- Clinical Trials Methods and Outcomes Lab, Palliative and Advanced Illness Research Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
| | - Fernando G Zampieri
- HCor Research Institute, São Paulo, Brazil
- Department of Critical Care Medicine, Faculty of Medicine and Dentistry, University of Alberta, Alberta, Canada
| | - Anders Perner
- Department of Intensive Care 4131, Copenhagen University Hospital - Rigshospitalet, DK-2100, Copenhagen, Denmark
| | - Morten Hylander Møller
- Department of Intensive Care 4131, Copenhagen University Hospital - Rigshospitalet, DK-2100, Copenhagen, Denmark
| | - Aksel Karl Georg Jensen
- Section of Biostatistics, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
106
|
McManus D, Davis MW, Ortiz A, Britto-Leon C, Dela Cruz CS, Topal JE. Immunomodulatory Agents for Coronavirus Disease-2019 Pneumonia. Clin Chest Med 2023; 44:299-319. [PMID: 37085221 PMCID: PMC9678826 DOI: 10.1016/j.ccm.2022.11.009] [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] [Indexed: 11/23/2022]
Abstract
Morbidity and mortality from COVID-19 is due to severe inflammation and end-organ damage caused by a hyperinflammatory response. Multiple immunomodulatory agents to attenuate this response have been studied. Corticosteroids, specifically dexamethasone, have been shown to reduce mortality in hospitalized patients who require supplemental oxygen. Interleukin-6 antagonist, tocilizimab, and Janus kinase inhibitors have also been shown to reduce mortality. However, patients who have severe pulmonary end-organ damage requiring mechanical ventilation or extracorporeal membrane oxygenation appear not to benefit from immunomodulatory therapies. This highlights the importance of appropriate timing to initiate immunomodulatory therapies in the management of severe COVID-19 disease.
Collapse
Affiliation(s)
- Dayna McManus
- Department of Pharmacy Services, Yale New Haven Hospital, 20 York Street, New Haven, CT 06510, USA.
| | - Matthew W Davis
- Department of Pharmacy Services, Yale New Haven Hospital, 20 York Street, New Haven, CT 06510, USA
| | - Alex Ortiz
- Pulmonary, Critical Care & Sleep Medicine, 300 Cedar Street, P.O. Box 208057, New Haven, CT 06520-8057, USA
| | - Clemente Britto-Leon
- Pulmonary, Critical Care & Sleep Medicine, 300 Cedar Street, P.O. Box 208057, New Haven, CT 06520-8057, USA
| | - Charles S Dela Cruz
- Pulmonary, Critical Care & Sleep Medicine, 300 Cedar Street, P.O. Box 208057, New Haven, CT 06520-8057, USA
| | - Jeffrey E Topal
- Department of Pharmacy Services, Yale New Haven Hospital, 20 York Street, New Haven, CT 06510, USA.
| |
Collapse
|
107
|
Myles PS, Yeung J, Beattie WS, Ryan EG, Heritier S, McArthur CJ. Platform trials for anaesthesia and perioperative medicine: a narrative review. Br J Anaesth 2023; 130:677-686. [PMID: 36456249 DOI: 10.1016/j.bja.2022.10.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/27/2022] [Accepted: 10/28/2022] [Indexed: 11/29/2022] Open
Abstract
Large randomised trials provide the most reliable evidence of effectiveness of new treatments in clinical practice. However, the time and resources required to complete such trials can be daunting. An overarching clinical trial platform focused on a single condition or type of surgery, aiming to compare several treatments, with an option to stop any or add in new treatment options, can provide greater efficiency. This has the potential to accelerate knowledge acquisition and identify effective, ineffective, or harmful treatments faster. The master protocol of the platform defines the study population(s) and standardised procedures. Ineffective or harmful treatments can be discarded or study drug dose modified during the life cycle of the trial. Other adaptive elements that can be modified include eligibility criteria, required sample size for any comparison(s), randomisation assignment ratio, and the addition of other promising treatment options. There are excellent opportunities for anaesthetists to establish platform trials in perioperative medicine. Platform trials are highly efficient, with the potential to provide quicker answers to important clinical questions that lead to improved patient care.
Collapse
Affiliation(s)
- Paul S Myles
- Department of Anaesthesiology and Perioperative Medicine, Alfred Health, Melbourne, VIC, Australia; Department of Anaesthesiology and Perioperative Medicine, Monash University, Melbourne, VIC, Australia.
| | - Joyce Yeung
- Warwick Medical School, University of Warwick, Coventry, UK; Department of Anaesthesia and Critical Care, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - W Scott Beattie
- Department of Anaesthesia and Pain Management, University of Toronto, Toronto, ON, Canada; University Health Network, Toronto, ON, Canada
| | - Elizabeth G Ryan
- Biostatistics Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Stephane Heritier
- Biostatistics Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Colin J McArthur
- Department of Critical Care Medicine, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|
108
|
Dal-Ré R, Voo TC, Holm S. Adaptive, platform trials assessing therapies for hospitalized COVID-19 patients: Informed consent forms omitted a few important elements of information. J Glob Health 2023; 13:06019. [PMID: 37166268 PMCID: PMC10173897 DOI: 10.7189/jogh.13.06019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
Background The information provided to participants of adaptive platform trials assessing therapies for COVID-19 inpatients is unknown. We aim to evaluate it by reviewing participant information sheets/informed consent forms (PIS/ICFs). Methods We searched the Cochrane COVID-19 Study Register and ClinicalTrials.gov (28 March 2022) to identify non-industry-sponsored adaptive platform phase 2+ trials with publicly available protocols and PIS/ICFs, selecting versions closest to the initial one. We assessed the elements of information included in the Good Clinical Practice guidelines and the Declaration of Helsinki as present, absent, or deficient (incompletely described). Results We included PIS/ICFs of 11 trials (ACCORD-2, ACTIV-1IM, Bari-SolidAct, CATALYST, Discovery, HEAL-COVID, ITAC, RECOVERY, REMAP-COVID, Solidarity and TACTIC-R), which were 4-32 pages long (median (md) = 11). Between two and 11 (md = 6) of the 25 different elements of information assessed were omitted or deficiently described in the PIS/ICFs of the 11 trials. Information about providing trial results, investigators' conflicts of interest, post-study provisions, payment to and anticipated expenses for participants, number of participants, and on whether participants will receive new information that could impact their decision on staying in the trial, were omitted or deficiently described in at least five PIS/ICFs. Conclusions Investigators failed to include a few important elements of information in the trial's PIS/ICF deemed relevant by international standards. In protocols of future trials, investigators should explain why elements of information specified in the Good Clinical Practice guidelines and/or by the Declaration of Helsinki were omitted from the PIS/ICFs.
Collapse
Affiliation(s)
- Rafael Dal-Ré
- Epidemiology Unit, Health Research Institute-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid, Madrid, Spain
| | - Teck Chuan Voo
- Centre for Biomedical Ethics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Søren Holm
- Centre for Social Ethics and Policy, Department of Law, School of Social Sciences, University of Manchester, University of Manchester, UK
- Center for Medical Ethics, HELSAM, Faculty of Medicine, University of Oslo, Norway
| |
Collapse
|
109
|
Saul S, Karim M, Ghita L, Huang PT, Chiu W, Durán V, Lo CW, Kumar S, Bhalla N, Leyssen P, Alem F, Boghdeh NA, Tran DH, Cohen CA, Brown JA, Huie KE, Tindle C, Sibai M, Ye C, Khalil AM, Martinez-Sobrido L, Dye JM, Pinsky BA, Ghosh P, Das S, Solow-Cordero DE, Jin J, Wikswo JP, Jochmans D, Neyts J, Jonghe SD, Narayanan A, Einav S. Anticancer pan-ErbB inhibitors reduce inflammation and tissue injury and exert broad-spectrum antiviral effects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2021.05.15.444128. [PMID: 34159337 PMCID: PMC8219101 DOI: 10.1101/2021.05.15.444128] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Targeting host factors exploited by multiple viruses could offer broad-spectrum solutions for pandemic preparedness. Seventeen candidates targeting diverse functions emerged in a screen of 4,413 compounds for SARS-CoV-2 inhibitors. We demonstrated that lapatinib and other approved inhibitors of the ErbB family receptor tyrosine kinases suppress replication of SARS-CoV-2, Venezuelan equine encephalitis virus (VEEV), and other emerging viruses with a high barrier to resistance. Lapatinib suppressed SARS-CoV-2 entry and later stages of the viral life cycle and showed synergistic effect with the direct-acting antiviral nirmatrelvir. We discovered that ErbB1, 2 and 4 bind SARS-CoV-2 S1 protein and regulate viral and ACE2 internalization, and they are required for VEEV infection. In human lung organoids, lapatinib protected from SARS-CoV-2-induced activation of ErbB-regulated pathways implicated in non-infectious lung injury, pro-inflammatory cytokine production, and epithelial barrier injury. Lapatinib suppressed VEEV replication, cytokine production and disruption of the blood-brain barrier integrity in microfluidic-based human neurovascular units, and reduced mortality in a lethal infection murine model. We validated lapatinib-mediated inhibition of ErbB activity as an important mechanism of antiviral action. These findings reveal regulation of viral replication, inflammation, and tissue injury via ErbBs and establish a proof-of-principle for a repurposed, ErbB-targeted approach to combat emerging viruses.
Collapse
|
110
|
Kuperminc E, Heming N, Carlos M, Annane D. Corticosteroids in ARDS. J Clin Med 2023; 12:jcm12093340. [PMID: 37176780 PMCID: PMC10179626 DOI: 10.3390/jcm12093340] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is frequently associated with sepsis. ARDS and sepsis exhibit a common pathobiology, namely excessive inflammation. Corticosteroids are powerful anti-inflammatory agents that are routinely used in septic shock and in oxygen-dependent SARS-CoV-2 related acute respiratory failure. Recently, corticosteroids were found to reduce mortality in severe community-acquired pneumonia. Corticosteroids may therefore also have a role to play in the treatment of ARDS. This narrative review was undertaken following a PubMed search for English language reports published before January 2023 using the terms acute respiratory distress syndrome, sepsis and steroids. Additional reports were identified by examining the reference lists of selected articles and based on personnel knowledge of the authors of the field. High-quality research is needed to fully understand the role of corticosteroids in the treatment of ARDS and to determine the optimal timing, dosing and duration of treatment.
Collapse
Affiliation(s)
- Emmanuelle Kuperminc
- Department of Intensive Care, Hôpital Raymond Poincaré, APHP University Versailles Saint Quentin-University Paris Saclay, 92380 Garches, France
| | - Nicholas Heming
- Department of Intensive Care, Hôpital Raymond Poincaré, APHP University Versailles Saint Quentin-University Paris Saclay, 92380 Garches, France
- Laboratory of Infection & Inflammation-U1173, School of Medicine Simone Veil, University Versailles Saint Quentin-University Paris Saclay, INSERM, 92380 Garches, France
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), 92380 Garches, France
| | - Miguel Carlos
- Department of Intensive Care, Hôpital Raymond Poincaré, APHP University Versailles Saint Quentin-University Paris Saclay, 92380 Garches, France
| | - Djillali Annane
- Department of Intensive Care, Hôpital Raymond Poincaré, APHP University Versailles Saint Quentin-University Paris Saclay, 92380 Garches, France
- Laboratory of Infection & Inflammation-U1173, School of Medicine Simone Veil, University Versailles Saint Quentin-University Paris Saclay, INSERM, 92380 Garches, France
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), 92380 Garches, France
| |
Collapse
|
111
|
Tsotsolis S, Kotoulas SC, Lavrentieva A. Invasive Pulmonary Aspergillosis in Coronavirus Disease 2019 Patients Lights and Shadows in the Current Landscape. Adv Respir Med 2023; 91:185-202. [PMID: 37218799 DOI: 10.3390/arm91030016] [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: 02/07/2023] [Revised: 04/17/2023] [Accepted: 04/30/2023] [Indexed: 05/24/2023]
Abstract
Invasive pulmonary aspergillosis (IPA) presents a known risk to critically ill patients with SARS-CoV-2; quantifying the global burden of IPA in SARS-CoV-2 is extremely challenging. The true incidence of COVID-19-associated pulmonary aspergillosis (CAPA) and the impact on mortality is difficult to define because of indiscriminate clinical signs, low culture sensitivity and specificity and variability in clinical practice between centers. While positive cultures of upper airway samples are considered indicative for the diagnosis of probable CAPA, conventional microscopic examination and qualitative culture of respiratory tract samples have quite low sensitivity and specificity. Thus, the diagnosis should be confirmed with serum and BAL GM test or positive BAL culture to mitigate the risk of overdiagnosis and over-treatment. Bronchoscopy has a limited role in these patients and should only be considered when diagnosis confirmation would significantly change clinical management. Varying diagnostic performance, availability, and time-to-results turnaround time are important limitations of currently approved biomarkers and molecular assays for the diagnosis of IA. The use of CT scans for diagnostic purposes is controversial due to practical concerns and the complex character of lesions presented in SARS-CoV-2 patients. The key objective of management is to improve survival by avoiding misdiagnosis and by initiating early, targeted antifungal treatment. The main factors that should be considered upon selection of treatment options include the severity of the infection, concomitant renal or hepatic injury, possible drug interactions, requirement for therapeutic drug monitoring, and cost of therapy. The optimal duration of antifungal therapy for CAPA is still under debate.
Collapse
Affiliation(s)
- Stavros Tsotsolis
- Medical School, Aristotle University of Thessaloniki, Leoforos Agiou Dimitriou, 54124 Thessaloniki, Greece
| | | | - Athina Lavrentieva
- 1st ICU, General Hospital of Thessaloniki "Georgios Papanikolaou", Leoforos Papanikolaou, 57010 Thessaloniki, Greece
| |
Collapse
|
112
|
Ramos R, de la Villa S, García-Ramos S, Padilla B, García-Olivares P, Piñero P, Garrido A, Hortal J, Muñoz P, Caamaño E, Benito P, Cedeño J, Garutti I. COVID-19 associated infections in the ICU setting: A retrospective analysis in a tertiary-care hospital. ENFERMEDADES INFECCIOSAS Y MICROBIOLOGIA CLINICA (ENGLISH ED.) 2023; 41:278-283. [PMID: 37142346 PMCID: PMC10151902 DOI: 10.1016/j.eimce.2021.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/20/2021] [Indexed: 05/06/2023]
Abstract
INTRODUCTION Our work describes the frequency of superinfections in COVID-19 ICU patients and identifies risk factors for its appearance. Second, we evaluated ICU length of stay, in-hospital mortality and analyzed a subgroup of multidrug-resistant microorganisms (MDROs) infections. METHODS Retrospective study conducted between March and June 2020. Superinfections were defined as appeared ≥48h. Bacterial and fungal infections were included, and sources were ventilator-associated lower respiratory tract infection (VA-LRTI), primary bloodstream infection (BSI), secondary BSI, and urinary tract infection (UTI). We performed a univariate analysis and a multivariate analysis of the risk factors. RESULTS Two-hundred thirteen patients were included. We documented 174 episodes in 95 (44.6%) patients: 78 VA-LRTI, 66 primary BSI, 9 secondary BSI and 21 UTI. MDROs caused 29.3% of the episodes. The median time from admission to the first episode was 18 days and was longer in MDROs than in non-MDROs (28 vs. 16 days, p<0.01). In multivariate analysis use of corticosteroids (OR 4.9, 95% CI 1.4-16.9, p 0.01), tocilizumab (OR 2.4, 95% CI 1.1-5.9, p 0.03) and broad-spectrum antibiotics within first 7 days of admission (OR 2.5, 95% CI 1.2-5.1, p<0.01) were associated with superinfections. Patients with superinfections presented respect to controls prolonged ICU stay (35 vs. 12 days, p<0.01) but not higher in-hospital mortality (45.3% vs. 39.7%, p 0.13). CONCLUSIONS Superinfections in ICU patients are frequent in late course of admission. Corticosteroids, tocilizumab, and previous broad-spectrum antibiotics are identified as risk factors for its development.
Collapse
Affiliation(s)
- Rafael Ramos
- Anesthesiology and Reanimation Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Sofía de la Villa
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| | - Sergio García-Ramos
- Anesthesiology and Reanimation Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Belén Padilla
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Pablo García-Olivares
- Intensive Care Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Patricia Piñero
- Anesthesiology and Reanimation Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Alberto Garrido
- Intensive Care Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Javier Hortal
- Anesthesiology and Reanimation Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Patricia Muñoz
- Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain; CIBER Enfermedades Respiratorias-CIBERES (CB06/06/0058), Medicine Department, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Estrela Caamaño
- Anesthesiology and Reanimation Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Pilar Benito
- Anesthesiology and Reanimation Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Jamil Cedeño
- Intensive Care Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Ignacio Garutti
- Anesthesiology and Reanimation Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| |
Collapse
|
113
|
Kim JH, Na YS, Lee SI, Moon YY, Hwang BS, Baek AR, Kim WY, Lee BY, Seong GM, Baek MS. Corticosteroid outcome may be dependent of duration of use in severe COVID-19. Korean J Intern Med 2023; 38:382-392. [PMID: 37038264 PMCID: PMC10175872 DOI: 10.3904/kjim.2022.201] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/27/2022] [Accepted: 01/01/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND/AIMS For patients hospitalized with coronavirus disease 2019 (COVID-19) who require supplemental oxygen, the evidence of the optimal duration of corticosteroid is limited. This study aims to identify whether long-term use of corticosteroids is associated with decreased mortality. METHODS Between February 10, 2020 and October 31, 2021, we analyzed consecutive hospitalized patients with COVID-19 with severe hypoxemia. The patients were divided into short-term (≤ 14 days) and long-term (> 14 days) corticosteroid users. The primary outcome was 60-day mortality. We performed propensity score (PS) analysis to mitigate the effect of confounders and conducted Kaplan-Meier curve analysis. RESULTS There were 141 (52%) short-term users and 130 (48%) long-term corticosteroid users. The median age was 68 years and the median PaO2/FiO2 at admission was 158. Of the patients, 40.6% required high-flow nasal cannula, 48.3% required mechanical ventilation, and 11.1% required extracorporeal membrane oxygenation. The overall 60-day mortality rate was 23.2%, and that of patients with hospital-acquired pneumonia (HAP) was 22.9%. The Kaplan-Meier curve for 60- day survival in the PS-matched cohort showed that corticosteroid for > 14 days was associated with decreased mortality (p = 0.0033). There were no significant differences in bacteremia and HAP between the groups. An adjusted odds ratio for the risk of 60-day mortality in short-term users was 5.53 (95% confidence interval, 1.90-18.26; p = 0.003). CONCLUSION For patients with severe COVID-19, long-term use of corticosteroids was associated with decreased mortality, with no increase in nosocomial complications. Corticosteroid use for > 14 days can benefit patients with severe COVID-19.
Collapse
Affiliation(s)
- Jin Hyoung Kim
- Division of Respiratory and Critical Care Medicine, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan,
Korea
| | - Yong Sub Na
- Department of Pulmonology and Critical Care Medicine, Chosun University Hospital, Gwangju,
Korea
| | - Song-I Lee
- Department of Pulmonary and Critical Care Medicine, Chungnam National University Hospital, Daejeon,
Korea
| | - Youn Young Moon
- Department of Applied Statistics, Chung-Ang University, Seoul,
Korea
| | - Beom Seuk Hwang
- Department of Applied Statistics, Chung-Ang University, Seoul,
Korea
| | - Ae-Rin Baek
- Division of Allergy and Pulmonology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon,
Korea
| | - Won-Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul,
Korea
| | - Bo Young Lee
- Division of Allergy and Respiratory Diseases, Soonchunhyang University Hospital, Seoul,
Korea
| | - Gil Myeong Seong
- Department of Internal Medicine, Jeju National University College of Medicine, Jeju,
Korea
| | - Moon Seong Baek
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul,
Korea
| |
Collapse
|
114
|
Dechamps M, De Poortere J, Octave M, Ginion A, Robaux V, Pirotton L, Bodart J, Gruson D, Van Dievoet MA, Douxfils J, Haguet H, Morimont L, Derive M, Jolly L, Bertrand L, Laterre PF, Horman S, Beauloye C. Dexamethasone Modulates the Cytokine Response but Not COVID-19-Induced Coagulopathy in Critically Ill. Int J Mol Sci 2023; 24:ijms24087278. [PMID: 37108440 PMCID: PMC10138864 DOI: 10.3390/ijms24087278] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/29/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
Severe forms of coronavirus 2019 (COVID-19) disease are caused by an exaggerated systemic inflammatory response and subsequent inflammation-related coagulopathy. Anti-inflammatory treatment with low dose dexamethasone has been shown to reduce mortality in COVID-19 patients requiring oxygen therapy. However, the mechanisms of action of corticosteroids have not been extensively studied in critically ill patients in the context of COVID-19. Plasma biomarkers of inflammatory and immune responses, endothelial and platelet activation, neutrophil extracellular trap formation, and coagulopathy were compared between patients treated or not by systemic dexamethasone for severe forms of COVID-19. Dexamethasone treatment significantly reduced the inflammatory and lymphoid immune response in critical COVID-19 patients but had little effect on the myeloid immune response and no effect on endothelial activation, platelet activation, neutrophil extracellular trap formation, and coagulopathy. The benefits of low dose dexamethasone on outcome in critical COVID-19 can be partially explained by a modulation of the inflammatory response but not by reduction of coagulopathy. Future studies should explore the impact of combining dexamethasone with other immunomodulatory or anticoagulant drugs in severe COVID-19.
Collapse
Affiliation(s)
- Mélanie Dechamps
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
- Department of Cardiovascular Intensive Care, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Julien De Poortere
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Marie Octave
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Audrey Ginion
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Valentine Robaux
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Laurence Pirotton
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Julie Bodart
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Damien Gruson
- Department of Clinical Biology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | | | - Jonathan Douxfils
- Department of Pharmacy, Namur Research Institute for Life Sciences (Narilis), 5000 Namur, Belgium
- Qualiblood, s.a., 5000 Namur, Belgium
| | - Hélène Haguet
- Department of Pharmacy, Namur Research Institute for Life Sciences (Narilis), 5000 Namur, Belgium
- Qualiblood, s.a., 5000 Namur, Belgium
| | - Laure Morimont
- Department of Pharmacy, Namur Research Institute for Life Sciences (Narilis), 5000 Namur, Belgium
- Qualiblood, s.a., 5000 Namur, Belgium
| | - Marc Derive
- Inotrem s.a., 54500 Vandoeuvre-les-Nancy, France
| | - Lucie Jolly
- Inotrem s.a., 54500 Vandoeuvre-les-Nancy, France
| | - Luc Bertrand
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO) Department, WEL Research Institute, 1300 Wavre, Belgium
| | - Pierre-François Laterre
- Department of Intensive Care, Centre Hospitalier Regional Mons-Hainaut, 7000 Mons, Belgium
- Critical Care Coordinating Center (4Cs), 1200 Brussels, Belgium
| | - Sandrine Horman
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Christophe Beauloye
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
- Department of Cardiology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
115
|
O'Grady HK, Bhimani Z, Dalziel S, Dolanjski B, Sandhu G, Santos M, Smith K, Murthy S, Marshall JC, Kho ME. Co-designing and pilot testing an infographic to support patients/families through the REMAP-CAP consent process: a mixed-methods study protocol. Pilot Feasibility Stud 2023; 9:58. [PMID: 37055859 PMCID: PMC10098229 DOI: 10.1186/s40814-023-01290-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 03/30/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Informed consent is critical to the ethical conduct of clinical research and requires understanding of a trial including its purpose, process, potential risks and benefits, and alternatives to participation. This can be challenging for complex trials, such as platform trials, and in high-stress environments, such as the intensive care unit (ICU). REMAP-CAP (randomized, embedded, multifactorial, adaptive platform trial for community-acquired pneumonia) is a platform trial which studies treatments for ICU patients with community-acquired pneumonia, including COVID-19. Patient/family partners (PFP) identified challenges during the REMAP-CAP consent process. METHODS This is a patient-centred co-design study to refine and test an infographic to supplement current REMAP-CAP consent documents. Infographic prototypes were developed by patients, substitute decision-makers (SDMs), and researchers with lived experience in the ICU or with ICU research. We will apply a two-phase exploratory sequential, mixed-methods research design. In phase 1, we will conduct focus groups with ICU patients, SDMs, and research coordinators (RCs). We will use inductive content analysis to inform infographic refinement, to be pilot tested in phase 2. Phase 2 is a prospective study within a trial (SWAT) at ≤ 5 REMAP-CAP sites. We will collect self-reported data from patients/SDMs and RCs. The primary outcome is feasibility (eligible consent encounters, receipt of infographic, consent to follow-up, completion of follow-up surveys). Data will be integrated to understand if/how quantitative results build upon the qualitatively informed infographic. DISCUSSION Phase 1 results will be used to co-design an infographic, directly informed by the perspectives of patients, SDMs, and RCs involved in ICU research consent discussions. Results from phase 2 will determine the feasibility of infographic implementation in REMAP-CAP consent encounters. These feasibility data will inform a larger SWAT to evaluate our consent infographic. If successful, use of a co-designed infographic to support REMAP-CAP consent documents may improve the experience of consent for patients, SDMs, and RCs. TRIAL REGISTRATION The Northern Ireland Hub for Trials Methodology Research SWAT Repository (SWAT no. 176).
Collapse
Affiliation(s)
- Heather K O'Grady
- Faculty of Health Sciences, School of Rehabilitation Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - Zahra Bhimani
- St. Michael's Hospital Unity Health Toronto, Toronto, Ontario, Canada
| | - Sandra Dalziel
- St. Michael's Hospital Unity Health Toronto, Toronto, Ontario, Canada
| | - Barbara Dolanjski
- St. Michael's Hospital Unity Health Toronto, Toronto, Ontario, Canada
| | - Gyan Sandhu
- St. Michael's Hospital Unity Health Toronto, Toronto, Ontario, Canada
| | - Marlene Santos
- St. Michael's Hospital Unity Health Toronto, Toronto, Ontario, Canada
| | - Kathy Smith
- St. Michael's Hospital Unity Health Toronto, Toronto, Ontario, Canada
| | - Srinivas Murthy
- Faculty of Medicine, Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - John C Marshall
- St. Michael's Hospital Unity Health Toronto, Toronto, Ontario, Canada
| | - Michelle E Kho
- Faculty of Health Sciences, School of Rehabilitation Sciences, McMaster University, Hamilton, Ontario, Canada
- Physiotherapy Department, St. Joseph's Healthcare, Hamilton, Ontario, Canada
| |
Collapse
|
116
|
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
|
117
|
Lawler PR, Derde LPG, van de Veerdonk FL, McVerry BJ, Huang DT, Berry LR, Lorenzi E, van Kimmenade R, Gommans F, Vaduganathan M, Leaf DE, Baron RM, Kim EY, Frankfurter C, Epelman S, Kwan Y, Grieve R, O'Neill S, Sadique Z, Puskarich M, Marshall JC, Higgins AM, Mouncey PR, Rowan KM, Al-Beidh F, Annane D, Arabi YM, Au C, Beane A, van Bentum-Puijk W, Bonten MJM, Bradbury CA, Brunkhorst FM, Burrell A, Buzgau A, Buxton M, Cecconi M, Cheng AC, Cove M, Detry MA, Estcourt LJ, Ezekowitz J, Fitzgerald M, Gattas D, Godoy LC, Goossens H, Haniffa R, Harrison DA, Hills T, Horvat CM, Ichihara N, Lamontagne F, Linstrum KM, McAuley DF, McGlothlin A, McGuinness SP, McQuilten Z, Murthy S, Nichol AD, Owen DRJ, Parke RL, Parker JC, Pollock KM, Reyes LF, Saito H, Santos MS, Saunders CT, Seymour CW, Shankar-Hari M, Singh V, Turgeon AF, Turner AM, Zarychanski R, Green C, Lewis RJ, Angus DC, Berry S, Gordon AC, McArthur CJ, Webb SA. Effect of Angiotensin-Converting Enzyme Inhibitor and Angiotensin Receptor Blocker Initiation on Organ Support-Free Days in Patients Hospitalized With COVID-19: A Randomized Clinical Trial. JAMA 2023; 329:1183-1196. [PMID: 37039790 PMCID: PMC10326520 DOI: 10.1001/jama.2023.4480] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/07/2023] [Indexed: 04/12/2023]
Abstract
IMPORTANCE Overactivation of the renin-angiotensin system (RAS) may contribute to poor clinical outcomes in patients with COVID-19. Objective To determine whether angiotensin-converting enzyme (ACE) inhibitor or angiotensin receptor blocker (ARB) initiation improves outcomes in patients hospitalized for COVID-19. DESIGN, SETTING, AND PARTICIPANTS In an ongoing, adaptive platform randomized clinical trial, 721 critically ill and 58 non-critically ill hospitalized adults were randomized to receive an RAS inhibitor or control between March 16, 2021, and February 25, 2022, at 69 sites in 7 countries (final follow-up on June 1, 2022). INTERVENTIONS Patients were randomized to receive open-label initiation of an ACE inhibitor (n = 257), ARB (n = 248), ARB in combination with DMX-200 (a chemokine receptor-2 inhibitor; n = 10), or no RAS inhibitor (control; n = 264) for up to 10 days. MAIN OUTCOMES AND MEASURES The primary outcome was organ support-free days, a composite of hospital survival and days alive without cardiovascular or respiratory organ support through 21 days. The primary analysis was a bayesian cumulative logistic model. Odds ratios (ORs) greater than 1 represent improved outcomes. RESULTS On February 25, 2022, enrollment was discontinued due to safety concerns. Among 679 critically ill patients with available primary outcome data, the median age was 56 years and 239 participants (35.2%) were women. Median (IQR) organ support-free days among critically ill patients was 10 (-1 to 16) in the ACE inhibitor group (n = 231), 8 (-1 to 17) in the ARB group (n = 217), and 12 (0 to 17) in the control group (n = 231) (median adjusted odds ratios of 0.77 [95% bayesian credible interval, 0.58-1.06] for improvement for ACE inhibitor and 0.76 [95% credible interval, 0.56-1.05] for ARB compared with control). The posterior probabilities that ACE inhibitors and ARBs worsened organ support-free days compared with control were 94.9% and 95.4%, respectively. Hospital survival occurred in 166 of 231 critically ill participants (71.9%) in the ACE inhibitor group, 152 of 217 (70.0%) in the ARB group, and 182 of 231 (78.8%) in the control group (posterior probabilities that ACE inhibitor and ARB worsened hospital survival compared with control were 95.3% and 98.1%, respectively). CONCLUSIONS AND RELEVANCE In this trial, among critically ill adults with COVID-19, initiation of an ACE inhibitor or ARB did not improve, and likely worsened, clinical outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02735707.
Collapse
Affiliation(s)
- Patrick R Lawler
- Peter Munk Cardiac Centre at University Health Network, Toronto, Canada
- McGill University Health Centre, Montreal, QC, Canada
| | | | | | | | | | | | | | | | - Frank Gommans
- Radboud University Medical Centre, Nijmegen, Netherlands
| | | | - David E Leaf
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rebecca M Baron
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Edy Y Kim
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Slava Epelman
- Peter Munk Cardiac Centre at University Health Network, Toronto, Canada
| | - Yvonne Kwan
- Peter Munk Cardiac Centre at University Health Network, Toronto, Canada
| | - Richard Grieve
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Stephen O'Neill
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Zia Sadique
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | | | | | - Paul R Mouncey
- Intensive Care National Audit & Research Centre (ICNARC), London, United Kingdom
| | - Kathryn M Rowan
- Intensive Care National Audit & Research Centre (ICNARC), London, United Kingdom
| | | | - Djillali Annane
- Hospital Raymond Poincaré (Assistance Publique Hôpitaux de Paris), Garches, France
- Université Versailles SQY - Université Paris Saclay, Montigny-le-Bretonneux, France
| | - Yaseen M Arabi
- King Saud bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Carly Au
- Intensive Care National Audit & Research Centre (ICNARC), London, United Kingdom
| | - Abi Beane
- University of Oxford, Oxford, England
| | | | | | | | | | | | | | - Meredith Buxton
- Global Coalition for Adaptive Research, Larkspur, California
| | | | | | - Matthew Cove
- Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | | | | | | | | | - David Gattas
- The George Institute for Global Health, Sydney, Australia
| | - Lucas C Godoy
- Peter Munk Cardiac Centre at University Health Network, Toronto, Canada
| | | | - Rashan Haniffa
- University of Oxford, Bangkok, Thailand
- National Intensive Care Surveillance (NICST), Colombo, Sri Lanka
| | - David A Harrison
- Intensive Care National Audit & Research Centre (ICNARC), London, United Kingdom
| | - Thomas Hills
- Medical Research Institute of New Zealand (MRINZ), Wellington, New Zealand
| | | | | | | | | | - Daniel F McAuley
- Queen's University Belfast, Belfast, Northern Ireland
- Royal Victoria Hospital, Belfast, Northern Ireland
| | | | - Shay P McGuinness
- Monash University, Melbourne, Australia
- Auckland City Hospital, Auckland, New Zealand
| | | | | | - Alistair D Nichol
- Monash University, Melbourne, Australia
- University College Dublin, Dublin, Ireland
| | - David R J Owen
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- UK Dementia Research Institute of Imperial College London, London, United Kingdom
| | - Rachael L Parke
- Auckland City Hospital, Auckland, New Zealand
- University of Auckland, Auckland, New Zealand
| | | | | | - Luis Felipe Reyes
- Universidad de La Sabana, Chia, Colombia
- Clinica Universidad de La Sabana, Chia, Colombia
| | - Hiroki Saito
- St Marianna University School of Medicine, Yokohama City Seibu Hospital, Yokohama, Japan
| | | | | | | | | | | | - Alexis F Turgeon
- Université Laval, Québec City, Canada
- CHU de Québec-Université Laval Research Center, Québec City, Canada
| | - Anne M Turner
- Medical Research Institute of New Zealand (MRINZ), Wellington, New Zealand
| | | | | | - Roger J Lewis
- Berry Consultants, Austin, Texas
- Harbor-UCLA Medical Center, Torrance, California
- Statistical Editor, JAMA
| | - Derek C Angus
- University of Pittsburgh, Pittsburgh, Pennsylvania
- Senior Editor, JAMA
| | | | - Anthony C Gordon
- Imperial College London, London, United Kingdom
- Imperial College Healthcare NHS Trust, St Mary's Hospital, London, United Kingdom
| | | | - Steve A Webb
- Monash University, Melbourne, Australia
- St John of God Hospital, Subiaco, Australia
| |
Collapse
|
118
|
Mondini L, Salton F, Trotta L, Bozzi C, Pozzan R, Barbieri M, Tavano S, Lerda S, Hughes M, Confalonieri M, Confalonieri P, Ruaro B. Host-Based Treatments for Severe COVID-19. Curr Issues Mol Biol 2023; 45:3102-3121. [PMID: 37185727 PMCID: PMC10136924 DOI: 10.3390/cimb45040203] [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: 03/03/2023] [Revised: 03/29/2023] [Accepted: 04/02/2023] [Indexed: 05/17/2023] Open
Abstract
COVID-19 has been a global health problem since 2020. There are different spectrums of manifestation of this disease, ranging from asymptomatic to extremely severe forms requiring admission to intensive care units and life-support therapies, mainly due to severe pneumonia. The progressive understanding of this disease has allowed researchers and clinicians to implement different therapeutic alternatives, depending on both the severity of clinical involvement and the causative molecular mechanism that has been progressively explored. In this review, we analysed the main therapeutic options available to date based on modulating the host inflammatory response to SARS-CoV-2 infection in patients with severe and critical illness. Although current guidelines are moving toward a personalised treatment approach titrated on the timing of presentation, disease severity, and laboratory parameters, future research is needed to identify additional biomarkers that can anticipate the disease course and guide targeted interventions on an individual basis.
Collapse
Affiliation(s)
- Lucrezia Mondini
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Francesco Salton
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Liliana Trotta
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Chiara Bozzi
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Riccardo Pozzan
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Mariangela Barbieri
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Stefano Tavano
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Selene Lerda
- Graduate School, University of Milan, 20149 Milano, Italy
| | - Michael Hughes
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester & Salford Royal NHS Foundation Trust, Manchester M6 8HD, UK
| | - Marco Confalonieri
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Paola Confalonieri
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| | - Barbara Ruaro
- Pulmonology Unit, Department of Medical Surgical and Health Sciences, University Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy
| |
Collapse
|
119
|
Files DC, Aggarwal N, Albertson T, Auld S, Beitler JR, Berger P, Burnham EL, Calfee CS, Cobb N, Crippa A, Discacciati A, Eklund M, Esserman L, Friedman E, Gandotra S, Khan K, Koff J, Kumar S, Liu KD, Martin TR, Matthay MA, Meyer NJ, Obermiller T, Robinson P, Russell D, Thomas K, Wong SF, Wunderink RG, Wurfel MM, Yen A, Youssef FA, Darmanian A, Dzierba AL, Garcia I, Gosek K, Madahar P, Mittel AM, Muir J, Rosen A, Schicchi J, Serra AL, Wahab R, Gibbs KW, Landreth L, LaRose M, Parks L, Wynn A, Ittner CA, Mangalmurti NS, Reilly JP, Harris D, Methukupally A, Patel S, Boerger L, Kazianis J, Higgins C, McKeehan J, Daniel B, Fields S, Hurst-Hopf J, Jauregui A, Brown Swigart L, Blevins D, Nguyen C, Suarez A, Tanios MA, Sarafian F, Shah U, Adelman M, Creel-Bulos C, Detelich J, Harris G, Nugent K, Spainhour C, Yang P, Haczku A, Hardy E, Harper R, Morrissey B, Sandrock C, Budinger GRS, Donnelly HK, Singer BD, Moskowitz A, Coleman M, Levitt J, Lu R, Henderson P, Asare A, Dunn I, Botello Barragan A. Report of the first seven agents in the I-SPY COVID trial: a phase 2, open label, adaptive platform randomised controlled trial. EClinicalMedicine 2023; 58:101889. [PMID: 36883141 PMCID: PMC9981330 DOI: 10.1016/j.eclinm.2023.101889] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Background An urgent need exists to rapidly screen potential therapeutics for severe COVID-19 or other emerging pathogens associated with high morbidity and mortality. Methods Using an adaptive platform design created to rapidly evaluate investigational agents, hospitalised patients with severe COVID-19 requiring ≥6 L/min oxygen were randomised to either a backbone regimen of dexamethasone and remdesivir alone (controls) or backbone plus one open-label investigational agent. Patients were enrolled to the arms described between July 30, 2020 and June 11, 2021 in 20 medical centres in the United States. The platform contained up to four potentially available investigational agents and controls available for randomisation during a single time-period. The two primary endpoints were time-to-recovery (<6 L/min oxygen for two consecutive days) and mortality. Data were evaluated biweekly in comparison to pre-specified criteria for graduation (i.e., likely efficacy), futility, and safety, with an adaptive sample size of 40-125 individuals per agent and a Bayesian analytical approach. Criteria were designed to achieve rapid screening of agents and to identify large benefit signals. Concurrently enrolled controls were used for all analyses. https://clinicaltrials.gov/ct2/show/NCT04488081. Findings The first 7 agents evaluated were cenicriviroc (CCR2/5 antagonist; n = 92), icatibant (bradykinin antagonist; n = 96), apremilast (PDE4 inhibitor; n = 67), celecoxib/famotidine (COX2/histamine blockade; n = 30), IC14 (anti-CD14; n = 67), dornase alfa (inhaled DNase; n = 39) and razuprotafib (Tie2 agonist; n = 22). Razuprotafib was dropped from the trial due to feasibility issues. In the modified intention-to-treat analyses, no agent met pre-specified efficacy/graduation endpoints with posterior probabilities for the hazard ratios [HRs] for recovery ≤1.5 between 0.99 and 1.00. The data monitoring committee stopped Celecoxib/Famotidine for potential harm (median posterior HR for recovery 0.5, 95% credible interval [CrI] 0.28-0.90; median posterior HR for death 1.67, 95% CrI 0.79-3.58). Interpretation None of the first 7 agents to enter the trial met the prespecified criteria for a large efficacy signal. Celecoxib/Famotidine was stopped early for potential harm. Adaptive platform trials may provide a useful approach to rapidly screen multiple agents during a pandemic. Funding Quantum Leap Healthcare Collaborative is the trial sponsor. Funding for this trial has come from: the COVID R&D Consortium, Allergan, Amgen Inc., Takeda Pharmaceutical Company, Implicit Bioscience, Johnson & Johnson, Pfizer Inc., Roche/Genentech, Apotex Inc., FAST Grant from Emergent Venture George Mason University, The DoD Defense Threat Reduction Agency (DTRA), The Department of Health and Human ServicesBiomedical Advanced Research and Development Authority (BARDA), and The Grove Foundation. Effort sponsored by the U.S. Government under Other Transaction number W15QKN-16-9-1002 between the MCDC, and the Government.
Collapse
|
120
|
Taher A, Lashkari M, Keramat F, Hashemi SH, Sedighi L, Poorolajal J, Mehrpooya M. Comparison of the efficacy of equivalent doses of dexamethasone, methylprednisolone, and hydrocortisone for treatment of COVID-19-related acute respiratory distress syndrome: a prospective three-arm randomized clinical trial. Wien Med Wochenschr 2023; 173:140-151. [PMID: 36624180 PMCID: PMC9838299 DOI: 10.1007/s10354-022-00993-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/24/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND This prospective controlled clinical trial aimed to compare the efficacy of methylprednisolone, dexamethasone, and hydrocortisone at equivalent doses in patients with severe COVID-19. METHODS In total, 106 patients with mild to moderate COVID-19-related acute respiratory distress syndrome (ARDS) were randomized to receive either dexamethasone (6 mg once a day), methylprednisolone (16 mg twice a day), or hydrocortisone (50 mg thrice a day) for up to 10 days. All participants received a standard of care for COVID-19. The primary and secondary efficacy outcomes included all-cause 28-day mortality, clinical status on day 28 assessed using the World Health Organization (WHO) eight-category ordinal clinical scale, number of patients requiring mechanical ventilation and intensive care unit (ICU) care, number of ventilator-free days, length of hospital and ICU stay, change in PaO2:FiO2 ratios during the first 5 days after treatment, and incidence of serious adverse events. P-values below 0.008 based on Bonferroni's multiple-testing correction method were considered statistically significant. RESULTS According to the obtained results, there was a trend toward more favorable clinical outcomes in terms of needing mechanical ventilation and ICU care, number of ventilator-free days, change in PaO2:FiO2 ratios during the first 5 days after treatment, clinical status score at day 28, length of ICU and hospital stay, and overall 28-day mortality in patients receiving dexamethasone compared to those receiving methylprednisolone or hydrocortisone; however, likely due to the study's small sample size, the difference between groups reached a significant level only in the case of clinical status score on day 28 (p-value = 0.003). There was no significant difference in the incidence of serious adverse events between the study groups. CONCLUSION Based on the results, severe cases of COVID-19 treated with dexamethasone might have a better clinical status at 28-day follow-up compared to methylprednisolone and hydrocortisone at an equivalent dose. Larger multicenter trials are required to confirm our findings.
Collapse
Affiliation(s)
- Abbas Taher
- Department of Anesthesiology and Critical Care, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marjan Lashkari
- Department of Anesthesiology and Critical Care, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fariba Keramat
- Department Infectious Disease, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyyed Hamid Hashemi
- Department Infectious Disease, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ladan Sedighi
- Department of medical and surgical nursing, school of nursing and midwifery, Shahid Beheshti university of Medical Sciences, Tehran, Iran
| | - Jalal Poorolajal
- Department of Epidemiology, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Maryam Mehrpooya
- Department of Clinical Pharmacy, School of Pharmacy, Medicinal Plants and Natural Products Research Center, Hamadan University of Medical Sciences, Shahid Fahmideh Ave, 6517838678 Hamadan, Iran
| |
Collapse
|
121
|
Dahmer M, Jennings A, Parker M, Sanchez-Pinto LN, Thompson A, Traube C, Zimmerman JJ. Pediatric Critical Care in the Twenty-first Century and Beyond. Crit Care Clin 2023; 39:407-425. [PMID: 36898782 DOI: 10.1016/j.ccc.2022.09.013] [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: 11/16/2022]
Abstract
Pediatric critical care addresses prevention, diagnosis, and treatment of organ dysfunction in the setting of increasingly complex patients, therapies, and environments. Soon burgeoning data science will enable all aspects of intensive care: driving facilitated diagnostics, empowering a learning health-care environment, promoting continuous advancement of care, and informing the continuum of critical care outside the intensive care unit preceding and following critical illness/injury. Although novel technology will progressively objectify personalized critical care, humanism, practiced at the bedside, defines the essence of pediatric critical care now and in the future.
Collapse
Affiliation(s)
- Mary Dahmer
- Division of Critical Care, Department of Pediatrics, University of Michigan, 1500 East Medical Center Drive, F6790/5243, Ann Arbor, MI, USA
| | - Aimee Jennings
- Division of Critical Care Medicine, Advanced Practice, FA.2.112, Seattle Children's Hospital, 4800 Sandpoint Way Northeast, Seattle, WA 98105, USA
| | - Margaret Parker
- Department of Pediatrics, Stony Brook University, 7762 Bloomfield Road, Easton, MD 21601, USA
| | - Lazaro N Sanchez-Pinto
- Department of Pediatrics, Ann and Robert H Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, 225 East Chicago Avenue, Box 73, Chicago, IL 60611-2605, USA
| | - Ann Thompson
- Department of Critical Care Medicine, University of Pittsburgh, 3550 Terrace Street, Pittsburgh, PA 15261, USA
| | - Chani Traube
- Department of Pediatrics, Weill Cornell Medicine, 525 East 68th Street, Box 225, New York, NY 10065, USA
| | - Jerry J Zimmerman
- Department of Pediatrics, FA.2.300B Seattle Children's Hospital, 4800 Sandpoint Way Northeast, Seattle, WA 98105, USA; Pediatric Critical Care Medicine, Seattle Children's Hospital, Harborview Medical Center, University of Washington, School of Medicine, FA.2.300B, Seattle Children's Hospital, 4800 Sand Point Way Northeast, Seattle, WA 98105, USA.
| |
Collapse
|
122
|
Jawa NA, Maslove DM. Bayes' Theorem in Neurocritical Care: Principles and Practice. Neurocrit Care 2023; 38:517-528. [PMID: 36635494 DOI: 10.1007/s12028-022-01665-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/13/2022] [Indexed: 01/13/2023]
Abstract
Patients with critical neurological illness are diverse. As a result of the heterogeneity of this patient population, standardized approaches to patient management might not confer benefit. A precision medicine approach to neurocritical care is therefore urgently needed to improve our understanding of neurocritical illness and the care provided to this vulnerable cohort. Research designs and approaches based on Bayesian models have the potential to meet this need, as they are specifically designed to evolve with emerging evidence. This adaptability provides a benefit over the popular frequentist statistical approach, as it provides a way of adjusting hypotheses and trial procedures to maximize efficacy. This review summarizes the current state of knowledge on Bayes' theorem, and its potential applications to the field of neurocritical care. We review the basic principles underlying Bayes' theorem, compare the use of Bayesian versus frequentist statistics in medicine, and discuss the relevance of Bayesian statistics to the field of neuroscience and to clinical research. Finally, we explore the potential benefits of employing Bayesian methods within the field of neurocritical care as a steppingstone toward implementing precision medicine approaches to improve patient outcomes for complex, heterogeneous disorders.
Collapse
Affiliation(s)
- Natasha A Jawa
- Center for Neuroscience Studies, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - David M Maslove
- Department of Critical Care Medicine, Queen's University, Kingston, ON, Canada.
- Department of Medicine, Queen's University, Kingston, ON, Canada.
- Kingston Health Sciences Centre, Kingston, ON, Canada.
| |
Collapse
|
123
|
Pitre T, Su J, Mah J, Helmeczi W, Danho S, Plaxton W, Giilck S, Rochwerg B, Zeraatkar D. Higher- versus Lower-Dose Corticosteroids for Severe to Critical COVID-19: A Systematic Review and Dose-Response Meta-analysis. Ann Am Thorac Soc 2023; 20:596-604. [PMID: 36449393 PMCID: PMC10112407 DOI: 10.1513/annalsats.202208-720oc] [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: 08/22/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022] Open
Abstract
Rationale: Corticosteroids are standard of care for patients with severe coronavirus disease (COVID-19). However, the optimal dose is uncertain. Objectives: To compare higher doses of corticosteroids with lower doses in patients with COVID-19. Methods: We searched MEDLINE, Embase, Cochrane Central Register of Controlled Trials, MedRxiv, and Web of Science from inception to August 1, 2022, for trials that randomized patients with severe-to-critical COVID-19 to corticosteroids, standard care, or placebo. Reviewers, working in duplicate, screened references, extracted data, and assessed risk of bias using a modified version of the Cochrane risk of bias 2.0 tool. We performed a dose-response meta-analysis and used the Grading of Recommendations Assessment, Development and Evaluation (GRADE) framework to assess the certainty of evidence. We present our results both in relative risk and absolute risk difference per 1,000, with 95% confidence intervals (CIs). Results: We included 20 trials, with 10,155 patients. We show that, compared with lower-dose corticosteroids, higher-dose corticosteroids probably reduce mortality (absolute risk difference, 14 fewer deaths per 1,000 [95% CI, 26 fewer to 2 fewer]; moderate certainty) and may reduce the need for mechanical ventilation (absolute risk difference, 11.6 fewer per 1,000 [95% CI, 23.2 fewer to 6.9 more]; low certainty). The effect of corticosteroids on nosocomial infections is uncertain (16.7 fewer infections per 1,000 [95% CI, 5.4 fewer to 25.0 fewer]; very low certainty). Conclusions: Relatively higher doses of corticosteroids may be beneficial in patients with severe-to-critical COVID-19 and may not increase the risk of nosocomial infections.
Collapse
Affiliation(s)
| | | | - Jasmine Mah
- Department of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Wryan Helmeczi
- Division of Internal Medicine, University of Ottawa, Ottawa, Ontario, Canada; and
| | | | | | - Stephen Giilck
- Department of Medicine, Grand River Hospital, Kitchener, Ontario, Canada
| | - Bram Rochwerg
- Department of Health Research Methods Evidence and Impact, and
| | - Dena Zeraatkar
- Department of Health Research Methods Evidence and Impact, and
- Department of Anesthesiology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
124
|
Pulse Methylprednisolone Versus Dexamethasone in COVID-19: A Multicenter Cohort Study. Crit Care Explor 2023; 5:e0886. [PMID: 36998527 PMCID: PMC10047604 DOI: 10.1097/cce.0000000000000886] [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: 03/29/2023] Open
Abstract
Although pulse (high-dose) methylprednisolone therapy can hypothetically control immune system flare-ups effectively, the clinical benefit of pulse methylprednisolone compared with dexamethasone in COVID-19 remains inconclusive.
Collapse
|
125
|
Liao Q, Pu Y, Jin X, Zhuang Z, Xu X, Ren X, Liu G, Ding Q. Physiological and clinical variables identify ARDS classes and therapeutic heterogeneity to glucocorticoids: a retrospective study. BMC Pulm Med 2023; 23:92. [PMID: 36944959 PMCID: PMC10028772 DOI: 10.1186/s12890-023-02384-w] [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: 10/14/2022] [Accepted: 03/13/2023] [Indexed: 03/23/2023] Open
Abstract
OBJECTIVE We aimed to identify new classes in acute respiratory distress syndrome (ARDS) using physiological and clinical variables and to explore heterogeneity in the effects of glucocorticoid therapy between classes. METHODS Using the Medical Information Mart for Intensive Care-IV database, we identified patients with ARDS. Potential profile analysis was used to identify classes with physiological and clinical data as delineating variables. Baseline characteristics and clinical outcomes were compared between classes. The effect of glucocorticoid treatment was explored by stratifying by class and glucocorticoid treatment. RESULTS From 2008 to 2019, 1104 patients with ARDS were enrolled in the study. The 2-class potential analysis model had the best fit (P < 0.0001), with 78% of patients falling into class 1 and 22% into class 2. Additional classes did not improve the model fit. Patients in class 2 had higher anion gap, lactate, creatinine, and glucose levels and lower residual base, blood pressure, and bicarbonate compared with class 1. In-hospital mortality and 28-day mortality were significantly higher among patients in class 2 than those in class 1 (P < 0.001). Heterogeneity of glucocorticoid treatment was observed, stratified by class and treatment, with no significant effect in class 1 (P = 0.496), increased mortality in class 2 (P = 0.001), and a significant interaction (P = 0.0381). In class 2, 28-day survival was significantly lower with glucocorticoid treatment compared with no hormone treatment (P = 0.001). CONCLUSION We used clinical and physiological variables to identify two classes of non-COVID-19-associated ARDS with different baseline characteristics and clinical outcomes. The response to glucocorticoid therapy varied among different classes of patients.
Collapse
Affiliation(s)
- Qingbo Liao
- The Affiliated Suzhou Hospital of Nanjing Medical University, 26 Daoqian Road, Suzhou, 215000, PR China
- Gusu School, Nanjing Medical University, 458 Shizi Road, Suzhou, 215000, PR China
| | - Yufan Pu
- The Affiliated Suzhou Hospital of Nanjing Medical University, 26 Daoqian Road, Suzhou, 215000, PR China
- Gusu School, Nanjing Medical University, 458 Shizi Road, Suzhou, 215000, PR China
| | - Xiaoer Jin
- The Affiliated Suzhou Hospital of Nanjing Medical University, 26 Daoqian Road, Suzhou, 215000, PR China
- Gusu School, Nanjing Medical University, 458 Shizi Road, Suzhou, 215000, PR China
| | - Zhiwei Zhuang
- Department of Emergency, Suzhou Municipal Hospital, 26 Daoqian Road, Suzhou, 215000, PR China
| | - Xiaowen Xu
- Department of Emergency, Suzhou Municipal Hospital, 26 Daoqian Road, Suzhou, 215000, PR China
| | - Xiaoqiang Ren
- Department of Emergency, Suzhou Municipal Hospital, 26 Daoqian Road, Suzhou, 215000, PR China
| | - Gaoqing Liu
- First Affiliated Hospital of Suzhou University, 899 Pinghai Road, Suzhou, 215000, PR China
| | - Qi Ding
- The Affiliated Suzhou Hospital of Nanjing Medical University, 26 Daoqian Road, Suzhou, 215000, PR China.
- Gusu School, Nanjing Medical University, 458 Shizi Road, Suzhou, 215000, PR China.
- Department of Emergency, Suzhou Municipal Hospital, 26 Daoqian Road, Suzhou, 215000, PR China.
| |
Collapse
|
126
|
COVID-19 Management Strategies in Solid Organ Transplant Recipients. Infect Dis Clin North Am 2023:S0891-5520(23)00024-7. [PMID: 37142512 PMCID: PMC10028355 DOI: 10.1016/j.idc.2023.03.003] [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: 03/24/2023]
Abstract
Solid organ transplant recipients are at high risk of severe coronavirus disease-2019 (COVID-19). If left untreated, it results in high rates of hospitalization, intensive care unit admission and death. Early diagnosis of COVID-19 is essential to ensure the early administration of therapeutics. Treatment of mild-to-moderate COVID-19 with remdesivir, ritonavir-boosted nirmatrelvir, or an anti-spike neutralizing monoclonal antibody may prevent progression to severe and critical COVID-19. Among patients with severe and critical COVID-19, treatment with intravenous remdesivir and immunomodulation is recommended. This review article discusses strategies in the management of solid organ transplant recipients with COVID-19.
Collapse
|
127
|
Akinosoglou K, Schinas G, Rigopoulos EA, Polyzou E, Tzouvelekis A, Adonakis G, Gogos C. COVID-19 Pharmacotherapy in Pregnancy: A Literature Review of Current Therapeutic Choices. Viruses 2023; 15:787. [PMID: 36992497 PMCID: PMC10054527 DOI: 10.3390/v15030787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
The clinical management of COVID-19 in pregnant women, who are considered a vulnerable population, remains uncertain even as the pandemic subsides. SARS-CoV-2 affects pregnant individuals in multiple ways and has been associated with severe maternal morbidity and mortality, as well as neonatal complications. The unique anatomy and physiology of gestation make managing COVID-19 in this population a complex and challenging task, emphasizing the importance of spreading knowledge and expertise in this area. Therapeutic interventions require distinct clinical consideration, taking into account differences in pharmacokinetics, vertical transmission, drug toxicities, and postnatal care. Currently, there is limited data on antiviral and immunomodulating COVID-19 pharmacotherapy in pregnancy. Some medication has been shown to be safe and well tolerated among pregnant women with COVID-19; however, the lack of randomized clinical trials and studies in this patient population is evident. Available vaccines are considered safe and effective, with no evidence of harm to the fetus, embryo development, or short-term postnatal development. Pregnant women should be counseled about the risks of SARS-CoV-2 infection and informed of available ways to protect themselves and their families. Effective treatments for COVID-19 should not be withheld from pregnant individuals, and more research is needed to ensure the best outcomes.
Collapse
Affiliation(s)
- Karolina Akinosoglou
- Department of Medicine, University of Patras, 26504 Patras, Greece
- Department of Infectious Diseases, University General Hospital of Patras, 26504 Patras, Greece
- Division of Internal Medicine, University General Hospital of Patras, 26504 Patras, Greece
| | - Georgios Schinas
- Department of Medicine, University of Patras, 26504 Patras, Greece
| | | | - Eleni Polyzou
- Department of Medicine, University of Patras, 26504 Patras, Greece
- Division of Internal Medicine, University General Hospital of Patras, 26504 Patras, Greece
| | - Argyrios Tzouvelekis
- Department of Medicine, University of Patras, 26504 Patras, Greece
- Department of Pulmonology, University General Hospital of Patras, 26504 Patras, Greece
| | - George Adonakis
- Department of Medicine, University of Patras, 26504 Patras, Greece
- Department of Obstetrics and Gynecology, University General Hospital of Patras, 26504 Patras, Greece
| | | |
Collapse
|
128
|
Coelho L, Falcão F, Póvoa P, Viegas E, Martins AP, Carmo E, Fonseca C, Campos L, Mansinho K, Carmo I, Soares J, Solano M, Mendes D, Miranda AC, Carvalho A, Mirco A, Farinha H, Aldir I, Correia J. Remdesivir and corticosteroids in the treatment of hospitalized COVID-19 patients. Sci Rep 2023; 13:4482. [PMID: 36934143 PMCID: PMC10024012 DOI: 10.1038/s41598-023-31544-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 03/14/2023] [Indexed: 03/20/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a pandemic infection caused by the newly discovered severe acute respiratory syndrome coronavirus 2. Remdesivir (RDV) and corticosteroids are used mainly in COVID-19 patients with acute respiratory failure. The main objective of the study was to assess the effectiveness of remdesivir with and without corticosteroids in the treatment of COVID-19 patients. We conducted a prospective observational study, including adult patients consecutively hospitalized with confirmed COVID-19 and acute respiratory failure. Patients were divided according to treatment strategy: RDV alone versus RDV with corticosteroids. The primary outcome was the time to recovery in both treatment groups. We included 374 COVID-19 adult patients, 184 were treated with RDV, and 190 were treated with RDV and corticosteroid. Patients in the RDV group had a shorter time to recovery in comparison with patients in the RDV plus corticosteroids group at 28 days after admission [11 vs. 16 days (95% confidence Interval 9.7-12.8; 14.9-17.1; p = .016)]. Patients treated with RDV alone had a shorter length of hospital stay. The use of corticosteroids as adjunctive therapy of RDV was not associated with improvement in mortality of COVID-19 patients.
Collapse
Affiliation(s)
- Luís Coelho
- Polyvalent Intensive Care Unit, Hospital de São Francisco Xavier, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal.
- NOVA Medical School, CHRC, New University of Lisbon, Lisbon, Portugal.
| | - Fatima Falcão
- Pharmacy Department, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- Faculty of Pharmacy of the University of Lisbon, Lisbon, Portugal
| | - Pedro Póvoa
- Polyvalent Intensive Care Unit, Hospital de São Francisco Xavier, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- NOVA Medical School, CHRC, New University of Lisbon, Lisbon, Portugal
- Center for Clinical Epidemiology and Research Unit of Clinical Epidemiology, OUH Odense University Hospital, Odense, Denmark
| | - Erica Viegas
- Pharmacy Department, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- Faculty of Pharmacy of the University of Lisbon, Lisbon, Portugal
| | - Antonio Pais Martins
- Surgical Intensive Care Unit, Hospital de S. Francisco Xavier, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Eduarda Carmo
- Polyvalent Intensive Care Unit, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Candida Fonseca
- NOVA Medical School, CHRC, New University of Lisbon, Lisbon, Portugal
- Internal Medicine, Hospital de S. Francisco Xavier, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Luis Campos
- NOVA Medical School, CHRC, New University of Lisbon, Lisbon, Portugal
- Internal Medicine, Hospital de S. Francisco Xavier, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Kamal Mansinho
- NOVA Medical School, CHRC, New University of Lisbon, Lisbon, Portugal
- Infecciology, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Inês Carmo
- Pharmacy Department, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Joana Soares
- Pharmacy Department, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Mariana Solano
- Pharmacy Department, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Dina Mendes
- Pharmacy Department, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Ana Cláudia Miranda
- Infecciology, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Antonio Carvalho
- Internal Medicine, Hospital de S. Francisco Xavier, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- Pharmacy and Therapeutics Committee, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Ana Mirco
- Faculty of Pharmacy of the University of Lisbon, Lisbon, Portugal
- Pharmacy and Therapeutics Committee, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Helena Farinha
- Faculty of Pharmacy of the University of Lisbon, Lisbon, Portugal
- Pharmacy and Therapeutics Committee, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - Isabel Aldir
- NOVA Medical School, CHRC, New University of Lisbon, Lisbon, Portugal
- Infecciology, Hospital de Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- Pharmacy and Therapeutics Committee, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| | - José Correia
- Pharmacy and Therapeutics Committee, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
| |
Collapse
|
129
|
Modlin C, Sugarman J, Chongwe G, Kass N, Nazziwa W, Tegli J, Shrestha P, Ali J. Towards achieving transnational research partnership equity: lessons from implementing adaptive platform trials in low- and middle-income countries. Wellcome Open Res 2023. [DOI: 10.12688/wellcomeopenres.18915.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
Background: Use of adaptive clinical trials, particularly adaptive platform trials, has grown exponentially in response to the coronavirus disease (COVID-19) pandemic. Implementation of these trials in low- and middle-income countries (LMICs) has been fostered through the formation or modification of transnational research partnerships, typically between research groups from LMICs and high-income countries (HICs). While these partnerships are important to promote collaboration and overcome the structural and economic disadvantages faced by LMIC health researchers, it is critical to focus attention on the multiple dimensions of partnership equity. Methods: Based on informal literature reviews and meetings with leaders of one of the multinational COVID-19 adaptive platform trials, we describe what can be learned about research partnership equity from these experiences. Results: We organize these considerations into eight thematic categories: 1) epistemic structures, 2) funding, 3) ethics oversight, 4) regulatory oversight, 5) leadership, 6) post-trial access to interventions, data, and specimens, 7) knowledge translation, and 8) research capacity strengthening and maintenance. Within each category we review the normative claims that support its relevance to research partnership equity followed by discussion of how adaptive platform trials highlight new dimensions, considerations, or challenges. Conclusion: These observations provide insight into procedural and substantive equity-building measures within transnational global health research partnerships more broadly.
Collapse
|
130
|
Bonilla H, Peluso MJ, Rodgers K, Aberg JA, Patterson TF, Tamburro R, Baizer L, Goldman JD, Rouphael N, Deitchman A, Fine J, Fontelo P, Kim AY, Shaw G, Stratford J, Ceger P, Costantine MM, Fisher L, O’Brien L, Maughan C, Quigley JG, Gabbay V, Mohandas S, Williams D, McComsey GA. Therapeutic trials for long COVID-19: A call to action from the interventions taskforce of the RECOVER initiative. Front Immunol 2023; 14:1129459. [PMID: 36969241 PMCID: PMC10034329 DOI: 10.3389/fimmu.2023.1129459] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/06/2023] [Indexed: 03/11/2023] Open
Abstract
Although most individuals recover from acute SARS-CoV-2 infection, a significant number continue to suffer from Post-Acute Sequelae of SARS-CoV-2 (PASC), including the unexplained symptoms that are frequently referred to as long COVID, which could last for weeks, months, or even years after the acute phase of illness. The National Institutes of Health is currently funding large multi-center research programs as part of its Researching COVID to Enhance Recover (RECOVER) initiative to understand why some individuals do not recover fully from COVID-19. Several ongoing pathobiology studies have provided clues to potential mechanisms contributing to this condition. These include persistence of SARS-CoV-2 antigen and/or genetic material, immune dysregulation, reactivation of other latent viral infections, microvascular dysfunction, and gut dysbiosis, among others. Although our understanding of the causes of long COVID remains incomplete, these early pathophysiologic studies suggest biological pathways that could be targeted in therapeutic trials that aim to ameliorate symptoms. Repurposed medicines and novel therapeutics deserve formal testing in clinical trial settings prior to adoption. While we endorse clinical trials, especially those that prioritize inclusion of the diverse populations most affected by COVID-19 and long COVID, we discourage off-label experimentation in uncontrolled and/or unsupervised settings. Here, we review ongoing, planned, and potential future therapeutic interventions for long COVID based on the current understanding of the pathobiological processes underlying this condition. We focus on clinical, pharmacological, and feasibility data, with the goal of informing future interventional research studies.
Collapse
Affiliation(s)
- Hector Bonilla
- Department of Medicine and Infectious Diseases, Stanford University, Palo Alto, CA, United States
| | - Michael J. Peluso
- Department of Medicine and Infectious Diseases, University of California, San Francisco, San Francisco, CA, United States
| | - Kathleen Rodgers
- Center for Innovations in Brain Science, University of Arizona, Tucson, AZ, United States
| | - Judith A. Aberg
- Department of Medicine, Infectious Diseases, Icahn School of Medicine at Mount Sinai, Chief, Division of Infectious Disease, New York, NY, United States
| | - Thomas F. Patterson
- Department of Medicine, Infectious Diseases, The University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Robert Tamburro
- Division of Intramural Research, National Institute of Health, Bethesda, MD, United States
| | - Lawrence Baizer
- National Heart Lung and Blood Institute, Division of Lung Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jason D. Goldman
- Department of Medicine, Organ Transplant and Liver Center, Swedish Medical Center, Seattle, WA, United States
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Nadine Rouphael
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States
| | - Amelia Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Jeffrey Fine
- Department of Rehabilitation Medicine at New York University (NYU) Grossman School of Medicine, Physical Medicine and Rehabilitation Service, New York University (NYU), New York University Medical Center, New York, NY, United States
| | - Paul Fontelo
- Applied Clinical Informatics Branch, National Library of Medicine, National Institutes of Health, Bethesda, MD, United States
| | - Arthur Y. Kim
- Department of Medicine at Harvard Medical School, Division of Infectious Disease, Boston, MA, United States
| | - Gwendolyn Shaw
- Research Triangle Institute (RTI), International, Durham, NC, United States
| | - Jeran Stratford
- Research Triangle Institute (RTI), International, Durham, NC, United States
| | - Patricia Ceger
- Research Triangle Institute (RTI), International, Durham, NC, United States
| | - Maged M. Costantine
- Department of Obstetrics and Gynecology, The Ohio State University, Columbus, OH, United States
| | - Liza Fisher
- Long COVID Families, Houston, TX, United States
| | - Lisa O’Brien
- Utah Covid-19 Long Haulers, Salt Lake City, UT, United States
| | | | - John G. Quigley
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Vilma Gabbay
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, United States
| | - Sindhu Mohandas
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - David Williams
- Department of Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Grace A. McComsey
- Department of Pediatrics and Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
131
|
Meyer EL, Mesenbrink P, Di Prospero NA, Pericàs JM, Glimm E, Ratziu V, Sena E, König F. Designing an exploratory phase 2b platform trial in NASH with correlated, co-primary binary endpoints. PLoS One 2023; 18:e0281674. [PMID: 36893087 PMCID: PMC9997886 DOI: 10.1371/journal.pone.0281674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/28/2023] [Indexed: 03/10/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is the progressive form of nonalcoholic fatty liver disease (NAFLD) and a disease with high unmet medical need. Platform trials provide great benefits for sponsors and trial participants in terms of accelerating drug development programs. In this article, we describe some of the activities of the EU-PEARL consortium (EU Patient-cEntric clinicAl tRial pLatforms) regarding the use of platform trials in NASH, in particular the proposed trial design, decision rules and simulation results. For a set of assumptions, we present the results of a simulation study recently discussed with two health authorities and the learnings from these meetings from a trial design perspective. Since the proposed design uses co-primary binary endpoints, we furthermore discuss the different options and practical considerations for simulating correlated binary endpoints.
Collapse
Affiliation(s)
- Elias Laurin Meyer
- Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Peter Mesenbrink
- Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, United States of America
| | | | - Juan M. Pericàs
- Liver Unit, Internal Medicine Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute for Research (VHIR), Barcelona, Spain
- Centros de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), ISCIII, Madrid, Spain
| | - Ekkehard Glimm
- Novartis Pharma AG, Basel, Switzerland
- Institute of Biometry and Medical Informatics, University of Magdeburg, Magdeburg, Germany
| | - Vlad Ratziu
- Assistance Publique-Hôpitaux de Paris, Hôpital Pitie-Salpetriere, University of Paris, Paris, France
| | - Elena Sena
- Liver Unit, Internal Medicine Department, Vall d’Hebron University Hospital, Vall d’Hebron Institute for Research (VHIR), Barcelona, Spain
| | - Franz König
- Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
132
|
Ali Abdelhamid Y, Deane A, Bellomo R. Recent advances in critical care. Med J Aust 2023; 218:153-156. [PMID: 36773964 DOI: 10.5694/mja2.51850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/06/2022] [Accepted: 01/18/2023] [Indexed: 02/13/2023]
Affiliation(s)
| | - Adam Deane
- University of Melbourne, Melbourne, VIC.,Royal Melbourne Hospital, Melbourne, VIC
| | - Rinaldo Bellomo
- University of Melbourne, Melbourne, VIC.,Austin Hospital, Melbourne, VIC
| |
Collapse
|
133
|
Estrup S, Barot E, Mortensen CB, Anthon CT, Crescioli E, Kjaer MBN, Vesterlund GK, Bruun CRL, Collet MO, Rasmussen BS, Sivapalan P, Poulsen LM, Møller MH, Perner A, Granholm A. Patient and public involvement in contemporary large intensive care trials: A meta-epidemiological study. Acta Anaesthesiol Scand 2023; 67:256-263. [PMID: 36537664 DOI: 10.1111/aas.14183] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/22/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Patient and public involvement in randomised clinical trials has received increased focus, including in intensive care trials, but the frequency, method and extent is unknown. This meta-epidemiological study investigated patient and public involvement in contemporary, large ICU trials. METHODS We systematically searched PubMed for large (≥225 randomised patients), contemporary trials (published between 1 January 2019 and 31 January 2022) assessing interventions in adult patients in ICU settings. Abstracts and full-text articles were assessed independently and in duplicate. Data were extracted using a pre-defined, pilot-tested data extraction form with details on trials, patient and public involvement including categories and numbers of individuals involved, methods of involvement, and trial stage(s) with involvement. Trials authors were contacted as necessary. RESULTS We included 100 trials, with 18 using patient and public involvement; these were larger and conducted in more centres than trials without patient and public involvement. Among trials with patient and public involvement, patients (in 14/18 trials), clinicians (13 trials), and family members (12 trials) were primarily involved, mainly in the development of research design (15 trials) and development of research focus (13 trials) stages and mostly by discussion (12 trials) and solo interviews (10 trials). A median of 65 individuals (range 1-6894) were involved. CONCLUSIONS We found patient and public involvement in a fifth of large, contemporary ICU trials. Primarily patients, families, and clinicians were included, particularly in the trial planning stages and mostly through interviews and discussions. Increased patient and public involvement in ICU trials is warranted.
Collapse
Affiliation(s)
- Stine Estrup
- Department of Anaesthesiology and Intensive Care, Zealand University Hospital, Køge, Denmark
| | - Emily Barot
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | | | - Carl Thomas Anthon
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Elena Crescioli
- Department of Anaesthesiology and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
| | | | - Gitte Kingo Vesterlund
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | | | - Marie Oxenbøll Collet
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Bodil Steen Rasmussen
- Department of Anaesthesiology and Intensive Care, Aalborg University Hospital, Aalborg, Denmark
| | - Praleene Sivapalan
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Lone Musaeus Poulsen
- Department of Anaesthesiology and Intensive Care, Zealand University Hospital, Køge, Denmark
| | - Morten Hylander Møller
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Anders Perner
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Anders Granholm
- Department of Intensive Care, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
134
|
Johns H, Campbell B, Bernhardt J, Churilov L. Generalised pairwise comparisons for trend: An extension to the win ratio and win odds for dose-response and prognostic variable analysis with arbitrary statements of outcome preference. Stat Methods Med Res 2023; 32:609-625. [PMID: 36573043 DOI: 10.1177/09622802221146306] [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: 12/28/2022]
Abstract
The win ratio is a novel approach for handling complex patient outcomes that have seen considerable interest in the medical statistics literature, and operates by considering all-to-all pairwise statements of preference on outcomes. Recent extensions to the method have focused on the two-group case, with few developments made for considering the impact of a well-ordered explanatory variable, which would allow for dose-response analysis or the analysis of links between complex patient outcomes and prognostic variables. Where such methods have been developed, they are semiparametric methods that can only be applied to survival outcomes. In this article, we introduce the generalised pairwise comparison for trend, a modified form of Agresti's generalised odds ratio. This approach is capable of considering arbitrary statements of preference, thus enabling its use across all types of outcome data. We provide a simulation study validating the approach and illustrate it with three clinical applications in stroke research.
Collapse
Affiliation(s)
- Hannah Johns
- Melbourne Medical School, University of Melbourne, Melbourne, Australia
| | - Bruce Campbell
- Department of Medicine and Neurology, Melbourne Brain Centre and Royal Melbourne Hospital, University of Melbourne, Melbourne, Australia
| | - Julie Bernhardt
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Leonid Churilov
- Melbourne Medical School, University of Melbourne, Melbourne, Australia
| |
Collapse
|
135
|
Issak ER, Amin MM. Timing of corticosteroids in non-severe non-hospitalized COVID-19 patients: open-label, two-center, randomized controlled study (TICS-COV19 study). Korean J Intern Med 2023; 38:207-217. [PMID: 36646988 PMCID: PMC9993104 DOI: 10.3904/kjim.2022.232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND/AIMS Corticosteroids (CSs) are frequently used in coronavirus disease 2019 (COVID-19); however, their utility remains controversial in mild to moderate cases. The timing of CSs initiation during the disease course remains unaddressed. The study aims to evaluate the impact of early CSs in non-severe COVID-19. METHODS A randomized controlled, open-label study was conducted on 754 COVID-19 patients randomized into a study group (n = 377) in which patients received CSs with COVID-19 protocol and a control group (n = 377) in which patients received COVID-19 protocol only. RESULTS Both groups were comparable regarding baseline characteristics, presenting symptoms, and inflammatory markers. The composite endpoint (need for O2, need for hospitalization or 28-day mortality) was significantly (p = 0.004) lower in the CS group 42 (11.14%) versus the control group 70 (18.67%) with odds ratio 0.55 (95% confidence interval [CI], 0.36 to 0.83), absolute risk reduction 7.53% (95% CI, 2.46% to 12.59%) and number needed to treat of 13.29 (95% CI, 7.94 to 40.61). Regarding severity at day 10, only (11.1%) of the study group patients were severe versus (18.7%) of the control group patients (p < 0.001). The median time-to-return to daily activity in the CS group was 8.0 days, while in the control group, it was 22.0 days (p < 0.001). CONCLUSION In non-severe COVID-19, CS may decrease hospitalization, severity, and mortality.
Collapse
Affiliation(s)
- Emad R. Issak
- Department of Internal Medicine, Allergy and Clinical Immunology, Faculty of Medicine, Ain Shams University, Cairo,
Egypt
- Department of Internal Medicine, Asalam Center, Cairo,
Egypt
| | - Mariam M. Amin
- Department of Internal Medicine, Allergy and Clinical Immunology, Faculty of Medicine, Ain Shams University, Cairo,
Egypt
| |
Collapse
|
136
|
Li S, Zhang Y, Guan Z, Ye M, Li H, You M, Zhou Z, Zhang C, Zhang F, Lu B, Zhou P, Peng K. SARS-CoV-2 Z-RNA activates the ZBP1-RIPK3 pathway to promote virus-induced inflammatory responses. Cell Res 2023; 33:201-214. [PMID: 36650286 PMCID: PMC9844202 DOI: 10.1038/s41422-022-00775-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
SARS-CoV-2 infection can trigger strong inflammatory responses and cause severe lung damage in COVID-19 patients with critical illness. However, the molecular mechanisms by which the infection induces excessive inflammatory responses are not fully understood. Here, we report that SARS-CoV-2 infection results in the formation of viral Z-RNA in the cytoplasm of infected cells and thereby activates the ZBP1-RIPK3 pathway. Pharmacological inhibition of RIPK3 by GSK872 or genetic deletion of MLKL reduced SARS-CoV-2-induced IL-1β release. ZBP1 or RIPK3 deficiency leads to reduced production of both inflammatory cytokines and chemokines during SARS-CoV-2 infection both in vitro and in vivo. Furthermore, deletion of ZBP1 or RIPK3 alleviated SARS-CoV-2 infection-induced immune cell infiltration and lung damage in infected mouse models. These results suggest that the ZBP1-RIPK3 pathway plays a critical role in SARS-CoV-2-induced inflammatory responses and lung damage. Our study provides novel insights into how SARS-CoV-2 infection triggers inflammatory responses and lung pathology, and implicates the therapeutic potential of targeting ZBP1-RIPK3 axis in treating COVID-19.
Collapse
Affiliation(s)
- Shufen Li
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Yulan Zhang
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Zhenqiong Guan
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Meidi Ye
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Huiling Li
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Miaomiao You
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhenxing Zhou
- University of Science and Technology of China, Hefei, Anhui, China
| | - Chongtao Zhang
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Fan Zhang
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Ben Lu
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Peng Zhou
- Guangzhou Laboratory, Guangzhou, Guangdong, China.
| | - Ke Peng
- State Key Laboratory of Virology, Center for Antiviral Research, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China.
| |
Collapse
|
137
|
Unilateral Facial Palsy After SARS-CoV-2 Booster Vaccination. J Clin Neuromuscul Dis 2023; 24:166-168. [PMID: 36809208 DOI: 10.1097/cnd.0000000000000416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
|
138
|
Ssentongo P, Yu N, Voleti N, Reddy S, Ingram D, Chinchilli VM, Paules CI. Optimal Duration of Systemic Corticosteroids in Coronavirus Disease 2019 Treatment: A Systematic Review and Meta-analysis. Open Forum Infect Dis 2023; 10:ofad105. [PMID: 36949880 PMCID: PMC10026544 DOI: 10.1093/ofid/ofad105] [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: 10/11/2022] [Accepted: 02/22/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Corticosteroids confer a survival benefit in individuals hospitalized with coronavirus disease 2019 (COVID-19) who require oxygen. This meta-analysis seeks to determine the duration of corticosteroids needed to optimize this mortality benefit. METHODS Electronic databases were searched to 9 March 2022, for studies reporting corticosteroid versus no corticosteroid treatment in hospitalized COVID-19 patients. We estimated the effect of corticosteroids on mortality by random-effects meta-analyses. Subgroup analyses and meta-analyses were conducted to assess the optimal duration of corticosteroid treatment while adjusting for the severity of disease, age, duration of symptoms, and proportion of control group given steroids. RESULTS We identified 27 eligible studies consisting of 13 404 hospitalized COVID-19 patients. Seven randomized controlled trials and 20 observational studies were included in the meta-analysis of mortality, which suggested a protective association with corticosteroid therapy (risk ratio [RR], 0.71 [95% confidence interval {CI}, .58-.87]). Pooled analysis of 18 studies showed the greatest survival benefit for a treatment duration up to 6 days (RR, 0.54 [95% CI, .39-.74]). Survival benefit was 0.65 (95% CI, .51-.83) up to 7 days, and no additional survival benefit was observed beyond 7 days of treatment (RR, 0.64 [95% CI, .44-.93]). The survival benefit was not confounded by severity of disease, age, duration of symptoms, or proportion of control group given steroids. CONCLUSIONS In this meta-analysis, optimal duration of corticosteroid treatment for hospitalized COVID-19 patients was up to 6 days, with no additional survival benefit with >7 days of treatment.
Collapse
Affiliation(s)
- Paddy Ssentongo
- Department of Medicine, Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Nyein Yu
- Division of Infectious Diseases, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Navya Voleti
- Department of Medicine, Penn State College of Medicine and Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Surya Reddy
- Department of Medicine, Osmania Medical College, Telangana, Hyderabad, India
| | - David Ingram
- Division of Infectious Diseases, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Vernon M Chinchilli
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Catharine I Paules
- Division of Infectious Diseases, Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania, USA
| |
Collapse
|
139
|
Piplani S, Singh P, Petrovsky N, Winkler DA. Identifying SARS-CoV-2 Drugs Binding to the Spike Fatty Acid Binding Pocket Using In Silico Docking and Molecular Dynamics. Int J Mol Sci 2023; 24:ijms24044192. [PMID: 36835602 PMCID: PMC9966092 DOI: 10.3390/ijms24044192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Drugs against novel targets are needed to treat COVID-19 patients, especially as SARS-CoV-2 is capable of rapid mutation. Structure-based de novo drug design and repurposing of drugs and natural products is a rational approach to discovering potentially effective therapies. These in silico simulations can quickly identify existing drugs with known safety profiles that can be repurposed for COVID-19 treatment. Here, we employ the newly identified spike protein free fatty acid binding pocket structure to identify repurposing candidates as potential SARS-CoV-2 therapies. Using a validated docking and molecular dynamics protocol effective at identifying repurposing candidates inhibiting other SARS-CoV-2 molecular targets, this study provides novel insights into the SARS-CoV-2 spike protein and its potential regulation by endogenous hormones and drugs. Some of the predicted repurposing candidates have already been demonstrated experimentally to inhibit SARS-CoV-2 activity, but most of the candidate drugs have yet to be tested for activity against the virus. We also elucidated a rationale for the effects of steroid and sex hormones and some vitamins on SARS-CoV-2 infection and COVID-19 recovery.
Collapse
Affiliation(s)
- Sakshi Piplani
- College of Medicine and Public Health, Flinders University, Bedford Park 5046, Australia
- Vaxine Pty Ltd., 11 Walkley Avenue, Warradale 5046, Australia
| | - Puneet Singh
- College of Medicine and Public Health, Flinders University, Bedford Park 5046, Australia
- Vaxine Pty Ltd., 11 Walkley Avenue, Warradale 5046, Australia
| | - Nikolai Petrovsky
- College of Medicine and Public Health, Flinders University, Bedford Park 5046, Australia
- Vaxine Pty Ltd., 11 Walkley Avenue, Warradale 5046, Australia
- Correspondence:
| | - David A. Winkler
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne 3086, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
140
|
Dahlberg J, Linder A, Mellhammar L. Use of healthcare before and after sepsis in Sweden: a case-control study. BMJ Open 2023; 13:e065967. [PMID: 36806063 PMCID: PMC9944643 DOI: 10.1136/bmjopen-2022-065967] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
OBJECTIVES The aim of this study was to compare readmissions and death between sepsis and non-sepsis hospitalisations the first year after discharge, and to investigate what diagnoses patients with sepsis present with at readmission. The aim was also to evaluate to what degree patients hospitalised for sepsis seek medical attention prior to hospitalisation. DESIGN Retrospective case-control study with data validated through clinical chart review. A disproportionate stratified sampling model was used to include a relatively larger number of sepsis hospitalisations. SETTING All eight public hospitals in region Scania, Sweden (1 January to 3 December 2019). PARTICIPANTS There were 447 patients hospitalised for sepsis (cases), and 541 hospitalised for other causes (control) identified through clinical chart review. OUTCOME MEASURES Cox regression was used to analyse readmission and death the year after discharge, and logistic regression was used to analyse healthcare the week prior to hospitalisation. Both analyses were made unadjusted, and adjusted for age, sex and comorbidities. RESULTS Out of patients who survived a sepsis hospitalisation, 48% were readmitted the year after discharge, compared with 39% for patients without sepsis (HR 1.50, 95% CI 1.03 to 2.19), p=0.04. The majority (52%) of readmissions occurred within 90 days and 75% within 180 days. The readmissions were most often caused by infection (32%), and 18% by cardiovascular disease. Finally, 34% of patients with sepsis had sought prehospital contact with a physician the week before hospitalisation, compared with 22% for patients without sepsis (OR 1.80, 95% CI 1.06 to 3.04), p=0.03. CONCLUSION Patients hospitalised for sepsis had a higher risk of readmission the year after discharge compared with patients without sepsis. The most common diagnoses at readmission were infection followed by cardiovascular disease. With better follow-up, some of these readmissions could potentially be prevented. Patients hospitalised for sepsis had sought prehospital contact the week prior to hospitalisation to a greater extent than patients without sepsis.
Collapse
Affiliation(s)
- Jacob Dahlberg
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Adam Linder
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Lisa Mellhammar
- Division of Infection Medicine, Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
141
|
Arrieta A, Galvis AE, Osborne S, Morphew T, Imfeld K, Enriquez C, Hoang J, Swearingen M, Nieves DJ, Ashouri N, Singh J, Nugent D. Use of COVID-19 Convalescent Plasma for Treatment of Symptomatic SARS-CoV-2 Infection at a Children's Hospital: A Contribution to a Still Inadequate Body of Evidence. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10020350. [PMID: 36832478 PMCID: PMC9955755 DOI: 10.3390/children10020350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/13/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
Data on COVID-19 convalescent plasma (CCP) safety and efficacy in children and young adults are limited. This single-center prospective, open-label trial evaluates CCP safety, neutralizing antibody kinetics, and outcomes in children and young adults with moderate/severe COVID-19 (April 2020-March 2021). A total of 46 subjects received CCP; 43 were included in the safety analysis (SAS); 7.0% < 2 years old, 2.3% 2-<6, 27.9% 6-<12, 39.5% 12-<19, and 23.3% > 19 years old; 28 were included in the antibody kinetic analysis (AbKS); 10.7% < 2 years old, 10.7% 6-<12, 53.8% 12-<19, and 25.0% > 19 years old. No adverse events occurred. The median COVID-19 severity score improved (5.0 pre-CCP to 1.0 by day 7; p < 0.001). A rapid increase in the median percentage of inhibition was observed in AbKS (22.5% (13.0%, 41.5%) pre-infusion to 52% (23.7%, 72%) 24 h post-infusion); a similar increase was observed in nine immune-competent subjects (28% (23%, 35%) to 63% (53%, 72%)). The inhibition percentage increased until day 7 and persisted at 21 and 90 days. CCP is well tolerated in children and young adults, providing rapid and robust increased antibodies. CCP should remain a therapeutic option for this population for whom vaccines are not fully available and given that the safety and efficacy of existing monoclonal antibodies and antiviral agents have not been established.
Collapse
Affiliation(s)
- Antonio Arrieta
- Pediatrics Infectious Diseases, CHOC Children’s Hospital, Orange, CA 92868, USA
- Department of Pediatrics, University of California Irvine School of Medicine, Irvine, CA 92697, USA
| | - Alvaro E. Galvis
- Pediatrics Infectious Diseases, CHOC Children’s Hospital, Orange, CA 92868, USA
- Department of Pediatrics, University of California Irvine School of Medicine, Irvine, CA 92697, USA
| | - Stephanie Osborne
- Research Administration, CHOC Children’s Hospital, Orange, CA 92868, USA
| | - Tricia Morphew
- Morphew Consulting, LLC, CHOC Research Institute, CHOC Children’s Hospital, Orange, CA 92868, USA
| | - Karen Imfeld
- Hematology Advanced Diagnostics Laboratory, CHOC Children’s Hospital, Orange, CA 92868, USA
| | - Claudia Enriquez
- Research Administration, CHOC Children’s Hospital, Orange, CA 92868, USA
| | - Janet Hoang
- Hematology Advanced Diagnostics Laboratory, CHOC Children’s Hospital, Orange, CA 92868, USA
| | - Marcia Swearingen
- Hematology Advanced Diagnostics Laboratory, CHOC Children’s Hospital, Orange, CA 92868, USA
| | - Delma J. Nieves
- Pediatrics Infectious Diseases, CHOC Children’s Hospital, Orange, CA 92868, USA
- Department of Pediatrics, University of California Irvine School of Medicine, Irvine, CA 92697, USA
- Correspondence: ; Tel.: +714-509-8403; Fax: +714-509-3303
| | - Negar Ashouri
- Pediatrics Infectious Diseases, CHOC Children’s Hospital, Orange, CA 92868, USA
- Department of Pediatrics, University of California Irvine School of Medicine, Irvine, CA 92697, USA
| | - Jasjit Singh
- Pediatrics Infectious Diseases, CHOC Children’s Hospital, Orange, CA 92868, USA
- Department of Pediatrics, University of California Irvine School of Medicine, Irvine, CA 92697, USA
| | - Diane Nugent
- Department of Pediatrics, University of California Irvine School of Medicine, Irvine, CA 92697, USA
- Pediatric Hematology, CHOC Children’s Hospital, Orange, CA 92868, USA
| |
Collapse
|
142
|
Yarnell CJ, Haas B. Transfusion Strategies and the Pragmatic Appeal of Bayesian Analysis for Trauma Research. JAMA Netw Open 2023; 6:e230426. [PMID: 36811864 DOI: 10.1001/jamanetworkopen.2023.0426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Affiliation(s)
- Christopher J Yarnell
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
- Medical-Surgical ICU, Toronto General Hospital, Toronto, Ontario, Canada
| | - Barbara Haas
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Ontario, Canada
- Sunnybrook Health Sciences Center, Toronto, Ontario, Canada
| |
Collapse
|
143
|
Wallis RS, O'Garra A, Sher A, Wack A. Host-directed immunotherapy of viral and bacterial infections: past, present and future. Nat Rev Immunol 2023; 23:121-133. [PMID: 35672482 PMCID: PMC9171745 DOI: 10.1038/s41577-022-00734-z] [Citation(s) in RCA: 113] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2022] [Indexed: 02/06/2023]
Abstract
The advent of COVID-19 and the persistent threat of infectious diseases such as tuberculosis, malaria, influenza and HIV/AIDS remind us of the marked impact that infections continue to have on public health. Some of the most effective protective measures are vaccines but these have been difficult to develop for some of these infectious diseases even after decades of research. The development of drugs and immunotherapies acting directly against the pathogen can be equally challenging, and such pathogen-directed therapeutics have the potential disadvantage of selecting for resistance. An alternative approach is provided by host-directed therapies, which interfere with host cellular processes required for pathogen survival or replication, or target the host immune response to infection (immunotherapies) to either augment immunity or ameliorate immunopathology. Here, we provide a historical perspective of host-directed immunotherapeutic interventions for viral and bacterial infections and then focus on SARS-CoV-2 and Mycobacterium tuberculosis, two major human pathogens of the current era, to indicate the key lessons learned and discuss candidate immunotherapeutic approaches, with a focus on drugs currently in clinical trials.
Collapse
Affiliation(s)
- Robert S Wallis
- The Aurum Institute, Johannesburg, South Africa.
- Vanderbilt University, Nashville, TN, USA.
- Rutgers University, Newark, NJ, USA.
- Case Western Reserve University, Cleveland, OH, USA.
| | - Anne O'Garra
- Immunoregulation and Infection Laboratory, The Francis Crick Institute, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
144
|
Abstract
Coronavirus disease 2019 (COVID-19) pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in significant mortality in pandemic proportions. Inflammation in response to the infection contributes to the pathogenesis of pneumonia. This review will discuss prior studies on the use of glucocorticoids to treat respiratory infections, the rationale for the use glucocorticoids in COVID-19, and review of existing data. We will also highlight outstanding research questions for future studies.
Collapse
Affiliation(s)
- Francesco Amati
- Respiratory Unit, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Antonio Tonutti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - John Huston
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Critical Care and Sleep Medicine, Center for Pulmonary Infection Research and Treatment, Yale School of Medicine, New Haven, Connecticut
| | - Charles S. Dela Cruz
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Critical Care and Sleep Medicine, Center for Pulmonary Infection Research and Treatment, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
145
|
Souan L, Al-Khairy Z, Battah A, Sughayer MA. Non-Dexamethasone Corticosteroid Therapy's Effect on COVID-19 Prognosis in Cancer Patients: A Retrospective Study. Vaccines (Basel) 2023; 11:vaccines11020290. [PMID: 36851168 PMCID: PMC9964883 DOI: 10.3390/vaccines11020290] [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: 12/19/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Anti-inflammatory corticosteroids are used in cancer treatment and COVID-19 infections. Data on the impact of non-dexamethasone corticosteroids on COVID-19 infection severity in cancer patients are minimal. This study investigates if corticosteroid treatment affects the disease severity in adult cancer patients. METHODS A total of 116 COVID-19-infected cancer patients on hydrocortisone (H) or prednisone (P) were compared to 343 untreated patients. The study included patients who received corticosteroids before (B), after (A), or both before and after (B and A) COVID-19 infections. Ventilation support, hospitalization and mortality were investigated. RESULTS Our data showed that a significantly greater number of patients taking H or P required ventilation support and hospitalization and that mortality rates were higher than the control group. Patients who received H or P after COVID-19 infection had a significantly worse prognosis than the other sub-groups and the control group. CONCLUSION Corticosteroids impacted cancer patients' COVID-19 prognosis. Despite the limited sample size, H- and P-treated patients' corticosteroids performed worse than the control, especially if treatments were received after COVID-19 infection. Hence, when a cancer patient already on H or P treatment is diagnosed with COVID-19, we recommend switching to a steroid treatment as suggested by international guidelines.
Collapse
Affiliation(s)
- Lina Souan
- Department of Pathology & Laboratory Medicine, King Hussein Cancer Center, Amman 11941, Jordan
- Correspondence: (L.S.); (M.A.S.)
| | - Zienab Al-Khairy
- Department of Pathology & Laboratory Medicine, King Hussein Cancer Center, Amman 11941, Jordan
| | - Abdelkader Battah
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Maher A. Sughayer
- Department of Pathology & Laboratory Medicine, King Hussein Cancer Center, Amman 11941, Jordan
- Correspondence: (L.S.); (M.A.S.)
| |
Collapse
|
146
|
Yuan Y, Jiao B, Qu L, Yang D, Liu R. The development of COVID-19 treatment. Front Immunol 2023; 14:1125246. [PMID: 36776881 PMCID: PMC9909293 DOI: 10.3389/fimmu.2023.1125246] [Citation(s) in RCA: 119] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/03/2023] [Indexed: 01/27/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused a pandemic named coronavirus disease 2019 (COVID-19) that has become the greatest worldwide public health threat of this century. Recent studies have unraveled numerous mysteries of SARS-CoV-2 pathogenesis and thus largely improved the studies of COVID-19 vaccines and therapeutic strategies. However, important questions remain regarding its therapy. In this review, the recent research advances on COVID-19 mechanism are quickly summarized. We mainly discuss current therapy strategies for COVID-19, with an emphasis on antiviral agents, neutralizing antibody therapies, Janus kinase inhibitors, and steroids. When necessary, specific mechanisms and the history of therapy are present, and representative strategies are described in detail. Finally, we discuss key outstanding questions regarding future directions of the development of COVID-19 treatment.
Collapse
Affiliation(s)
- Yongliang Yuan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Baihai Jiao
- Division of Nephrology, Department of Medicine, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Lili Qu
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Duomeng Yang
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, CT, United States,*Correspondence: Ruijuan Liu, ; Duomeng Yang,
| | - Ruijuan Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,*Correspondence: Ruijuan Liu, ; Duomeng Yang,
| |
Collapse
|
147
|
Dallavalasa S, Tulimilli SV, Prakash J, Ramachandra R, Madhunapantula SV, Veeranna RP. COVID-19: Diabetes Perspective-Pathophysiology and Management. Pathogens 2023; 12:pathogens12020184. [PMID: 36839456 PMCID: PMC9967788 DOI: 10.3390/pathogens12020184] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/05/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
Recent evidence relating to the impact of COVID-19 on people with diabetes is limited but continues to emerge. COVID-19 pneumonia is a newly identified illness spreading rapidly throughout the world and causes many disabilities and fatal deaths. Over the ensuing 2 years, the indirect effects of the pandemic on healthcare delivery have become prominent, along with the lingering effects of the virus on those directly infected. Diabetes is a commonly identified risk factor that contributes not only to the severity and mortality of COVID-19 patients, but also to the associated complications, including acute respiratory distress syndrome (ARDS) and multi-organ failure. Diabetic patients are highly affected due to increased viral entry into the cells and decreased immunity. Several hypotheses to explain the increased incidence and severity of COVID-19 infection in people with diabetes have been proposed and explained in detail recently. On the other hand, 20-50% of COVID-19 patients reported new-onset hyperglycemia without diabetes and new-onset diabetes, suggesting the two-way interactions between COVID-19 and diabetes. A systematic review is required to confirm diabetes as a complication in those patients diagnosed with COVID-19. Diabetes and diabetes-related complications in COVID-19 patients are primarily due to the acute illness caused during the SARS-CoV-2 infection followed by the release of glucocorticoids, catecholamines, and pro-inflammatory cytokines, which have been shown to drive hyperglycemia positively. This review provides brief insights into the potential mechanisms linking COVID-19 and diabetes, and presents clinical management recommendations for better handling of the disease.
Collapse
Affiliation(s)
- Siva Dallavalasa
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Centre), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, India
| | - SubbaRao V. Tulimilli
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Centre), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, India
| | - Janhavi Prakash
- Department of Biochemistry, Council of Scientific and Industrial Research (CSIR)-Central Food Technological Research Institute (CFTRI), Mysuru 570020, India
| | - Ramya Ramachandra
- Department of Biochemistry, Council of Scientific and Industrial Research (CSIR)-Central Food Technological Research Institute (CFTRI), Mysuru 570020, India
| | - SubbaRao V. Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR) Laboratory (DST-FIST Supported Centre), Department of Biochemistry (DST-FIST Supported Department), JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, India
- Leader, Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, India
| | - Ravindra P. Veeranna
- Department of Biochemistry, Council of Scientific and Industrial Research (CSIR)-Central Food Technological Research Institute (CFTRI), Mysuru 570020, India
- Correspondence:
| |
Collapse
|
148
|
Asif S, Frithiof R, Larsson A, Franzén S, Anderberg SB, Kristensen B, Hultström M, Lipcsey M. Immuno-Modulatory Effects of Dexamethasone in Severe COVID-19-A Swedish Cohort Study. Biomedicines 2023; 11:biomedicines11010164. [PMID: 36672672 PMCID: PMC9855905 DOI: 10.3390/biomedicines11010164] [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: 11/26/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
Dexamethasone (Dex) has been shown to decrease mortality in severe coronavirus disease 2019 (COVID-19), but the mechanism is not fully elucidated. We aimed to investigate the physiological and immunological effects associated with Dex administration in patients admitted to intensive care with severe COVID-19. A total of 216 adult COVID-19 patients were included-102 (47%) received Dex, 6 mg/day for 10 days, and 114 (53%) did not. Standard laboratory parameters, plasma expression of cytokines, endothelial markers, immunoglobulin (Ig) IgA, IgM, and IgG against SARS-CoV-2 were analyzed post-admission to intensive care. Patients treated with Dex had higher blood glucose but lower blood lactate, plasma cortisol, IgA, IgM, IgG, D-dimer, cytokines, syndecan-1, and E-selectin and received less organ support than those who did not receive Dex (Without-Dex). There was an association between Dex treatment and IL-17A, macrophage inflammatory protein 1 alpha, syndecan-1 as well as E-selectin in predicting 30-day mortality. Among a subgroup of patients who received Dex early, within 14 days of COVID-19 debut, the adjusted mortality risk was 0.4 (95% CI 0.2-0.8), i.e., 40% compared with Without-Dex. Dex administration in a cohort of critically ill COVID-19 patients resulted in altered immunological and physiologic responses, some of which were associated with mortality.
Collapse
Affiliation(s)
- Sana Asif
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden
- Correspondence:
| | - Robert Frithiof
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden
| | - Anders Larsson
- Department of Medical Sciences, Uppsala University, 751 85 Uppsala, Sweden
| | - Stephanie Franzén
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden
| | - Sara Bülow Anderberg
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden
| | | | - Michael Hultström
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden
- Unit for Integrative Physiology, Department of Medical Cell Biology, Uppsala University, 751 85 Uppsala, Sweden
| | - Miklos Lipcsey
- Anaesthesiology and Intensive Care Medicine, Department of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden
- Hedenstierna Laboratory, Department of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden
| |
Collapse
|
149
|
Naorungroj T, Viarasilpa T, Tongyoo S, Detkaew A, Pinpak T, Wimolwattanaphan R, Ratanarat R, Promsin P, Thamrongpiroj P, Phumpichet A, Permpikul C. Characteristics, outcomes, and risk factors for in-hospital mortality of COVID-19 patients: A retrospective study in Thailand. Front Med (Lausanne) 2023; 9:1061955. [PMID: 36687414 PMCID: PMC9846200 DOI: 10.3389/fmed.2022.1061955] [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: 10/05/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction Data on the characteristics and outcomes of patients hospitalized for Coronavirus Disease 2019 (COVID-19) in Thailand are limited. Objective To determine characteristics and outcomes and identify risk factors for hospital mortality for hospitalized patients with COVID-19. Methods We retrospectively reviewed the medical records of patients who had COVID-19 infection and were admitted to the cohort ward or ICUs at Siriraj Hospital between January 2020 and December 2021. Results Of the 2,430 patients included in this study, 229 (9.4%) died; the mean age was 54 years, 40% were men, 81% had at least one comorbidity, and 13% required intensive care unit (ICU). Favipiravir (86%) was the main antiviral treatment. Corticosteroids and rescue anti-inflammatory therapy were used in 74 and 6%, respectively. Admission to the ICU was the only factor associated with reduced mortality [odds ratio (OR) 0.01, 95% confidence interval (CI) 0.01-0.05, P < 0.001], whereas older age (OR 14.3, 95%CI 5.76-35.54, P < 0.001), high flow nasal cannula (HFNC; OR 9.2, 95% CI 3.9-21.6, P < 0.001), mechanical ventilation (OR 269.39, 95%CI 3.6-2173.63, P < 0.001), septic shock (OR 7.79, 95%CI, 2.01-30.18, P = 0.003), and hydrocortisone treatment (OR 27.01, 95%CI 5.29-138.31, P < 0.001) were factors associated with in-hospital mortality. Conclusion The overall mortality of hospitalized patients with COVID-19 was 9%. The only factor associated with reduced mortality was admission to the ICU. Therefore, appropriate selection of patients for admission to the ICU, strategies to limit disease progression and prevent intubation, and early detection and prompt treatment of nosocomial infection can improve survival in these patients.
Collapse
|
150
|
Higgins AM, Berry LR, Lorenzi E, Murthy S, McQuilten Z, Mouncey PR, Al-Beidh F, Annane D, Arabi YM, Beane A, van Bentum-Puijk W, Bhimani Z, Bonten MJM, Bradbury CA, Brunkhorst FM, Burrell A, Buzgau A, Buxton M, Charles WN, Cove M, Detry MA, Estcourt LJ, Fagbodun EO, Fitzgerald M, Girard TD, Goligher EC, Goossens H, Haniffa R, Hills T, Horvat CM, Huang DT, Ichihara N, Lamontagne F, Marshall JC, McAuley DF, McGlothlin A, McGuinness SP, McVerry BJ, Neal MD, Nichol AD, Parke RL, Parker JC, Parry-Billings K, Peters SEC, Reyes LF, Rowan KM, Saito H, Santos MS, Saunders CT, Serpa-Neto A, Seymour CW, Shankar-Hari M, Stronach LM, Turgeon AF, Turner AM, van de Veerdonk FL, Zarychanski R, Green C, Lewis RJ, Angus DC, McArthur CJ, Berry S, Derde LPG, Gordon AC, Webb SA, Lawler PR. Long-term (180-Day) Outcomes in Critically Ill Patients With COVID-19 in the REMAP-CAP Randomized Clinical Trial. JAMA 2023; 329:39-51. [PMID: 36525245 PMCID: PMC9857594 DOI: 10.1001/jama.2022.23257] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022]
Abstract
Importance The longer-term effects of therapies for the treatment of critically ill patients with COVID-19 are unknown. Objective To determine the effect of multiple interventions for critically ill adults with COVID-19 on longer-term outcomes. Design, Setting, and Participants Prespecified secondary analysis of an ongoing adaptive platform trial (REMAP-CAP) testing interventions within multiple therapeutic domains in which 4869 critically ill adult patients with COVID-19 were enrolled between March 9, 2020, and June 22, 2021, from 197 sites in 14 countries. The final 180-day follow-up was completed on March 2, 2022. Interventions Patients were randomized to receive 1 or more interventions within 6 treatment domains: immune modulators (n = 2274), convalescent plasma (n = 2011), antiplatelet therapy (n = 1557), anticoagulation (n = 1033), antivirals (n = 726), and corticosteroids (n = 401). Main Outcomes and Measures The main outcome was survival through day 180, analyzed using a bayesian piecewise exponential model. A hazard ratio (HR) less than 1 represented improved survival (superiority), while an HR greater than 1 represented worsened survival (harm); futility was represented by a relative improvement less than 20% in outcome, shown by an HR greater than 0.83. Results Among 4869 randomized patients (mean age, 59.3 years; 1537 [32.1%] women), 4107 (84.3%) had known vital status and 2590 (63.1%) were alive at day 180. IL-6 receptor antagonists had a greater than 99.9% probability of improving 6-month survival (adjusted HR, 0.74 [95% credible interval {CrI}, 0.61-0.90]) and antiplatelet agents had a 95% probability of improving 6-month survival (adjusted HR, 0.85 [95% CrI, 0.71-1.03]) compared with the control, while the probability of trial-defined statistical futility (HR >0.83) was high for therapeutic anticoagulation (99.9%; HR, 1.13 [95% CrI, 0.93-1.42]), convalescent plasma (99.2%; HR, 0.99 [95% CrI, 0.86-1.14]), and lopinavir-ritonavir (96.6%; HR, 1.06 [95% CrI, 0.82-1.38]) and the probabilities of harm from hydroxychloroquine (96.9%; HR, 1.51 [95% CrI, 0.98-2.29]) and the combination of lopinavir-ritonavir and hydroxychloroquine (96.8%; HR, 1.61 [95% CrI, 0.97-2.67]) were high. The corticosteroid domain was stopped early prior to reaching a predefined statistical trigger; there was a 57.1% to 61.6% probability of improving 6-month survival across varying hydrocortisone dosing strategies. Conclusions and Relevance Among critically ill patients with COVID-19 randomized to receive 1 or more therapeutic interventions, treatment with an IL-6 receptor antagonist had a greater than 99.9% probability of improved 180-day mortality compared with patients randomized to the control, and treatment with an antiplatelet had a 95.0% probability of improved 180-day mortality compared with patients randomized to the control. Overall, when considered with previously reported short-term results, the findings indicate that initial in-hospital treatment effects were consistent for most therapies through 6 months.
Collapse
Affiliation(s)
| | | | | | - Srinivas Murthy
- University of British Columbia, Vancouver, British Columbia, Canada
| | - Zoe McQuilten
- Monash University, Melbourne, Victoria, Australia
- Monash Health, Melbourne, Victoria, Australia
| | - Paul R Mouncey
- Intensive Care National Audit & Research Centre (ICNARC), London, United Kingdom
| | | | - Djillali Annane
- Hospital Raymond Poincaré (Assistance Publique Hôpitaux de Paris), Garches, France
- Université Versailles SQY - Université Paris Saclay, Montigny-le-Bretonneux, France
| | - Yaseen M Arabi
- King Saud bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center, Riyadh, Kingdom of Saudi Arabia
| | - Abi Beane
- University of Oxford, Oxford, United Kingdom
| | | | - Zahra Bhimani
- St Michael's Hospital Unity Health, Toronto, Ontario, Canada
| | | | | | | | | | | | | | - Walton N Charles
- Intensive Care National Audit & Research Centre (ICNARC), London, United Kingdom
| | - Matthew Cove
- Yong Loo Lin Scholle of Medicine, National University Singapore, Singapore
| | | | | | | | | | | | - Ewan C Goligher
- Peter Munk Cardiac Centre at University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | | | - Rashan Haniffa
- University of Oxford, Bangkok, Thailand
- National Intensive Care Surveillance (NICST), Colombo, Sri Lanka
| | - Thomas Hills
- Medical Research Institute of New Zealand (MRINZ), Wellington, New Zealand
| | | | | | | | | | - John C Marshall
- St Michael's Hospital Unity Health, Toronto, Ontario, Canada
| | - Daniel F McAuley
- Queen's University Belfast, Belfast, Northern Ireland
- Royal Victoria Hospital, Belfast, Northern Ireland
| | | | - Shay P McGuinness
- Monash University, Melbourne, Victoria, Australia
- Auckland City Hospital, Auckland, New Zealand
| | | | | | - Alistair D Nichol
- Monash University, Melbourne, Victoria, Australia
- University College Dublin, Dublin, Ireland
| | - Rachael L Parke
- Auckland City Hospital, Auckland, New Zealand
- University of Auckland, Auckland, New Zealand
| | | | - Karen Parry-Billings
- Intensive Care National Audit & Research Centre (ICNARC), London, United Kingdom
| | - Sam E C Peters
- Intensive Care National Audit & Research Centre (ICNARC), London, United Kingdom
| | - Luis F Reyes
- Universidad de La Sabana, Chia, Colombia
- Clinica Universidad de La Sabana, Chia, Colombia
| | - Kathryn M Rowan
- Intensive Care National Audit & Research Centre (ICNARC), London, United Kingdom
| | - Hiroki Saito
- St Marianna University School of Medicine, Yokohama City Seibu Hospital, Yokohama, Japan
| | | | | | - Ary Serpa-Neto
- Monash University, Melbourne, Victoria, Australia
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | | | - Manu Shankar-Hari
- King's College London, London, United Kingdom
- Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | | | - Alexis F Turgeon
- Université Laval, Québec City, Québec, Canada
- CHU de Québec-Université Laval Research Center, Québec City, Canada
| | - Anne M Turner
- Medical Research Institute of New Zealand (MRINZ), Wellington, New Zealand
| | | | | | | | - Roger J Lewis
- Berry Consultants, Austin, Texas
- Harbor-UCLA Medical Center, Torrance, California
| | | | | | | | | | - Anthony C Gordon
- Imperial College London, London, United Kingdom
- Imperial College Healthcare NHS Trust, St Mary's Hospital, London, United Kingdom
| | - Steve A Webb
- Monash University, Melbourne, Victoria, Australia
- St John of God Hospital, Subiaco, Australia
| | - Patrick R Lawler
- Peter Munk Cardiac Centre at University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|