101
|
Cron RQ. IL-1 Family Blockade in Cytokine Storm Syndromes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:553-563. [PMID: 39117838 DOI: 10.1007/978-3-031-59815-9_36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Interleukin-1 is a prototypic proinflammatory cytokine that is elevated in cytokine storm syndromes (CSSs), such as secondary hemophagocytic lymphohistiocytosis (sHLH) and macrophage activation syndrome (MAS). IL-1 has many pleotropic and redundant roles in both innate and adaptive immune responses. Blockade of IL-1 with recombinant human interleukin-1 receptor antagonist has shown efficacy in treating CSS. Recently, an IL-1 family member, IL-18, has been demonstrated to be contributory to CSS in autoinflammatory conditions, such as in inflammasomopathies (e.g., NLRC4 mutations). Anecdotally, recombinant IL-18 binding protein can be of benefit in treating IL-18-driven CSS. Lastly, another IL-1 family member, IL-33, has been postulated to contribute to CSS in an animal model of disease. Targeting of IL-1 and related cytokines holds promise in treating a variety of CSS.
Collapse
|
102
|
Keenan C, Albeituni S, Nichols KE, Hines M. JAK Inhibitors in Cytokine Storm Syndromes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:583-600. [PMID: 39117841 DOI: 10.1007/978-3-031-59815-9_39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Cytokine storm syndromes (CSSs) comprise a group of severe and often fatal hyperinflammatory conditions driven by the overproduction of pro-inflammatory cytokines by activated cells of the immune system. Many of the CSS-associated cytokines mediate their downstream effects by signaling through the Janus kinases (JAKs) and signal transducers and activators of transcription (STATs). In addition, several of these cytokines are produced downstream of JAK/STAT pathway activation. Therefore, targeting JAK/STAT signaling using small molecule JAK inhibitors has become an increasingly appealing therapeutic option to dampen hyperinflammation in patients with CSSs. Application of JAK inhibitors in preclinical CSS models has shown improvements in multiple sequelae of hyperinflammation, and there is growing clinical evidence supporting the efficacy of JAK inhibition in patients with these conditions. Although generally well tolerated, JAK inhibitor use is not without potential for toxicity, especially in settings like CSSs where end-organ dysfunction is common. More prospective clinical trials incorporating JAK inhibitors, alone or in combination with other immunomodulatory therapies, are necessary to determine the optimal dosing, schedule, efficacy, and tolerability of these agents for patients experiencing CSSs.
Collapse
Affiliation(s)
- Camille Keenan
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sabrin Albeituni
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kim E Nichols
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Melissa Hines
- Department of Pediatric Medicine, Division of Critical Care Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
103
|
Conroy GM, Bauer SR, Pallotta AM, Duggal A, Wang L, Sacha GL. Baricitinib versus tocilizumab in critically ill COVID-19 patients: A retrospective cohort study. Pharmacotherapy 2024; 44:28-38. [PMID: 37593883 PMCID: PMC10961678 DOI: 10.1002/phar.2867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/06/2023] [Accepted: 07/14/2023] [Indexed: 08/19/2023]
Abstract
OBJECTIVES The immunomodulators tocilizumab and baricitinib improve outcomes in severely ill patients with coronavirus disease 2019 (COVID-19); however, comparative analyses of clinical outcomes related to these agents are lacking. A tocilizumab national shortage shifted treatment to baricitinib in critically ill patients, allowing for an outcome comparison in a similar population. The purpose of this study is to compare clinical outcomes in critically ill COVID-19 patients who received tocilizumab and those who received baricitinib. DESIGN Retrospective, observational cohort study using generalized estimating equation models, accounting for clustering by hospital and known confounders, to estimate the proportional odds of the ordinal World Health Organization Clinical Progression Scale (WHO-CPS) score at day 14, the primary outcome. Secondary outcomes included WHO-CPS score at day 7. SETTING Multiple hospitals within the Cleveland Clinic Health System. PATIENTS Adult patients admitted for COVID-19 between January 2021 and November 2021. INTERVENTIONS Receipt of tocilizumab, before its shortage, or baricitinib, during shortage. MEASUREMENTS AND MAIN RESULTS In total, 507 patients were included; 217 received tocilizumab and 290 received baricitinib. Over 96% of patients required ICU admission and 98% received concomitant dexamethasone. Tocilizumab recipients had higher (worse) baseline WHO-CPS scores. After adjustment, tocilizumab use was associated with higher odds of a worse day 14 WHO-CPS score compared with baricitinib (adjusted odds ratio [OR] 1.65 [95% confidence interval (CI) 1.10-2.48]). Similarly, after adjustment, tocilizumab use was associated with higher odds of a worse day 7 WHO-CPS score (adjusted OR 1.65 [95% CI 1.22-2.24]). CONCLUSIONS Baricitinib use was associated with better WHO-CPS scores at day 14 and day 7 compared with tocilizumab in a cohort of critically ill patients with COVID-19. The odds of having a one unit increase in WHO-CPS score at day 14 was 71% higher with tocilizumab than baricitinib. No difference in mortality or adverse effects was noted.
Collapse
Affiliation(s)
| | - Seth R. Bauer
- Department of Pharmacy, Cleveland Clinic, Cleveland, Ohio
| | | | - Abhijit Duggal
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio
| | - Lu Wang
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio
| | | |
Collapse
|
104
|
Hodel K, Fonseca A, Barbosa I, Medina C, Alves B, Maciel C, Nascimento D, Oliveira-Junior G, Pedreira L, de Souza M, Godoy AL. Obesity and its Relationship with Covid-19: A Review of the Main Pharmaceutical Aspects. Curr Pharm Biotechnol 2024; 25:1651-1663. [PMID: 38258769 DOI: 10.2174/0113892010264503231108070917] [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: 06/15/2023] [Revised: 09/28/2023] [Accepted: 10/04/2023] [Indexed: 01/24/2024]
Abstract
Important physiological changes are observed in patients with obesity, such as intestinal permeability, gastric emptying, cardiac output, and hepatic and renal function. These differences can determine variations in the pharmacokinetics of different drugs and can generate different concentrations at the site of action, which can lead to sub therapeutic or toxic concentrations. Understanding the physiological and immunological processes that lead to the clinical manifestations of COVID-19 is essential to correlate obesity as a risk factor for increasing the prevalence, severity, and lethality of the disease. Several drugs have been suggested to control COVID- 19 like Lopinavir, Ritonavir, Ribavirin, Sofosbuvir, Remdesivir, Oseltamivir, Oseltamivir phosphate, Oseltamivir carboxylate, Hydroxychloroquine, Chloroquine, Azithromycin, Teicoplanin, Tocilizumab, Anakinra, Methylprednisolone, Prednisolone, Ciclesonide and Ivermectin. Similarly, these differences between healthy people and obese people can be correlated to mechanical factors, such as insufficient doses of the vaccine for high body mass, impairing the absorption and distribution of the vaccine that will be lower than desired or can be linked to the inflammatory state in obese patients, which can influence the humoral immune response. Additionally, different aspects make the obese population more prone to persistent symptoms of the disease (long COVID), which makes understanding these mechanisms fundamental to addressing the implications of the disease. Thus, this review provides an overview of the relationship between COVID-19 and obesity, considering aspects related to pharmacokinetics, immunosuppression, immunization, and possible implications of long COVID in these individuals.
Collapse
Affiliation(s)
- Katharine Hodel
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Ananda Fonseca
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Islania Barbosa
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Caio Medina
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Brenda Alves
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Carine Maciel
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Daniel Nascimento
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Gessualdo Oliveira-Junior
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Lorena Pedreira
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Monielly de Souza
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Ana Leonor Godoy
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| |
Collapse
|
105
|
Jaffal K, Davido B. [Early post-exposure and curative therapeutic strategies against COVID-19]. Rev Mal Respir 2024; 41:51-58. [PMID: 37993363 DOI: 10.1016/j.rmr.2023.10.007] [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: 04/22/2023] [Accepted: 10/25/2023] [Indexed: 11/24/2023]
Abstract
INTRODUCTION There now exist preventive and curative treatments available for both early and advanced stages of COVID-19 management. CURRENT KNOWLEDGE Antibiotics have no place in the initial therapeutic management of Sars-Cov-2 pneumonia. On the other hand, corticosteroids are recommended for patients requiring oxygen therapy≥2L/min. According to the latest recommendations, nirmatrelvir/ritonavir is indicated as an early treatment for adults not requiring oxygen therapy but at high risk of developing a severe form of COVID-19. In case of contraindication, remdesivir is an alternative therapy. PERSPECTIVES Although there is no indication for convalescent plasma outside of clinical trials, it seems promising for immunocompromised patients, particularly those with B lymphopenia. It is noteworthy that currently, with the predominance of the Omicron XBB.1.5 variant, monoclonal antibodies are no longer recommended as therapy except for sotrovimab, which still has partial efficacy and could be considered after expert opinion as salvage therapy in a previously well-established program. CONCLUSION Despite the evolution of variants, antivirals still appear to have activity and remain the first-line treatment for patients, in addition to vaccination.
Collapse
Affiliation(s)
- K Jaffal
- Service de maladies infectieuses, hôpital universitaire Raymond-Poincaré, AP-HP, 104, boulevard Raymond-Poincaré, 92380 Garches, France
| | - B Davido
- Service de maladies infectieuses, hôpital universitaire Raymond-Poincaré, AP-HP, 104, boulevard Raymond-Poincaré, 92380 Garches, France; UMR 1173, université Paris Saclay, Versailles, France.
| |
Collapse
|
106
|
van de Veerdonk FL. COVID-19 Pneumonia and Cytokine Storm Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:307-319. [PMID: 39117824 DOI: 10.1007/978-3-031-59815-9_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Virus-associated cytokine storm syndrome (CSS) has been recognized for a long time and the classic viruses associated are the herpes viruses EBV, CMV, and HHV-8 as described in chapters IVa,b. In addition, pandemic viruses such as influenza, SARS, and MERS can result in severe CSS that might ultimately lead to severe acute respiratory distress syndrome (ARDS) and death [1-3]. A new pandemic caused by SARS-CoV-2 that started in 2019 has defined another chapter in the virus-associated CSS. The clinical spectrum of SARS-CoV-2 infection has many faces. In most people, it will be asymptomatic, but it can also result in severe COVID-19 pneumonia, ARDS, and multiorgan failure depending on age, comorbidities, and immune status [4]. In addition, this pandemic has known many different stages and developed in a unique way in the first 2 years. It started in a setting where there was no immunity to the virus and after a year, highly effective vaccines were introduced and herd immunity built up over time. However, vaccine effectiveness was waning over time depending on multiple factors, and novel variant strains of the virus circulated across different areas in the world. Antiviral therapy was developed and introduced, and treatment changed from giving no immunomodulatory treatment, followed by the introduction of corticosteroids [5], and later the addition of more targeted strategies such as JAK inhibitors [6] and blocking IL-6 signaling [7]. Therefore, the scientific literature published on COVID-19 must be seen in the context of a highly dynamic and rapidly changing pandemic, making it difficult to compare results from early studies to more recent reports even within 2 years. Still, a lot has been learned over a very short period. It has become apparent that severe COVID-19 is predominantly a disease of immune dysregulation with components that can be defined as CSS. It has unique features and overlapping characteristics with other CSSs, and immunological treatment addressing the CSS has been extensively explored, which will be described here.
Collapse
|
107
|
Sundén-Cullberg J, Chen P, Häbel H, Skorup P, Janols H, Rasmuson J, Niward K, Östholm Balkhed Å, Chatzidionysiou K, Asgeirsson H, Blennow O, Parke Å, Svensson AK, Muvva JR, Ljunggren HG, Karolinska KI/K COVID-19 Treatment Working Group, Horne AC, Ådén U, Henter JI, Sönnerborg A, Vesterbacka J, Nowak P, Lampa J. Anakinra or tocilizumab in patients admitted to hospital with severe covid-19 at high risk of deterioration (IMMCoVA): A randomized, controlled, open-label trial. PLoS One 2023; 18:e0295838. [PMID: 38157348 PMCID: PMC10756513 DOI: 10.1371/journal.pone.0295838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Anakinra and tocilizumab are used for severe Covid-19, but only one previous randomized controlled trial (RCT) has studied both. We performed a multi-center RCT comparing anakinra or tocilizumab versus usual care (UC) for adults at high risk of deterioration. METHODS The study was conducted June 2020 to March 2021. Eligibility required ≥ 5 liters/minute of Oxygen to maintain peripheral oxygen saturation at ≥ 93%, CRP > 70 mg/L, ferritin > 500 μg/L and at least two points where one point was awarded for lymphocytes < 1x 109/L; D-dimer ≥ 0.5 mg/L and; lactate dehydrogenase ≥ 8 microkatal/L. Patients were randomly assigned 1:1:1 to receive either a single dose of tocilizumab (8 mg/kg) or anakinra 100 mg IV QID for seven days or UC alone. The primary outcome was time to recovery. RESULTS Recruitment was ended prematurely when tocilizumab became part of usual care. Out of a planned 195 patients, 77 had been randomized, 27 to UC, 28 to anakinra and 22 to tocilizumab. Median time to recovery was 15, 15 and 11 days. Rate ratio for recovery for UC vs anakinra was 0.91, 0.47 to 1.78, 95% [CI], p = 0.8 and for UC vs tocilizumab 1.13, 0.55 to 2.30; p = 0.7. There were non-significant trends favoring tocilizumab (and to limited degree anakinra) vs UC for some secondary outcomes. Safety profiles did not differ significantly. CONCLUSION Premature closure of trial precludes firm conclusions. Anakinra or tocilizumab did not significantly shorten time to clinical recovery compared to usual care. (IMMCoVA, NCT04412291, EudraCT: 2020-00174824).
Collapse
Affiliation(s)
- Jonas Sundén-Cullberg
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Puran Chen
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Henrike Häbel
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paul Skorup
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Helena Janols
- Department of Medical Sciences, Section of Infectious Diseases, Uppsala University, Uppsala, Sweden
| | - Johan Rasmuson
- Infection and Immunology, Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Katarina Niward
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Åse Östholm Balkhed
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Katerina Chatzidionysiou
- Rheumatology Division, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Hilmir Asgeirsson
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Ola Blennow
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Åsa Parke
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Anna-Karin Svensson
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Jagadeeswara Rao Muvva
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Hans-Gustav Ljunggren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | | | - Anna-Carin Horne
- Theme of Children’s and Women’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Ulrika Ådén
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Jan-Inge Henter
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Anders Sönnerborg
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jan Vesterbacka
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Piotr Nowak
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Jon Lampa
- Rheumatology Division, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
108
|
Arman BY, Brun J, Hill ML, Zitzmann N, von Delft A. An Update on SARS-CoV-2 Clinical Trial Results-What We Can Learn for the Next Pandemic. Int J Mol Sci 2023; 25:354. [PMID: 38203525 PMCID: PMC10779148 DOI: 10.3390/ijms25010354] [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: 11/28/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has claimed over 7 million lives worldwide, providing a stark reminder of the importance of pandemic preparedness. Due to the lack of approved antiviral drugs effective against coronaviruses at the start of the pandemic, the world largely relied on repurposed efforts. Here, we summarise results from randomised controlled trials to date, as well as selected in vitro data of directly acting antivirals, host-targeting antivirals, and immunomodulatory drugs. Overall, repurposing efforts evaluating directly acting antivirals targeting other viral families were largely unsuccessful, whereas several immunomodulatory drugs led to clinical improvement in hospitalised patients with severe disease. In addition, accelerated drug discovery efforts during the pandemic progressed to multiple novel directly acting antivirals with clinical efficacy, including small molecule inhibitors and monoclonal antibodies. We argue that large-scale investment is required to prepare for future pandemics; both to develop an arsenal of broad-spectrum antivirals beyond coronaviruses and build worldwide clinical trial networks that can be rapidly utilised.
Collapse
Affiliation(s)
- Benediktus Yohan Arman
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Juliane Brun
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Michelle L. Hill
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK;
| | - Nicole Zitzmann
- Antiviral Drug Discovery Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK; (J.B.); (N.Z.)
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
| | - Annette von Delft
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford OX1 3QU, UK
- Centre for Medicine Discovery, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
109
|
Edwards KM. The Therapy of SARS-CoV-2 Infection in Children. J Clin Med 2023; 13:120. [PMID: 38202127 PMCID: PMC10779459 DOI: 10.3390/jcm13010120] [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: 11/23/2023] [Revised: 12/11/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
The impact of SARS-CoV-2 infections in children has fortunately been lower than what has been seen in adults. However, even previously healthy children have developed severe disease, sometimes with subsequent mortality, and those who are infants or adolescents, are from racial and ethnic minority groups, or have certain chronic conditions are at higher risk of these outcomes. During the pandemic, extensive studies of therapeutic agents, including antivirals and immunomodulators, were conducted in adults. Few trials included children, and most were in older children and adolescents. Thus, the potential benefits of therapies in children must be extrapolated from adult evidence. Despite these limitations, advisory committees of the National Institute of Health (NIH), the Infectious Disease Society of America (IDSA), and the Pediatric Infectious Diseases Society (PIDS) were constituted, and expert consensus guidelines were developed. This review provides a synthesis of those comprehensive recommendations for therapy in children. These address treatment during the early infectious period with antiviral agents, including remdesivir and nirmatrelvir/ritonavir, as well as treatment in the later period of immune dysregulation with corticosteroids and immunomodulators. In addition, the therapeutic approach for multisystem inflammatory syndrome in children (MIS-C), also referred to as Pediatric Inflammatory Multisystem Syndrome temporally associated with SARS-CoV-2 (PIMS-TS), is also provided.
Collapse
Affiliation(s)
- Kathryn M Edwards
- Division of Infectious Diseases, Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| |
Collapse
|
110
|
Bragazzi Cunha J, Leix K, Sherman EJ, Mirabelli C, Frum T, Zhang CJ, Kennedy AA, Lauring AS, Tai AW, Sexton JZ, Spence JR, Wobus CE, Emmer BT. Type I interferon signaling induces a delayed antiproliferative response in respiratory epithelial cells during SARS-CoV-2 infection. J Virol 2023; 97:e0127623. [PMID: 37975674 PMCID: PMC10734423 DOI: 10.1128/jvi.01276-23] [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: 08/29/2023] [Accepted: 10/22/2023] [Indexed: 11/19/2023] Open
Abstract
ABSTRACT Disease progression during SARS-CoV-2 infection is tightly linked to the fate of lung epithelial cells, with severe cases of COVID-19 characterized by direct injury of the alveolar epithelium and an impairment in its regeneration from progenitor cells. The molecular pathways that govern respiratory epithelial cell death and proliferation during SARS-CoV-2 infection, however, remain unclear. We now report a high-throughput CRISPR screen for host genetic modifiers of the survival and proliferation of SARS-CoV-2-infected Calu-3 respiratory epithelial cells. The top four genes identified in our screen encode components of the same type I interferon (IFN-I) signaling complex—IFNAR1, IFNAR2, JAK1, and TYK2. The fifth gene, ACE2, was an expected control encoding the SARS-CoV-2 viral receptor. Surprisingly, despite the antiviral properties of IFN-I signaling, its disruption in our screen was associated with an increase in Calu-3 cell fitness. We validated this effect and found that IFN-I signaling did not sensitize SARS-CoV-2-infected cultures to cell death but rather inhibited the proliferation of surviving cells after the early peak of viral replication and cytopathic effect. We also found that IFN-I signaling alone, in the absence of viral infection, was sufficient to induce this delayed antiproliferative response in both Calu-3 cells and iPSC-derived type 2 alveolar epithelial cells. Together, these findings highlight a cell autonomous antiproliferative response by respiratory epithelial cells to persistent IFN-I signaling during SARS-CoV-2 infection. This response may contribute to the deficient alveolar regeneration that has been associated with COVID-19 lung injury and represents a promising area for host-targeted therapeutic development.
Collapse
Affiliation(s)
- Juliana Bragazzi Cunha
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kyle Leix
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Emily J. Sherman
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Carmen Mirabelli
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Tristan Frum
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Charles J. Zhang
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrew A. Kennedy
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Adam S. Lauring
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Andrew W. Tai
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Jonathan Z. Sexton
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason R. Spence
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - Christiane E. Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Brian T. Emmer
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
111
|
Mettananda C, Peiris C, Abeyrathna D, Gunasekara A, Egodage T, Dantanarayana C, Pathmeswaran A, Ranasinha C. Inhaled beclomethasone in the treatment of early COVID-19: a double-blind, placebo-controlled, randomised, hospital-based trial in Sri Lanka. BMJ Open 2023; 13:e075803. [PMID: 38101843 PMCID: PMC10729268 DOI: 10.1136/bmjopen-2023-075803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023] Open
Abstract
OBJECTIVES To study if early initiation of inhaled beclomethasone 1200 mcg in patients with asymptomatic, mild or moderate COVID-19 reduces disease progression to severe COVID-19. DESIGN Double-blinded, parallel-groups, randomised, placebo-controlled trial. SETTING A hospital-based study in Sri Lanka. PARTICIPANTS Adults with asymptomatic, mild or moderate COVID-19, presenting within the first 7 days of symptom onset or laboratory diagnosis of COVID-19, admitted to a COVID-19 intermediate treatment centre in Sri Lanka between July and November 2021. INTERVENTIONS All participants received inhaled beclomethasone 600 mcg or placebo two times per day, for 10 days from onset of symptoms/COVID-19 test becoming positive if asymptomatic or until reaching primary endpoint, whichever is earlier. PRIMARY OUTCOME MEASURE Progression of asymptomatic, mild or moderate COVID-19 to severe COVID-19. SECONDARY OUTCOME MEASURES The number of days with a temperature of 38°C or more and the time to self-reported clinical recovery. RESULTS A total of 385 participants were randomised to receive beclomethasone(n=193) or placebo(n=192) stratified by age (≤60 or >60 years) and sex. One participant from each arm withdrew from the study. All participants were included in final analysis. Primary outcome occurred in 24 participants in the beclomethasone group and 26 participants in the placebo group (RR 0.90 ; p=0.763). The median time for self-reported clinical recovery in all participants was 5 days (95% CI 3 to 7) in the beclomethasone group and 5 days (95% CI 3 to 8) in the placebo group (p=0.5). The median time for self-reported clinical recovery in patients with moderate COVID-19 was 5 days (95% CI 3 to 7) in the beclomethasone group and 6 days (95% CI 4 to 9) in the placebo group (p=0.05). There were no adverse events. CONCLUSIONS Early initiation of inhaled beclomethasone in patients with asymptomatic, mild or moderate COVID-19 did not reduce disease progression to severe COVID-19. TRIAL REGISTRATION NUMBER Sri Lanka Clinical Trials Registry; SLCTR/2021/017.
Collapse
Affiliation(s)
- Chamila Mettananda
- Department of Pharmacology, University of Kelaniya, Kelaniya, Sri Lanka
- Clinical trials unit, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Chathura Peiris
- COVID-19 Intermediate Treatment Centre, Base Hospital, Kandana, Sri Lanka
| | | | - Aloka Gunasekara
- Department of Pharmacology, University of Kelaniya, Kelaniya, Sri Lanka
| | - Thimira Egodage
- Department of Pharmacology, University of Kelaniya, Kelaniya, Sri Lanka
- Clinical trials unit, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | - Channaka Dantanarayana
- Department of Pharmacology, University of Kelaniya, Kelaniya, Sri Lanka
- Clinical trials unit, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| | | | - Channa Ranasinha
- Department of Pharmacology, University of Kelaniya, Kelaniya, Sri Lanka
- Clinical trials unit, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka
| |
Collapse
|
112
|
Waibel M, Wentworth JM, So M, Couper JJ, Cameron FJ, MacIsaac RJ, Atlas G, Gorelik A, Litwak S, Sanz-Villanueva L, Trivedi P, Ahmed S, Martin FJ, Doyle ME, Harbison JE, Hall C, Krishnamurthy B, Colman PG, Harrison LC, Thomas HE, Kay TWH. Baricitinib and β-Cell Function in Patients with New-Onset Type 1 Diabetes. N Engl J Med 2023; 389:2140-2150. [PMID: 38055252 DOI: 10.1056/nejmoa2306691] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
BACKGROUND Janus kinase (JAK) inhibitors, including baricitinib, block cytokine signaling and are effective disease-modifying treatments for several autoimmune diseases. Whether baricitinib preserves β-cell function in type 1 diabetes is unclear. METHODS In this phase 2, double-blind, randomized, placebo-controlled trial, we assigned patients with type 1 diabetes diagnosed during the previous 100 days to receive baricitinib (4 mg once per day) or matched placebo orally for 48 weeks. The primary outcome was the mean C-peptide level, determined from the area under the concentration-time curve, during a 2-hour mixed-meal tolerance test at week 48. Secondary outcomes included the change from baseline in the glycated hemoglobin level, the daily insulin dose, and measures of glycemic control assessed with the use of continuous glucose monitoring. RESULTS A total of 91 patients received baricitinib (60 patients) or placebo (31 patients). The median of the mixed-meal-stimulated mean C-peptide level at week 48 was 0.65 nmol per liter per minute (interquartile range, 0.31 to 0.82) in the baricitinib group and 0.43 nmol per liter per minute (interquartile range, 0.13 to 0.63) in the placebo group (P = 0.001). The mean daily insulin dose at 48 weeks was 0.41 U per kilogram of body weight per day (95% confidence interval [CI], 0.35 to 0.48) in the baricitinib group and 0.52 U per kilogram per day (95% CI, 0.44 to 0.60) in the placebo group. The levels of glycated hemoglobin were similar in the two trial groups. However, the mean coefficient of variation of the glucose level at 48 weeks, as measured by continuous glucose monitoring, was 29.6% (95% CI, 27.8 to 31.3) in the baricitinib group and 33.8% (95% CI, 31.5 to 36.2) in the placebo group. The frequency and severity of adverse events were similar in the two trial groups, and no serious adverse events were attributed to baricitinib or placebo. CONCLUSIONS In patients with type 1 diabetes of recent onset, daily treatment with baricitinib over 48 weeks appeared to preserve β-cell function as estimated by the mixed-meal-stimulated mean C-peptide level. (Funded by JDRF International and others; BANDIT Australian New Zealand Clinical Trials Registry number, ACTRN12620000239965.).
Collapse
Affiliation(s)
- Michaela Waibel
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - John M Wentworth
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Michelle So
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Jennifer J Couper
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Fergus J Cameron
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Richard J MacIsaac
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Gabby Atlas
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Alexandra Gorelik
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Sara Litwak
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Laura Sanz-Villanueva
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Prerak Trivedi
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Simi Ahmed
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Francis J Martin
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Madeleine E Doyle
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Jessica E Harbison
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Candice Hall
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Balasubramanian Krishnamurthy
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Peter G Colman
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Leonard C Harrison
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Helen E Thomas
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| | - Thomas W H Kay
- From St. Vincent's Institute of Medical Research (M.W., M.S., S.L., L.S.-V., P.T., M.E.D., C.H., B.K., H.E.T., T.W.H.K.), St. Vincent's Hospital Melbourne (R.J.M., B.K., T.W.H.K.), and the Department of Medicine at St. Vincent's Hospital, University of Melbourne (R.J.M., L.S.-V., M.E.D., B.K., H.E.T., T.W.H.K.), Fitzroy, the Walter and Eliza Hall Institute of Medical Research (J.M.W., P.G.C., L.C.H.), the Departments of Medical Biology (J.M.W., L.C.H.) and Medicine (A.G.), University of Melbourne, the Royal Melbourne Hospital (J.M.W., M.S., C.H., P.G.C., L.C.H.), the Royal Children's Hospital (F.J.C., G.A.), and the Murdoch Children's Research Institute (F.J.C.), Parkville, and the School of Public Health and Preventive Medicine, Monash University, Melbourne (A.G.), VIC, and Women's and Children's Hospital (J.J.C., J.E.H.) and the University of Adelaide (J.J.C.), Adelaide, SA - all in Australia; the New York Stem Cell Foundation, New York (S.A.); and Macromoltek, Austin, TX (F.J.M.)
| |
Collapse
|
113
|
Levinson T, Wasserman A, Shenhar-Tsarfaty S, Halutz O, Shapira I, Zeltser D, Rogowski O, Berliner S, Ziv-Baran T. Comparative analysis of CRP as a biomarker of the inflammatory response intensity among common viral infections affecting the lungs: COVID-19 versus influenza A, influenza B and respiratory syncytial virus. Clin Exp Med 2023; 23:5307-5313. [PMID: 37640989 DOI: 10.1007/s10238-023-01176-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
Severe acute respiratory syndrome-corona virus 2 (SARS-CoV-2) is associated with significant morbidity and mortality. C-reactive protein (CRP) is a useful inflammatory biomarker for patients admitted with an infection. This study aimed to compare CRP level as an indicator of inflammation severity between SARS-CoV-2 and common respiratory viral infections. A cross-sectional study of all adult patients hospitalized in the internal medicine department, geriatric department, or internal intensive care unit between 02/2012 and 06/2021 with laboratory-confirmed respiratory viral infection was performed. SARS-CoV-2, influenza A, influenza B, and respiratory syncytial virus (RSV) were studied. Patients with laboratory-confirmed concurrent viral or bacterial infections were excluded. Patients with malignancy were also excluded. Age, gender, comorbidities, and CRP level upon admission were compared between groups. Univariate and multivariable analyses were applied. Among 1124 patients, 18.2% had SARS‑CoV‑2, 48.3% influenza A, 18.9% RSV, and 14.6% influenza B. SARS‑CoV‑2 patients were significantly younger (median 69.4 vs. ≥ 76 years) and had lower Charlson score (median 3 vs. ≥ 4 in other groups) compared to patients with other viral pathogens. After adjustment for patients' age, gender and comorbidities, SARS‑CoV‑2 patients had a higher probability (OR = 1.84-2.02, p < 0.01) of having CRP values in the upper quartile (> 117 mg/L) compared to all other viral pathogens while between all others there was no significant difference. To conclude, a higher CRP level upon admission is approximately twice more common among SARS-CoV-2 patients compared to other widespread respiratory viruses which may demonstrate the higher intensity of inflammation caused by SARS-CoV-2.
Collapse
Affiliation(s)
- Tal Levinson
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
- Infectious Diseases Unit, Tel-Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Asaf Wasserman
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Shani Shenhar-Tsarfaty
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Ora Halutz
- Clinical Microbiology Laboratory, Tel-Aviv Sourasky Medical Center, 6 Weizmann Street, 6423906, Tel-Aviv, Israel
| | - Itzhak Shapira
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - David Zeltser
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Ori Rogowski
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Shlomo Berliner
- Department of Internal Medicine "C" and "E", Tel Aviv Sourasky Medical Center, Faculty of Medicine, Tel Aviv University, 6 Weizmann Street, 6423906, Tel Aviv, Israel
| | - Tomer Ziv-Baran
- Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medicine, Tel Aviv University, P.O.B. 39040, 6997801, Tel Aviv, Israel.
| |
Collapse
|
114
|
Goury A, Mourvillier B. Treatment of severe COVID-19: a role for JAK and complement inhibitors? THE LANCET. RESPIRATORY MEDICINE 2023; 11:1036-1037. [PMID: 37977160 DOI: 10.1016/s2213-2600(23)00423-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Affiliation(s)
- Antoine Goury
- CHU Reims, Médecine Intensive et Réanimation Polyvalente, F-51100 Reims, France
| | - Bruno Mourvillier
- CHU Reims, Médecine Intensive et Réanimation Polyvalente, F-51100 Reims, France; Université de Reims Champagne-Ardenne, Reims, France.
| |
Collapse
|
115
|
Hall FC, Cheriyan J, Cope AP, Galloway J, Wilkinson I, Bond S, Norton S, Banham-Hall E, Bayes H, Kostapanos M, Nodale M, Petchey WG, Sheeran T, Underwood J, Jayne DR. Efficacy and safety of baricitinib or ravulizumab in adult patients with severe COVID-19 (TACTIC-R): a randomised, parallel-arm, open-label, phase 4 trial. THE LANCET. RESPIRATORY MEDICINE 2023; 11:1064-1074. [PMID: 37977159 PMCID: PMC10682367 DOI: 10.1016/s2213-2600(23)00376-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 08/31/2023] [Accepted: 10/03/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND From early in the COVID-19 pandemic, evidence suggested a role for cytokine dysregulation and complement activation in severe disease. In the TACTIC-R trial, we evaluated the efficacy and safety of baricitinib, an inhibitor of Janus kinase 1 (JAK1) and JAK2, and ravulizumab, a monoclonal inhibitor of complement C5 activation, as an adjunct to standard of care for the treatment of adult patients hospitalised with COVID-19. METHODS TACTIC-R was a phase 4, randomised, parallel-arm, open-label platform trial that was undertaken in the UK with urgent public health designation to assess the potential of repurposing immunosuppressants for the treatment of severe COVID-19, stratified by a risk score. Adult participants (aged ≥18 years) were enrolled from 22 hospitals across the UK. Patients with a risk score indicating a 40% risk of admission to an intensive care unit or death were randomly assigned 1:1:1 to standard of care alone, standard of care with baricitinib, or standard of care with ravulizumab. The composite primary outcome was the time from randomisation to incidence (up to and including day 14) of the first event of death, invasive mechanical ventilation, extracorporeal membrane oxygenation, cardiovascular organ support, or renal failure. The primary interim analysis was triggered when 125 patient datasets were available up to day 14 in each study group and we included in the analysis all participants who were randomly assigned. The trial was registered on ClinicalTrials.gov (NCT04390464). FINDINGS Between May 8, 2020, and May 7, 2021, 417 participants were recruited and randomly assigned to standard of care alone (145 patients), baricitinib (137 patients), or ravulizumab (135 patients). Only 54 (39%) of 137 patients in the baricitinib group received the maximum 14-day course, whereas 132 (98%) of 135 patients in the ravulizumab group received the intended dose. The trial was stopped after the primary interim analysis on grounds of futility. The estimated hazard ratio (HR) for reaching the composite primary endpoint was 1·11 (95% CI 0·62-1·99) for patients on baricitinib compared with standard of care alone, and 1·53 (0·88-2·67) for ravulizumab compared with standard of care alone. 45 serious adverse events (21 deaths) were reported in the standard-of-care group, 57 (24 deaths) in the baricitinib group, and 60 (18 deaths) in the ravulizumab group. INTERPRETATION Neither baricitinib nor ravulizumab, as administered in this study, was effective in reducing disease severity in patients selected for severe COVID-19. Safety was similar between treatments and standard of care. The short period of dosing with baricitinib might explain the discrepancy between our findings and those of other trials. The therapeutic potential of targeting complement C5 activation product C5a, rather than the cleavage of C5, warrants further evaluation. FUNDING UK Medical Research Council, UK National Institute for Health Research Cambridge Biomedical Research Centre, Eli Lilly and Company, Alexion Pharmaceuticals, and Addenbrooke's Charitable Trust.
Collapse
Affiliation(s)
- Frances C Hall
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Joseph Cheriyan
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Andrew P Cope
- Centre for Rheumatic Diseases, Kings's College London, London, UK
| | - James Galloway
- Centre for Rheumatic Diseases, Kings's College London, London, UK
| | - Ian Wilkinson
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Simon Bond
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sam Norton
- Centre for Rheumatic Diseases, Kings's College London, London, UK
| | - Edward Banham-Hall
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Hannah Bayes
- Department of Respiratory Medicine, Glasgow Royal Infirmary, Glasgow, UK
| | - Michalis Kostapanos
- Department of Medicine, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Marianna Nodale
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - William G Petchey
- Department of Nephrology, West Suffolk NHS Foundation Trust, Bury St Edmunds, UK
| | - Thomas Sheeran
- Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, UK
| | | | - David R Jayne
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
116
|
Herrera S, Aguado JM, Candel FJ, Cordero E, Domínguez-Gil B, Fernández-Ruiz M, Los Arcos I, Len Ò, Marcos MÁ, Muñez E, Muñoz P, Rodríguez-Goncer I, Sánchez-Céspedes J, Valerio M, Bodro M. Executive summary of the consensus statement of the group for the study of infection in transplantation and other immunocompromised host (GESITRA-IC) of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC) on the treatment of SARS-CoV-2 infection in solid organ transplant recipients. Transplant Rev (Orlando) 2023; 37:100788. [PMID: 37591117 DOI: 10.1016/j.trre.2023.100788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023]
Affiliation(s)
- Sabina Herrera
- Department of Infectious Diseases, Hospital Clínic, IDIBAPS (Institut D'Investigacions Biomèdiques Agust Pi I Sunyer), Universitat de Barcelona, Barcelona, Spain
| | - Jose M Aguado
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Francisco Javier Candel
- Clinical Microbiology & Infectious Diseases, Transplant Coordination, Hospital Clínico Universitario San Carlos, Madrid 28040, Spain; Department of Clinical Microbiology and Infectious Diseases, Hospital Clínico San Carlos, Madrid, Spain
| | - Elisa Cordero
- Infectious Diseases Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina Sevilla, Sevilla, Spain
| | | | - Mario Fernández-Ruiz
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Ibai Los Arcos
- Infectious Diseases Department, Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | - Òscar Len
- Infectious Diseases Department, Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | | | - Elena Muñez
- Infectious Diseases Unit, Internal Medicine Department, University Hospital Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Patricia Muñoz
- Department of Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 9 Madrid, Spain
| | - Isabel Rodríguez-Goncer
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Javier Sánchez-Céspedes
- Infectious Diseases Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina Sevilla, Sevilla, Spain
| | - Maricela Valerio
- Department of Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 9 Madrid, Spain
| | - Marta Bodro
- Department of Infectious Diseases, Hospital Clínic, IDIBAPS (Institut D'Investigacions Biomèdiques Agust Pi I Sunyer), Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
117
|
McKinnon JE, Santiaguel J, Murta de Oliveira C, Yu D, Khursheed M, Moreau F, Klopp‐Schulze L, Shaw J, Roy S, Kao AH. Enpatoran in COVID-19 pneumonia: Safety and efficacy results from a phase II randomized trial. Clin Transl Sci 2023; 16:2640-2653. [PMID: 37873555 PMCID: PMC10719456 DOI: 10.1111/cts.13658] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/25/2023] Open
Abstract
Enpatoran is a selective inhibitor of toll-like receptors 7 and 8 (TLR7/8) that potentially targets pro-inflammatory pathways induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A phase II study conducted in Brazil, the Philippines, and the USA during the early pandemic phase assessed the safety and efficacy of enpatoran in patients hospitalized with COVID-19 pneumonia (NCT04448756). A total of 149 patients, who scored 4 on the World Health Organization's (WHO) 9-point ordinal severity scale, were randomized 1:1:1 and received enpatoran 50 mg (n = 54) or 100 mg (n = 46), or placebo (n = 49) twice daily (b.i.d.) for 14 days plus standard of care. The primary objectives were safety and time to recovery (WHO 9-point scale ≤3). Clinical deterioration (WHO 9-point scale ≥ 5) was a key secondary objective. Treatment-emergent adverse events (TEAEs) were comparable across groups (56.5%-63.0%). Treatment-related TEAEs were numerically higher with enpatoran 50 mg (14.8%) than 100 mg (10.9%) or placebo (8.2%). Serious TEAEs were numerically lower with enpatoran (50 mg 9.3%, 100 mg 2.2%) than placebo (18.4%). The primary efficacy objective was not met; median time to recovery was 3.4-3.9 days across groups, with placebo-treated patients recovering on average faster than anticipated. Clinical deterioration event-free rates up to Day 7 were 90.6%, 95.6%, and 81.6% with enpatoran 50 mg, 100 mg, and placebo, respectively. Enpatoran was well tolerated by patients acutely ill and hospitalized with COVID-19 pneumonia. Positive signals in some secondary end points suggested potential beneficial effects, supporting further evaluation of enpatoran in patients with hyperinflammation due to infection or autoimmunity.
Collapse
Affiliation(s)
- John E. McKinnon
- Division of Infectious Disease, Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Joel Santiaguel
- Division of Pulmonary MedicineUniversity of the PhilippinesManilaPhilippines
| | | | - Dongzi Yu
- Global Clinical Development, EMD SeronoBillericaMassachusettsUSA
| | - Mukhy Khursheed
- Global Patient SafetyMerck Serono Ltd. Feltham, UK, an affiliate of Merck KGaADarmstadtGermany
| | - Flavie Moreau
- Global Biostatistics, EMD SeronoBillericaMassachusettsUSA
| | - Lena Klopp‐Schulze
- Translational Medicine, the healthcare business of Merck KGaADarmstadtGermany
| | - Jamie Shaw
- Translational Medicine, EMD SeronoBillericaMassachusettsUSA
| | - Sanjeev Roy
- Global Clinical DevelopmentAres Trading SA, Eysins, Switzerland, an affiliate of Merck KGaADarmstadtGermany
| | - Amy H. Kao
- Research Unit – Neuroscience & ImmunologyEMD SeronoBillericaMassachusettsUSA
| |
Collapse
|
118
|
Sun J, Wang S, Ma X, Wei Q, Peng Y, Bai Y, Miao G, Meng C, Liu P. Efficacy and safety of baricitinib for the treatment of hospitalized adults with COVID-19: a systematic review and meta-analysis. Eur J Med Res 2023; 28:536. [PMID: 37990249 PMCID: PMC10661565 DOI: 10.1186/s40001-023-01403-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/28/2023] [Indexed: 11/23/2023] Open
Abstract
OBJECTIVES Several clinical trials have evaluated the efficacy and safety of baricitinib in COVID-19 patients. Recently, there have been reports on critical patients, which are different from previous research results. The meta-analysis was performed to investigate the effects of baricitinib in COVID-19, by pooling data from all clinically randomized controlled trials (RCTs) available to increase power to testify. METHODS Studies were searched in PubMed, Embase, and Cochrane Library databases on January 31, 2023. We performed a meta-analysis to estimate the efficacy and safety of baricitinib for the treatment of hospitalized adults with COVID-19. This study is registered with INPLASY, number 202310086. RESULTS A total of 3010 patients were included in our analyses. All included studies were randomized controlled trials or prospective study. There was no difference in 14-day mortality between the two groups [OR 0.23 (95% CI 0.03-1.84), I2 = 72%, P = 0.17]. In subgroup analyses we found that baricitinib did not seem to improve significantly in 24-day mortality critically ill patients [OR 0.60 (95% CI 0.35-1.02), I2 = 0%, P = 0.06]. Fortunately, baricitinib have led to faster recovery and shorter hospital stays for COVID-19 patients. There were no difference in infections and infestations, major adverse cardiovascular events, deep vein thrombosis and pulmonary embolism. CONCLUSIONS Baricitinib did not increase the incidence of adverse reactions. At the same time, we can find that it reduces the mortality of COVID-19 patients, not including the critically ill.
Collapse
Affiliation(s)
- Jing Sun
- Department of Critical Care Medicine, Emergency General Hospital, Beijing, China
| | - Shufang Wang
- Department of Emergency, Emergency General Hospital, Xibahe South Road 29, Chaoyang District, Beijing, 100028, People's Republic of China
| | - Xin Ma
- Department of Critical Care Medicine, Affiliated Hospital of Hebei University, Baoding, China
| | - Qingqing Wei
- Department of Critical Care Medicine, Emergency General Hospital, Beijing, China
| | - Yujuan Peng
- Department of Critical Care Medicine, Emergency General Hospital, Beijing, China
| | - Ying Bai
- Department of Critical Care Medicine, Emergency General Hospital, Beijing, China
| | - Guobin Miao
- Department of Emergency, Emergency General Hospital, Xibahe South Road 29, Chaoyang District, Beijing, 100028, People's Republic of China.
| | - Chang Meng
- Department of Emergency, Emergency General Hospital, Xibahe South Road 29, Chaoyang District, Beijing, 100028, People's Republic of China.
| | - Peng Liu
- Department of Cardiology, Ordos School of Clinical Medicine, Ordos Central Hospital, Inner Mongolia Medical University, 23 Yijin Huoluo West Street, Dongsheng District, Inner Mongolia, 017000, People's Republic of China.
| |
Collapse
|
119
|
Passeron T, King B, Seneschal J, Steinhoff M, Jabbari A, Ohyama M, Tobin DJ, Randhawa S, Winkler A, Telliez JB, Martin D, Lejeune A. Inhibition of T-cell activity in alopecia areata: recent developments and new directions. Front Immunol 2023; 14:1243556. [PMID: 38022501 PMCID: PMC10657858 DOI: 10.3389/fimmu.2023.1243556] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/05/2023] [Indexed: 12/01/2023] Open
Abstract
Alopecia areata (AA) is an autoimmune disease that has a complex underlying immunopathogenesis characterized by nonscarring hair loss ranging from small bald patches to complete loss of scalp, face, and/or body hair. Although the etiopathogenesis of AA has not yet been fully characterized, immune privilege collapse at the hair follicle (HF) followed by T-cell receptor recognition of exposed HF autoantigens by autoreactive cytotoxic CD8+ T cells is now understood to play a central role. Few treatment options are available, with the Janus kinase (JAK) 1/2 inhibitor baricitinib (2022) and the selective JAK3/tyrosine kinase expressed in hepatocellular carcinoma (TEC) inhibitor ritlecitinib (2023) being the only US Food and Drug Administration-approved systemic medications thus far for severe AA. Several other treatments are used off-label with limited efficacy and/or suboptimal safety and tolerability. With an increased understanding of the T-cell-mediated autoimmune and inflammatory pathogenesis of AA, additional therapeutic pathways beyond JAK inhibition are currently under investigation for the development of AA therapies. This narrative review presents a detailed overview about the role of T cells and T-cell-signaling pathways in the pathogenesis of AA, with a focus on those pathways targeted by drugs in clinical development for the treatment of AA. A detailed summary of new drugs targeting these pathways with expert commentary on future directions for AA drug development and the importance of targeting multiple T-cell-signaling pathways is also provided in this review.
Collapse
Affiliation(s)
- Thierry Passeron
- University Côte d’Azur, Centre Hospitalier Universitaire Nice, Department of Dermatology, Nice, France
- University Côte d’Azur, INSERM, U1065, C3M, Nice, France
| | - Brett King
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, United States
| | - Julien Seneschal
- Department of Dermatology and Paediatric Dermatology, National Reference Centre for Rare Skin Diseases, Saint-André Hospital, University of Bordeaux, Bordeaux, France
- Bordeaux University, Centre national de la recherche scientifique (CNRS), ImmunoConcept, UMR5164, Bordeaux, France
| | - Martin Steinhoff
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Dermatology and Venereology, Weill Cornell Medicine-Qatar, Doha, Qatar
- College of Medicine, Qatar University, Doha, Qatar
- Department of Dermatology, Weill Cornell Medicine, New York, NY, United States
- College of Health and Life Sciences, Hamad Bin Khalifa University-Qatar, Doha, Qatar
| | - Ali Jabbari
- Department of Dermatology, University of Iowa, Iowa City, IA, United States
- Iowa City VA Medical Center, Iowa City, IA, United States
| | - Manabu Ohyama
- Department of Dermatology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Desmond J. Tobin
- Charles Institute of Dermatology, UCD School of Medicine, University College Dublin, Dublin, Ireland
| | | | | | | | | | | |
Collapse
|
120
|
Bagri NK, Chew C, Ramanan AV. Scope of JAK Inhibitors in Children: Recent Evidence and Way Forward. Paediatr Drugs 2023; 25:635-647. [PMID: 37775678 DOI: 10.1007/s40272-023-00594-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 10/01/2023]
Abstract
Over the last decade, there has been an increase in the use of targeted therapy using small molecules such as Janus kinase (JAK) inhibitors. Since the introduction of ruxolitinib, the first non-selective JAK inhibitor approved for use in myelofibrosis, many other JAK inhibitors have been tried in a wide spectrum of immune-mediated disorders. Although various trials have shown the promising efficacy of JAK inhibitors in immune-mediated inflammatory disorders (IMIDs), there is a growing concern over the major cardiovascular events and malignancies associated with the use of these molecules in older adults, particularly those over 65 years of age. In this review, we aim to discuss the immunology of the JAK-STAT pathway, the scope of use of JAK inhibitors, and their safety in paediatric practice. Here, we discuss high-quality evidence favouring the use of JAK inhibitors in children with juvenile idiopathic arthritis (JIA) who are refractory to one or more conventional/biological disease-modifying drugs, demonstrated in two randomised controlled trials (RCTs). In addition to JIA, there are reports favouring the role of JAK inhibitors in other IMIDs such as systemic-onset JIA and interferonopathies. Thus far, the existing literature suggests an acceptable safety profile for JAK inhibitors in children. With the expanding scope of JAK inhibitors in a wide range of IMIDs in children, there is a significant need for long-term close vigilance for any potential harm.
Collapse
Affiliation(s)
- Narendra Kumar Bagri
- Division of Pediatric Rheumatology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Christine Chew
- Department of Paediatric Rheumatology, Bristol Royal Hospital for Children, Upper Maudlin Street, Bristol, UK
- School of Cellular and Molecular Medicine, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - A V Ramanan
- Department of Paediatric Rheumatology, Bristol Royal Hospital for Children, Upper Maudlin Street, Bristol, UK.
- Translational Health Sciences, University of Bristol, Bristol, UK.
| |
Collapse
|
121
|
Liu H, Chen J, Shao W, Yan S, Ding S. Efficacy and Safety of Novel Oral Antivirals in Hospitalized COVID-19 Patients: A Network Meta-Analysis of Randomized Clinical Trials. Clin Epidemiol 2023; 15:1041-1053. [PMID: 37933389 PMCID: PMC10625770 DOI: 10.2147/clep.s422386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/26/2023] [Indexed: 11/08/2023] Open
Abstract
OBJECTIVE Numerous pharmacological interventions are now under investigation for the treatment of the 2019 coronavirus pandemic (COVID-19), and the evidence is rapidly evolving. Our aim is to evaluate the comparative efficacy and safety of these drugs. METHODS We searched for randomized clinical trials (RCTs) on the efficacy and safety of novel oral antivirals for the treatment of hospitalized COVID-19 patients until November 30, 2022, including baricitinib, ivermectin (IVM), favipiravir (FVP), chloroquine (CQ), lopinavir and ritonavir (LPV/RTV), hydroxychloroquine (HCQ), and hydroxychloroquine plus azithromycin (HCQ+AZT). The main outcomes of this network meta-analysis (NMA) were in-hospital mortality, adverse event (AE), recovery time, and improvement in peripheral capillary oxygen saturation (SpO2). For dichotomous results, the odds ratio (OR) was used, and the 95% confidence interval (CI) was determined. We also used meta-regression to explore whether different treatments affected efficacy and safety. STATA 15.0 was used to conduct the NMA. The research protocol was registered with PROSPERO (#CRD 42023415743). RESULTS Thirty-six RCTs, with 33,555 hospitalized COVID-19 patients, were included in this analysis. First, we compared the efficacy of different novel oral antivirals. Baricitinib (OR 0.56, 95% CI: 0.35 to 0.90) showed the highest probability of being the optimal probiotic species in reducing in-hospital mortality and suggested that none of the interventions reduced AE better than placebo. In terms of safety outcomes, IVM ranked first in improving the recovery time of hospitalized COVID-19 patients (mean difference (MD) -1.36, 95% CI: -2.32 to -0.39). In addition, patients were most likely to increase SpO2 (OR 1.77, 95% CI: 0.09 to 3.45). The meta-regression revealed no significant differences between participants using different novel oral antivirals in all outcomes in hospitalized COVID-19 patients. CONCLUSION Currently, baricitinib has reduced in-hospital mortality in hospitalized COVID-19 patients, with moderate certainty of evidence. IVM appeared to be a safer option than placebo in improving recovery time, while FVP was associated with increased SpO2 safety outcomes. These preliminary evidence-based observations should guide clinical practice until more data are made public.
Collapse
Affiliation(s)
- Haoshuang Liu
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| | - Jingfeng Chen
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| | - Weihao Shao
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, People's Republic of China
| | - Su Yan
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| | - Suying Ding
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| |
Collapse
|
122
|
Sahanic S, Hilbe R, Dünser C, Tymoszuk P, Löffler-Ragg J, Rieder D, Trajanoski Z, Krogsdam A, Demetz E, Yurchenko M, Fischer C, Schirmer M, Theurl M, Lener D, Hirsch J, Holfeld J, Gollmann-Tepeköylü C, Zinner CP, Tzankov A, Zhang SY, Casanova JL, Posch W, Wilflingseder D, Weiss G, Tancevski I. SARS-CoV-2 activates the TLR4/MyD88 pathway in human macrophages: A possible correlation with strong pro-inflammatory responses in severe COVID-19. Heliyon 2023; 9:e21893. [PMID: 38034686 PMCID: PMC10686889 DOI: 10.1016/j.heliyon.2023.e21893] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 09/26/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Background Toll-like receptors (TLRs) play a pivotal role in the immunologic response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Exaggerated inflammatory response of innate immune cells, however, may drive morbidity and death in Coronavirus disease 19 (COVID-19). Objective We investigated the engagement of SARS-CoV-2 with TLR4 in order to better understand how to tackle hyperinflammation in COVID-19. Methods We combined RNA-sequencing data of human lung tissue and of bronchoalveolar lavage fluid cells derived from COVID-19 patients with functional studies in human macrophages using SARS-CoV-2 spike proteins and viable SARS-CoV-2. Pharmacological inhibitors as well as gene editing with CRISPR/Cas9 were used to delineate the signalling pathways involved. Results We found TLR4 to be the most abundantly upregulated TLR in human lung tissue irrespective of the underlying pathology. Accordingly, bronchoalveolar lavage fluid cells from patients with severe COVID-19 showed an NF-κB-pathway dominated immune response, whereas they were mostly defined by type I interferon signalling in moderate COVID-19. Mechanistically, we found the Spike ectodomain, but not receptor binding domain monomer to induce TLR4-dependent inflammation in human macrophages. By using pharmacological inhibitors as well as CRISPR/Cas9 deleted macrophages, we identify SARS-CoV-2 to engage canonical TLR4-MyD88 signalling. Importantly, we demonstrate that TLR4 blockage prevents exaggerated inflammatory responses in human macrophages infected with different SARS-CoV-2 variants, including immune escape variants B.1.1.7.-E484K and B.1.1.529 (omicron). Conclusion Our study critically extends the current knowledge on TLR-mediated hyperinflammatory responses to SARS-CoV-2 in human macrophages, paving the way for novel approaches to tackle severe COVID-19. Take-home message Our study combining human lung transcriptomics with functional studies in human macrophages clearly supports the design and development of TLR4 - directed therapeutics to mitigate hyperinflammation in severe COVID-19.
Collapse
Affiliation(s)
- Sabina Sahanic
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Richard Hilbe
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Christina Dünser
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Judith Löffler-Ragg
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Dietmar Rieder
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Zlatko Trajanoski
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Anne Krogsdam
- Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Egon Demetz
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Maria Yurchenko
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
- The Central Norway Regional Health Authority, St. Olavs Hospital HF, Trondheim, Norway
| | - Christine Fischer
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Schirmer
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Theurl
- Department of Internal Medicine III, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Lener
- Department of Internal Medicine III, Medical University of Innsbruck, Innsbruck, Austria
| | - Jakob Hirsch
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Johannes Holfeld
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Carl P. Zinner
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Alexandar Tzankov
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, 10065, USA
| | - Wilfried Posch
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Austria
| | - Doris Wilflingseder
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, Austria
| | - Guenter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
123
|
Mustafa SS, Stern RA, Patel PC, Chu DK. COVID-19 Treatments: Then and Now. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:3321-3333. [PMID: 37558163 DOI: 10.1016/j.jaip.2023.07.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has evolved over the past 3+ years, and strategies to prevent illness and treat infection have changed over time. As COVID-19 transitions from a pandemic to an endemic infection, widespread nonpharmaceutical interventions such as mask mandates and governmental policies requiring social distancing have given way to more selective strategies for risk mitigation. Monoclonal antibody therapies used for disease prevention and treatment lost utility owing to the emergence of resistant viral variants. Oral antiviral medications have become the mainstay of treatment in nonhospitalized individuals, whereas systemic corticosteroids remain the cornerstone of therapy in those requiring supplemental oxygen. Emerging literature also supports the use of additional immune-modulating therapies in select admitted patients. Importantly, the COVID-19 pandemic highlighted both unprecedented research and development of medical interventions while also drawing attention to significant pitfalls in the global response. This review provides a comprehensive update in prevention and management of COVID-19.
Collapse
Affiliation(s)
- S Shahzad Mustafa
- Department of Medicine, Rochester Regional Health, Rochester, NY; Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| | - Rebecca A Stern
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Pratish C Patel
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Derek K Chu
- Department of Medicine, Evidence in Allergy Group, Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ont, Canada
| |
Collapse
|
124
|
Piñana JL, Pérez A, Chorão P, Guerreiro M, García-Cadenas I, Solano C, Martino R, Navarro D. Respiratory virus infections after allogeneic stem cell transplantation: Current understanding, knowledge gaps, and recent advances. Transpl Infect Dis 2023; 25 Suppl 1:e14117. [PMID: 37585370 DOI: 10.1111/tid.14117] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023]
Abstract
Before the COVID-19 pandemic, common community-acquired seasonal respiratory viruses (CARVs) were a significant threat to the health and well-being of allogeneic hematopoietic cell transplant (allo-HCT) recipients, often resulting in severe illness and even death. The pandemic has further highlighted the significant risk that immunosuppressed patients, including allo-HCT recipients, face when infected with SARS-CoV-2. As preventive transmission measures are relaxed and CARVs circulate again among the community, including in allo-HSCT recipients, it is crucial to understand the current state of knowledge, gaps, and recent advances regarding CARV infection in allo-HCT recipients. Urgent research is needed to identify seasonal respiratory viruses as potential drivers for future pandemics.
Collapse
Affiliation(s)
- Jose L Piñana
- Hematology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Ariadna Pérez
- Hematology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Pedro Chorão
- Hematology Division, Hospital universitario y politécnico La Fe, Valencia, Spain
- Instituto de Investigación La Fe, Hospital Universitário y Politécncio La Fe, Valencia, Spain
| | - Manuel Guerreiro
- Hematology Division, Hospital universitario y politécnico La Fe, Valencia, Spain
- Instituto de Investigación La Fe, Hospital Universitário y Politécncio La Fe, Valencia, Spain
| | | | - Carlos Solano
- Hematology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
- Fundación INCLIVA, Instituto de Investigación Sanitaria Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - Rodrigo Martino
- Hematology Division, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - David Navarro
- Microbiology department, Hospital Clinico Universitario de Valencia, Spain
- Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain
| |
Collapse
|
125
|
He C, Xu Y, Zhou Y, Fan J, Cheng C, Meng R, Gamazon ER, Zhou D. Integrating population-level and cell-based signatures for drug repositioning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.25.564079. [PMID: 37961219 PMCID: PMC10634827 DOI: 10.1101/2023.10.25.564079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Drug repositioning presents a streamlined and cost-efficient way to expand the range of therapeutic possibilities. Furthermore, drugs with genetic evidence are more likely to progress successfully through clinical trials towards FDA approval. Exploiting these developments, single gene-based drug repositioning methods have been implemented, but approaches leveraging the entire spectrum of molecular signatures are critically underexplored. Most multi-gene-based approaches rely on differential gene expression (DGE) analysis, which is prone to identify the molecular consequence of disease and renders causal inference challenging. We propose a framework TReD (Transcriptome-informed Reversal Distance) that integrates population-level disease signatures robust to reverse causality and cell-based drug-induced transcriptome response profiles. TReD embeds the disease signature and drug profile in a high-dimensional normed space, quantifying the reversal potential of candidate drugs in a disease-related cell screen assay. The robustness is ensured by evaluation in additional cell screens. For an application, we implement the framework to identify potential drugs against COVID-19. Taking transcriptome-wide association study (TWAS) results from four relevant tissues and three DGE results as disease features, we identify 37 drugs showing potential reversal roles in at least four of the seven disease signatures. Notably, over 70% (27/37) of the drugs have been linked to COVID-19 from other studies, and among them, eight drugs are supported by ongoing/completed clinical trials. For example, TReD identifies the well-studied JAK1/JAK2 inhibitor baricitinib, the first FDA-approved immunomodulatory treatment for COVID-19. Novel potential candidates, including enzastaurin, a selective inhibitor of PKC-beta which can be activated by SARS-CoV-2, are also identified. In summary, we propose a comprehensive genetics-anchored framework integrating population-level signatures and cell-based screens that can accelerate the search for new therapeutic strategies.
Collapse
|
126
|
Harrington R, Harkins P, Conway R. Janus Kinase Inhibitors in Rheumatoid Arthritis: An Update on the Efficacy and Safety of Tofacitinib, Baricitinib and Upadacitinib. J Clin Med 2023; 12:6690. [PMID: 37892827 PMCID: PMC10607454 DOI: 10.3390/jcm12206690] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Janus kinase inhibitors (JAKis) are the most recent new drug class to arrive to the market for rheumatoid arthritis (RA) treatment. While they have proven to be a very effective treatment option, there remains significant concern regarding the risk of cardiovascular events, thrombosis and malignancy, particularly given the findings of the post-marketing ORAL Surveillance study and FDA black box warnings. This article reviews the key findings of the most impactful cohort of studies and registry data since ORAL Surveillance. It also evaluates the role of JAKis in practice and offers guidance on risk stratifying patients and determining their suitability for a JAKi.
Collapse
Affiliation(s)
- Robert Harrington
- Department of Rheumatology, St. James’s Hospital, James Street, Dublin 8, D08 NHY1 Dublin, Ireland;
| | | | - Richard Conway
- Department of Rheumatology, St. James’s Hospital, James Street, Dublin 8, D08 NHY1 Dublin, Ireland;
| |
Collapse
|
127
|
Guo W, Zheng Y, Feng S. Omicron related COVID-19 prevention and treatment measures for patients with hematological malignancy and strategies for modifying hematologic treatment regimes. Front Cell Infect Microbiol 2023; 13:1207225. [PMID: 37928188 PMCID: PMC10622671 DOI: 10.3389/fcimb.2023.1207225] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/20/2023] [Indexed: 11/07/2023] Open
Abstract
The Omicron variant of SARS-CoV-2 has rapidly become the dominant strain worldwide due to its high transmissibility, although it appears to be less pathogenic than previous strains. However, individuals with hematological malignancy (HM) and COVID-19 remain susceptible to severe infection and mortality, especially those with chronic lymphocytic leukemia (CLL) and those undergoing chimeric antigen receptor T-cell (CAR-T) treatment. Hematologists should thoroughly assess the severity of the patient's hematological disease and the potential risk of SARS-CoV-2 infection before initiating chemotherapy or immunosuppressive treatment. Vaccination and booster doses are strongly recommended and patients with a poor vaccine response may benefit from long-acting COVID-19 neutralizing monoclonal antibodies (such as Evusheld). Early use of small molecule antiviral drugs is recommended for managing mild COVID-19 in HM patients and those with severe immunodeficiency may benefit from SARS-CoV-2 neutralizing monoclonal antibody therapy and high-titer COVID-19 convalescent plasma (CCP). For moderate to severe cases, low-dose glucocorticoids in combination with early antiviral treatment can be administered, with cytokine receptor antagonists or JAK inhibitors added if the condition persists or worsens. In the treatment of hematological malignancies, delaying chemotherapy is preferable for CLL, acute leukemia (AL), and low-risk myelodysplastic syndrome (MDS), but if the disease progresses, appropriate adjustments in dosage and frequency of treatment are required, with the avoidance of anti-CD20 monoclonal antibody, CAR-T and hematopoietic stem cell transplantation (HSCT). Patients with chronic myelocytic leukemia (CML) and myeloproliferative neoplasms (MPNs) can continue current treatment. What's more, non-drug protective measures, the development of new vaccines and antiviral drugs, and monitoring of mutations in immunocompromised populations are particularly important.
Collapse
Affiliation(s)
- Wenjing Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yizhou Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
128
|
Richardson PJ, Smith DP, de Giorgio A, Snetkov X, Almond-Thynne J, Cronin S, Mead RJ, McDermott CJ, Shaw PJ. Janus kinase inhibitors are potential therapeutics for amyotrophic lateral sclerosis. Transl Neurodegener 2023; 12:47. [PMID: 37828541 PMCID: PMC10568794 DOI: 10.1186/s40035-023-00380-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a poorly treated multifactorial neurodegenerative disease associated with multiple cell types and subcellular organelles. As with other multifactorial diseases, it is likely that drugs will need to target multiple disease processes and cell types to be effective. We review here the role of Janus kinase (JAK)/Signal transducer and activator of transcription (STAT) signalling in ALS, confirm the association of this signalling with fundamental ALS disease processes using the BenevolentAI Knowledge Graph, and demonstrate that inhibitors of this pathway could reduce the ALS pathophysiology in neurons, glia, muscle fibres, and blood cells. Specifically, we suggest that inhibition of the JAK enzymes by approved inhibitors known as Jakinibs could reduce STAT3 activation and modify the progress of this disease. Analysis of the Jakinibs highlights baricitinib as a suitable candidate due to its ability to penetrate the central nervous system and exert beneficial effects on the immune system. Therefore, we recommend that this drug be tested in appropriately designed clinical trials for ALS.
Collapse
Affiliation(s)
| | | | | | | | | | - Sara Cronin
- BenevolentAI, 15 MetroTech Centre, 8th FL, Brooklyn, NY, 11201, USA
| | - Richard J Mead
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
| | - Christopher J McDermott
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, University of Sheffield and Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, University of Sheffield and Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
129
|
Marko M, Pawliczak R. Assessment of the available therapeutic approaches for severe COVID-19: a meta-analysis of randomized controlled trials. Sci Rep 2023; 13:17114. [PMID: 37816884 PMCID: PMC10564895 DOI: 10.1038/s41598-023-44463-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 10/09/2023] [Indexed: 10/12/2023] Open
Abstract
The study aimed to evaluate severe COVID-19 treatment approaches. We conducted a meta-analysis of randomized controlled trials (RTCs) with standard of care (SoC) as a control group and/or placebo. Database searching was performed separately for severe COVID-19 treatment such as anakinra, remdesivir, baricitinib, ivermectin, ritonavir, tocilizumab, sarilumab, sotrovimab, casirivimab/imdevimab. The results are presented as Risk Ratio (RR), 95% Confidence Interval (CI), and heterogeneity (I2). We obtained the most statistically significant outcomes favorable tocilizumab compared to SoC for death incidents RR 0.87 [95% CI 0.80, 0.95], overall effect p = 0.002, heterogeneity p = 0.85, I2 = 0%, need for mechanical ventilation RR 0.78 [95% CI 0.68, 0.89], overall effect p = 0.0004, heterogeneity p = 0.55, I2 = 0%, and number of patients discharged from hospital. RR 1.13 [95% CI 1.07, 1.20], overall effect p < 0.00001, heterogeneity p = 0.009, I2 = 85%. This meta-analysis has revealed that a considerable amount of research characterized by a very diverse methodology is available. Despite the limited data that met the criteria for inclusion in the meta-analysis, we showed that the available treatment options for severe COVID-19 are effective.
Collapse
Affiliation(s)
- Monika Marko
- Division of Biomedical Science, Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, 7/9 Zeligowskiego St, 90-752, Lodz, Poland
| | - Rafał Pawliczak
- Division of Biomedical Science, Department of Immunopathology, Faculty of Medicine, Medical University of Lodz, 7/9 Zeligowskiego St, 90-752, Lodz, Poland.
| |
Collapse
|
130
|
Obradović D, Milovančev A, Plećaš Đurić A, Sovilj-Gmizić S, Đurović V, Šović J, Đurđević M, Tubić S, Bulajić J, Mišić M, Jojić J, Pušara M, Lazić I, Đurković M, Bek Pupovac R, Vulić A, Jozing M. High-Flow Nasal Cannula oxygen therapy in COVID-19: retrospective analysis of clinical outcomes - single center experience. Front Med (Lausanne) 2023; 10:1244650. [PMID: 37849487 PMCID: PMC10577378 DOI: 10.3389/fmed.2023.1244650] [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: 06/22/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023] Open
Abstract
Background High-Flow Nasal Cannula (HFNC) oxygen therapy emerged as the therapy of choice in COVID-19-related pneumonia and moderate to severe acute hypoxemic respiratory failure (AHRF). HFNC oxygen therapy in COVID-19 has been recommended based its use to treat AHRF of other etiologies, and studies on assessing outcomes in COVID-19 patients are highly needed. This study aimed to examine outcomes in COVID-19 patients with pneumonia and severe AHRF treated with HFNC. Materials and methods The study included 235 COVID-19 patients with pneumonia treated with HFNC. Data extracted from medical records included demographic characteristics, comorbidities, laboratory parameters, clinical and oxygenation status, clinical complications, as well as the length of hospital stay. Patients were segregated into two groups based on their oxygen therapy needs: HDU group, those who exclusively required HFNC and ICU group, those whose oxygen therapy needed to be escalated at some point of hospital stay. The primary outcome was the need for respiratory support escalation (noninvasive or invasive mechanical ventilation) and the secondary outcome was the in-hospital all-cause mortality. Results The primary outcome was met in 113 (48%) of patients. The overall mortality was 70%, significantly higher in the ICU group [102 (90.2%) vs. 62 (50.1%), p < 0.001]. The rate of intrahospital infections was significantly higher in the ICU group while there were no significant differences in the length of hospital stay between the groups. The ICU group exhibited significant increases in D-dimer, NLR, and NEWS values, accompanied by a significant decrease in the SaO2/FiO2 ratio. The multivariable COX proportional regression analysis identified malignancy, higher levels of 4C Mortality Score and NEWS2 as significant predictors of mortality. Conclusion High-Flow Nasal Cannula oxygen therapy is a safe type of respiratory support in patients with COVID-19 pneumonia and acute hypoxemic respiratory failure with significantly less possibility for emergence of intrahospital infections. In 52% of patients, HFNC was successful in treating AHRF in COVID-19 patients. Overall, mortality in COVID-19 pneumonia with AHRF is still very high, especially in patients treated with noninvasive/invasive mechanical ventilation.
Collapse
Affiliation(s)
- Dušanka Obradović
- Faculty of Medicine Novi Sad, University of Novi Sad, Novi Sad, Serbia
- Institute for Pulmonary Diseases of Vojvodina, Sremska Kamenica, Serbia
| | - Aleksandra Milovančev
- Faculty of Medicine Novi Sad, University of Novi Sad, Novi Sad, Serbia
- Institute for Cardiovascular Diseases of Vojvodina, Sremska Kamenica, Serbia
| | - Aleksandra Plećaš Đurić
- Faculty of Medicine Novi Sad, University of Novi Sad, Novi Sad, Serbia
- Clinic of Anesthesiology, Intensive Care and Pain Therapy, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | | | - Vladimir Đurović
- Clinic of Nephrology and Clinical Immunology, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Jovica Šović
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Miloš Đurđević
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Stevan Tubić
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Jelena Bulajić
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Milena Mišić
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Jovana Jojić
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Miroslava Pušara
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Ivana Lazić
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Mladen Đurković
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Renata Bek Pupovac
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Aleksandra Vulić
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Marija Jozing
- Urgent Care Center, University Clinical Center of Vojvodina, Novi Sad, Serbia
| |
Collapse
|
131
|
Naik R, Avula S, Palleti SK, Gummadi J, Ramachandran R, Chandramohan D, Dhillon G, Gill AS, Paiwal K, Shaik B, Balachandran M, Patel B, Gurugubelli S, Mariswamy Arun Kumar AK, Nanjundappa A, Bellamkonda M, Rathi K, Sakhamuri PL, Nassar M, Bali A. From Emergence to Endemicity: A Comprehensive Review of COVID-19. Cureus 2023; 15:e48046. [PMID: 37916248 PMCID: PMC10617653 DOI: 10.7759/cureus.48046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2023] [Indexed: 11/03/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), later renamed coronavirus disease 2019 (COVID-19), was first identified in Wuhan, China, in early December 2019. Initially, the China office of the World Health Organization was informed of numerous cases of pneumonia of unidentified etiology in Wuhan, Hubei Province at the end of 2019. This would subsequently result in a global pandemic with millions of confirmed cases of COVID-19 and millions of deaths reported to the WHO. We have analyzed most of the data published since the beginning of the pandemic to compile this comprehensive review of SARS-CoV-2. We looked at the core ideas, such as the etiology, epidemiology, pathogenesis, clinical symptoms, diagnostics, histopathologic findings, consequences, therapies, and vaccines. We have also included the long-term effects and myths associated with some therapeutics of COVID-19. This study presents a comprehensive assessment of the SARS-CoV-2 virology, vaccines, medicines, and significant variants identified during the course of the pandemic. Our review article is intended to provide medical practitioners with a better understanding of the fundamental sciences, clinical treatment, and prevention of COVID-19. As of May 2023, this paper contains the most recent data made accessible.
Collapse
Affiliation(s)
- Roopa Naik
- Medicine, Geisinger Commonwealth School of Medicine, Scranton, USA
- Internal Medicine/Hospital Medicine, Geisinger Health System, Wilkes Barre, USA
| | - Sreekant Avula
- Diabetes, Endocrinology, and Metabolism, University of Minnesota, Minneapolis, USA
| | - Sujith K Palleti
- Nephrology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Jyotsna Gummadi
- Internal Medicine, MedStar Franklin Square Medical Center, Baltimore, USA
| | | | | | - Gagandeep Dhillon
- Physician Executive MBA, University of Tennessee, Knoxville, USA
- Internal Medicine, University of Maryland Baltimore Washington Medical Center, Glen Burnie, USA
| | | | - Kapil Paiwal
- Oral & Maxillofacial Pathology, Daswani Dental College & Research Center, Kota, IND
| | - Bushra Shaik
- Internal Medicine, Onslow Memorial Hospital, Jacksonville, USA
| | | | - Bhumika Patel
- Oral Medicine and Radiology, Howard University, Washington, D.C., USA
| | | | | | | | - Mahita Bellamkonda
- Hospital Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Kanika Rathi
- Internal Medicine, University of Florida, Gainesville, USA
| | | | - Mahmoud Nassar
- Endocrinology, Diabetes, and Metabolism, Jacobs School of Medicine and Biomedical Sciences, Buffalo, USA
| | - Atul Bali
- Internal Medicine/Nephrology, Geisinger Medical Center, Danville, USA
- Internal Medicine/Nephrology, Geisinger Health System, Wilkes-Barre, USA
- Medicine, Geisinger Commonwealth School of Medicine, Scranton, USA
| |
Collapse
|
132
|
Leegwater E, Dol L, Benard MR, Roelofsen EE, Delfos NM, van der Feltz M, Mollema FPN, Bosma LBE, Visser LE, Ottens TH, van Burgel ND, Arbous SM, El Bouazzaoui LH, Knevel R, Groenwold RHH, de Boer MGJ, Visser LG, Rosendaal FR, Wilms EB, van Nieuwkoop C. Rapid Response to Remdesivir in Hospitalised COVID-19 Patients: A Propensity Score Weighted Multicentre Cohort Study. Infect Dis Ther 2023; 12:2471-2484. [PMID: 37801280 PMCID: PMC10600071 DOI: 10.1007/s40121-023-00874-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/13/2023] [Indexed: 10/07/2023] Open
Abstract
INTRODUCTION Remdesivir is a registered treatment for hospitalised patients with COVID-19 that has moderate clinical effectiveness. Anecdotally, some patients' respiratory insufficiency seemed to recover particularly rapidly after initiation of remdesivir. In this study, we investigated if this rapid improvement was caused by remdesivir, and which patient characteristics might predict a rapid clinical improvement in response to remdesivir. METHODS This was a multicentre observational cohort study of hospitalised patients with COVID-19 who required supplemental oxygen and were treated with dexamethasone. Rapid clinical improvement in response to treatment was defined by a reduction of at least 1 L of supplemental oxygen per minute or discharge from the hospital within 72 h after admission. Inverse probability of treatment-weighted logistic regression modelling was used to assess the association between remdesivir and rapid clinical improvement. Secondary endpoints included in-hospital mortality, ICU admission rate and hospitalisation duration. RESULTS Of 871 patients included, 445 were treated with remdesivir. There was no influence of remdesivir on the occurrence of rapid clinical improvement (62% vs 61% OR 1.05, 95% CI 0.79-1.40; p = 0.76). The in-hospital mortality was lower (14.7% vs 19.8% OR 0.70, 95% CI 0.48-1.02; p = 0.06) for the remdesivir-treated patients. Rapid clinical improvement occurred more often in patients with low C-reactive protein (≤ 75 mg/L) and short duration of symptoms prior to hospitalisation (< 7 days) (OR 2.84, 95% CI 1.07-7.56). CONCLUSION Remdesivir generally does not increase the incidence of rapid clinical improvement in hospitalised patients with COVID-19, but it might have an effect in patients with short duration of symptoms and limited signs of systemic inflammation.
Collapse
Affiliation(s)
- Emiel Leegwater
- Department of Hospital Pharmacy, Haga Teaching Hospital, Els Borst-Eilersplein 275, 2545 AA, The Hague, The Netherlands.
- Apotheek Haagse Ziekenhuizen, The Hague, The Netherlands.
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands.
| | - Lisa Dol
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Department of Internal Medicine, Haga Teaching Hospital, The Hague, The Netherlands
- Department of Intensive Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Menno R Benard
- Alrijne Academy, Alrijne Hospital, Leiderdorp, The Netherlands
| | - Eveline E Roelofsen
- Department of Hospital Pharmacy, Haaglanden Medical Center, The Hague, The Netherlands
| | - Nathalie M Delfos
- Department of Internal Medicine, Alrijne Hospital, Leiderdorp, The Netherlands
| | | | - Femke P N Mollema
- Department of Internal Medicine, Haaglanden Medical Center, The Hague, The Netherlands
| | - Liesbeth B E Bosma
- Department of Hospital Pharmacy, Haga Teaching Hospital, Els Borst-Eilersplein 275, 2545 AA, The Hague, The Netherlands
| | - Loes E Visser
- Department of Hospital Pharmacy, Haga Teaching Hospital, Els Borst-Eilersplein 275, 2545 AA, The Hague, The Netherlands
- Department of Hospital Pharmacy, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Thomas H Ottens
- Department of Intensive Care, Haga Teaching Hospital, The Hague, The Netherlands
| | - Nathalie D van Burgel
- Department of Medical Microbiology, Haga Teaching Hospital, The Hague, The Netherlands
| | - Sesmu M Arbous
- Department of Intensive Care, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Rachel Knevel
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rolf H H Groenwold
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mark G J de Boer
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Leo G Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik B Wilms
- Department of Hospital Pharmacy, Haga Teaching Hospital, Els Borst-Eilersplein 275, 2545 AA, The Hague, The Netherlands
- Apotheek Haagse Ziekenhuizen, The Hague, The Netherlands
| | - Cees van Nieuwkoop
- Department of Internal Medicine, Haga Teaching Hospital, The Hague, The Netherlands
- Department of Public Health and Primary Care, Health Campus The Hague, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
133
|
Janoff EN, Brown ST, Belitskaya-Levy I, Curtis JL, Bonomo RA, Miller EK, Goldberg AM, Zehm L, Wills A, Hutchinson C, Dumont LJ, Gleason T, Shih MC, ADD Caitlin MS in CCTC website. Design of VA CoronavirUs Research and Efficacy Studies-1 (VA CURES-1): A double-blind, randomized placebo-controlled trial of COVID-19 convalescent plasma in hospitalized patients with early respiratory compromise. Contemp Clin Trials Commun 2023; 35:101190. [PMID: 37560085 PMCID: PMC10407261 DOI: 10.1016/j.conctc.2023.101190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 07/07/2023] [Accepted: 07/15/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Effective therapeutics for severe acute respiratory syndrome CoronaVirus-2 (SARS-CoV-2) infection are evolving. Under Emergency Use Authorization, COVID-19 convalescent plasma (CCP) was widely used in individuals hospitalized for COVID-19, but few randomized controlled trials supported its efficacy to limit respiratory failure or death. METHODS VA CoronavirUs Research and Efficacy Studies-1 (VA CURES-1) was a double-blind, multi-site, placebo-controlled, randomized clinical trial evaluating the efficacy and safety of CCP with conventional therapy in hospitalized Veterans with SARS-CoV-2 infection and early respiratory compromise (requirement for oxygen). Participants (planned sample size 702) were randomized 1:1 to receive CCP with high titer neutralizing activity or 0.9% saline, stratified by site and age (≥65 versus <65 years old). Participants were followed daily during initial hospitalization and at Days 15, 22 and 28. OUTCOMES The composite primary outcome was acute hypoxemic respiratory failure or all-cause death by Day 28. Secondary outcomes by day 28 included time-to-recovery, clinical severity, mortality, rehospitalization for COVID-19, and adverse events. Serial respiratory and blood samples were collected for safety, virologic and immunologic analyses and future studies. Key variables in predicting the success of CURES-1 were: (1) enrollment early in the course of severe infection; (2) use of plasma with high neutralizing activity; (3) reliance on unambiguous, clinically meaningful outcomes. CURES-1 was terminated for futility due to perceived inability to enroll in the lull between the Alpha and Delta waves of the SARS CoV-2 epidemic. CONCLUSIONS VA CURES-1 was a large multi-site trial designed to provide conclusive information about the efficacy of CCP in well-characterized patients at risk for progression of COVID-19. It utilized a rigorous study design with relevant initial timing, quality of product and outcomes. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04539275.
Collapse
Affiliation(s)
- Edward N. Janoff
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
- University of Colorado Denver School of Medicine, Aurora, CO, USA
| | - Sheldon T. Brown
- James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
- Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Ilana Belitskaya-Levy
- Department of Veterans Affairs, Cooperative Studies Program Coordinating Center, Palo Alto, CA, USA
| | - Jeffrey L. Curtis
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
- University of Michigan Medical School, Ann Arbor, MI, USA
| | - Robert A. Bonomo
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
- Case VA CARES, Case Western Reserve University School of Medicine, USA
| | - Elliott K. Miller
- Department of Veterans Affairs, Cooperative Studies Program Clinical Research Pharmacy Coordinating Center, Albuquerque, NM, USA
| | - Alexa M. Goldberg
- Department of Veterans Affairs, Cooperative Studies Program Clinical Research Pharmacy Coordinating Center, Albuquerque, NM, USA
| | - Lisa Zehm
- Department of Veterans Affairs, Cooperative Studies Program Coordinating Center, Palo Alto, CA, USA
| | - Ashlea Wills
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
| | | | - Larry J. Dumont
- University of Colorado Denver School of Medicine, Aurora, CO, USA
- Vitalant Research Institute, Denver, CO, USA
| | - Theresa Gleason
- Department of Veterans Affairs, Clinical Science Research and Development Service, Washington, DC, USA
| | - Mei-Chiung Shih
- Department of Veterans Affairs, Cooperative Studies Program Coordinating Center, Palo Alto, CA, USA
- Stanford University School of Medicine, Palo Alto, CA, USA
| | - ADD Caitlin MS in CCTC website
- Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
- University of Colorado Denver School of Medicine, Aurora, CO, USA
- James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, USA
- Icahn School of Medicine at Mt. Sinai, New York, NY, USA
- Department of Veterans Affairs, Cooperative Studies Program Coordinating Center, Palo Alto, CA, USA
- VA Ann Arbor Healthcare System, Ann Arbor, MI, USA
- University of Michigan Medical School, Ann Arbor, MI, USA
- Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
- Case VA CARES, Case Western Reserve University School of Medicine, USA
- Department of Veterans Affairs, Cooperative Studies Program Clinical Research Pharmacy Coordinating Center, Albuquerque, NM, USA
- Vitalant Research Institute, Denver, CO, USA
- Department of Veterans Affairs, Clinical Science Research and Development Service, Washington, DC, USA
- Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
134
|
Alrajhi NN. Post-COVID-19 pulmonary fibrosis: An ongoing concern. Ann Thorac Med 2023; 18:173-181. [PMID: 38058790 PMCID: PMC10697304 DOI: 10.4103/atm.atm_7_23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/05/2023] [Accepted: 07/10/2023] [Indexed: 12/08/2023] Open
Abstract
Coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 rapidly spread across the globe causing over 6 million deaths and major compromization of health facilities. The vast majority of survivors post-COVID-19 are left with variable degrees of health sequelae including pulmonary, neurological, psychological, and cardiovascular complications. Post-COVID-19 pulmonary fibrosis is one of the major concerns arising after the recovery from this pandemic. Risk factors for post-COVID-19 pulmonary fibrosis include age, male sex, and the severity of COVID-19 disease. High-resolution computed tomography provides diagnostic utility to diagnose pulmonary fibrosis as it provides more details regarding the pattern and the extent of pulmonary fibrosis. Emerging data showing similarities between post-COVID-19 pulmonary fibrosis and idiopathic pulmonary fibrosis, finding that needs further exploration. The management of post-COVID-19 pulmonary fibrosis depends on many factors but largely relies on excluding other causes of pulmonary fibrosis, the extent of fibrosis, and physiological impairment. Treatment includes immunosuppressants versus antifibrotics or both.
Collapse
Affiliation(s)
- Nuha Nasser Alrajhi
- Department of Medicine, Pulmonary Unit, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
135
|
Stricker M. Managing hospitalized patients with COVID-19. JAAPA 2023; 36:16-20. [PMID: 37751251 DOI: 10.1097/01.jaa.0000977664.94343.68] [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: 09/27/2023]
Abstract
ABSTRACT Treatment for COVID-19 has significantly changed since the beginning of the pandemic and continues to change as new evidence is published. This article describes which COVID-19 patients require hospitalization and how to manage hospitalized patients based on current evidence from randomized clinical trials.
Collapse
Affiliation(s)
- Mike Stricker
- Mike Stricker practices in hospital medicine at the Cleveland (Ohio) Clinic. The author has disclosed no potential conflicts of interest, financial or otherwise
| |
Collapse
|
136
|
Van Laethem J, Pierreux J, Wuyts SC, De Geyter D, Allard SD, Dauby N. Using risk factors and markers to predict bacterial respiratory co-/superinfections in COVID-19 patients: is the antibiotic steward's toolbox full or empty? Acta Clin Belg 2023; 78:418-430. [PMID: 36724448 DOI: 10.1080/17843286.2023.2167328] [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: 10/30/2022] [Accepted: 01/07/2023] [Indexed: 02/03/2023]
Abstract
BACKGROUND Adequate diagnosis of bacterial respiratory tract co-/superinfection (bRTI) in coronavirus disease (COVID-19) patients is challenging, as there is insufficient knowledge about the role of risk factors and (para)clinical parameters in the identification of bacterial co-/superinfection in the COVID-19 setting. Empirical antibiotic therapy is mainly based on COVID-19 severity and expert opinion, rather than on scientific evidence generated since the start of the pandemic. PURPOSE We report the best available evidence regarding the predictive value of risk factors and (para)clinical markers in the diagnosis of bRTI in COVID-19 patients. METHODS A multidisciplinary team identified different potential risk factors and (para)clinical predictors of bRTI in COVID-19 and formulated one or two research questions per topic. After a thorough literature search, research gaps were identified, and suggestions concerning further research were formulated. The quality of this narrative review was ensured by following the Scale for the Assessment of Narrative Review Articles. RESULTS Taking into account the scarcity of scientific evidence for markers and risk factors of bRTI in COVID-19 patients, to date, COVID-19 severity is the only parameter which can be associated with higher risk of developing bRTI. CONCLUSIONS Evidence on the usefulness of risk factors and (para)clinical factors as predictors of bRTI in COVID-19 patients is scarce. Robust studies are needed to optimise antibiotic prescribing and stewardship activities in the context of COVID-19.
Collapse
Affiliation(s)
- Johan Van Laethem
- Department of Internal Medicine and Infectious Diseases, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Jan Pierreux
- Department of Internal Medicine and Infectious Diseases, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Stephanie Cm Wuyts
- Universitair Ziekenhuis Brussel (UZ Brussel), Hospital Pharmacy, Brussels, Belgium
- Research group Clinical Pharmacology and Pharmacotherapy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Deborah De Geyter
- Microbiology and Infection Control Department, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Sabine D Allard
- Department of Internal Medicine and Infectious Diseases, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Nicolas Dauby
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Centre for Environmental Health and Occupational Health, School of Public Health, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Infectious Diseases, CHU Saint-Pierre - Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
137
|
Neupane M, Kadri SS. Nangibotide for precision immunomodulation in septic shock and COVID-19. THE LANCET. RESPIRATORY MEDICINE 2023; 11:855-857. [PMID: 37269869 DOI: 10.1016/s2213-2600(23)00220-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/05/2023]
Affiliation(s)
- Maniraj Neupane
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD, USA; Critical Care Medicine Branch, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA
| | - Sameer S Kadri
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD, USA; Critical Care Medicine Branch, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
138
|
Abstract
COVID-19, the illness caused by SARS-CoV-2, became a worldwide pandemic in 2020. Initial clinical manifestations range from asymptomatic infection to mild upper respiratory illness but may progress to pulmonary involvement with hypoxemia and, in some cases, multiorgan involvement, shock, and death. Older adults, pregnant persons, those with common comorbidities, and those with immunosuppression are at greatest risk for progression. Vaccination is effective in preventing symptomatic infection and reducing risk for severe disease, hospitalization, and death. Antiviral treatment and immunomodulators have been shown to benefit certain patients. This article summarizes current recommendations on prevention, diagnosis, management, and treatment of COVID-19.
Collapse
Affiliation(s)
| | - Roy M Gulick
- Weill Cornell Medicine, New York, New York (K.M.M., R.M.G.)
| |
Collapse
|
139
|
Patanwala AE, Jager NGL, Radosevich JJ, Brüggemann R. An update on drug-drug interactions for care of the acutely ill in the era of COVID-19. Am J Health Syst Pharm 2023; 80:1301-1308. [PMID: 37368815 PMCID: PMC10516707 DOI: 10.1093/ajhp/zxad152] [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: 06/25/2023] [Indexed: 06/29/2023] Open
Abstract
PURPOSE To provide key pharmacological concepts underlying drug-drug interactions (DDIs), a decision-making framework, and a list of DDIs that should be considered in the context of contemporary acutely ill patients with COVID-19. SUMMARY DDIs are frequently encountered in the acutely ill. The implications of DDIs include either increased risk of drug toxicity or decreased effectiveness, which may have severe consequences in the acutely ill due to lower physiological and neurocognitive reserves in these patients. In addition, an array of additional therapies and drug classes have been used for COVID-19 that were not typically used in the acute care setting. In this update on DDIs in the acutely ill, we provide key pharmacological concepts underlying DDIs, including a discussion of the gastric environment, the cytochrome P-450 (CYP) isozyme system, transporters, and pharmacodynamics in relation to DDIs. We also provide a decision-making framework that elucidates the identification of DDIs, risk assessment, selection of alternative therapies, and monitoring. Finally, important DDIs pertaining to contemporary acute care clinical practice related to COVID-19 are discussed. CONCLUSION Interpreting and managing DDIs should follow a pharmacologically based approach and a systematic decision-making process to optimize patient outcomes.
Collapse
Affiliation(s)
- Asad E Patanwala
- Faculty of Medicine and Health, School of Pharmacy, The University of Sydney, Camperdown, New South Wales, and Department of Pharmacy, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Nynke G L Jager
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, and Radboudumc Institute for Health Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - John J Radosevich
- Department of Pharmacy Services, Dignity Health–St. Joseph’s Hospital & Medical Center, Phoenix, AZ, USA
| | - Roger Brüggemann
- Department of Pharmacy, Radboud University Medical Center, Nijmegen, and Radboudumc Institute for Health Sciences Center of Expertise in Mycology Radboudumc/CWZ, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
140
|
Madi K, Flumian C, Olivier P, Sommet A, Montastruc F. Quality of reporting of adverse events in clinical trials of covid-19 drugs: systematic review. BMJ MEDICINE 2023; 2:e000352. [PMID: 37779893 PMCID: PMC10537984 DOI: 10.1136/bmjmed-2022-000352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 06/27/2023] [Indexed: 10/03/2023]
Abstract
Objective To assess the quality of reporting of adverse events in clinical trials of covid-19 drugs based on the CONSORT (Consolidated Standards of Reporting Trials) harms extension and according to clinical trial design, and to examine reporting of serious adverse events in drug trials published on PubMed versus clinical trial summaries on ClinicalTrials.gov. Design Systematic review. Data sources PubMed and ClinicalTrials.gov registries were searched from 1 December 2019 to 17 February 2022. Eligibility criteria for selecting studies Randomised clinical trials evaluating the efficacy and safety of drugs used to treat covid-19 disease in participants of all ages with suspected, probable, or confirmed SARS-CoV-2 infection were included. Clinical trials were screened on title, abstract, and text by two authors independently. Only articles published in French and English were selected. The Cochrane risk of bias tool for randomised trials (RoB 2) was used to assess risk of bias. Results The search strategy identified 1962 randomised clinical trials assessing the efficacy and safety of drugs used to treat covid-19, published in the PubMed database; 1906 articles were excluded after screening and 56 clinical trials were included in the review. Among the 56 clinical trials, no study had a high score for quality of reporting of adverse events, 60.7% had a moderate score, 33.9% had a low score, and 5.4% had a very low score. All clinical trials with a very low score for quality of reporting of adverse events were randomised open label trials. For reporting of serious adverse events, journal articles published on PubMed under-reported 51% of serious adverse events compared with clinical trial summaries published on ClinicalTrials.gov. Conclusions In one in three published clinical trials on covid-19 drugs, the quality of reporting of adverse events was low or very low. Differences were found in the number of serious adverse events reported in journal articles versus clinical trial summaries. During the covid-19 pandemic, risk assessment of drugs in clinical trials of covid-19 drugs did not comply with good practice recommendations for publication of results. Systematic review registration European Network of Centres for Pharmacoepidemiology and Pharmacovigilance (ENCePP) EUPAS45959.
Collapse
Affiliation(s)
- Karima Madi
- CIC 1436, Team PEPSS (Pharmacologie En Population cohorteS et biobanqueS), Toulouse University Hospital, Toulouse, France
| | - Clara Flumian
- CIC 1436, Team PEPSS (Pharmacologie En Population cohorteS et biobanqueS), Toulouse University Hospital, Toulouse, France
- Department of Medical and Clinical Pharmacology, Centre of PharmacoVigilance and Pharmacoepidemiology, Faculty of Medicine, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Pascale Olivier
- Department of Medical and Clinical Pharmacology, Centre of PharmacoVigilance and Pharmacoepidemiology, Faculty of Medicine, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Agnès Sommet
- CIC 1436, Team PEPSS (Pharmacologie En Population cohorteS et biobanqueS), Toulouse University Hospital, Toulouse, France
- Department of Medical and Clinical Pharmacology, Centre of PharmacoVigilance and Pharmacoepidemiology, Faculty of Medicine, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - François Montastruc
- CIC 1436, Team PEPSS (Pharmacologie En Population cohorteS et biobanqueS), Toulouse University Hospital, Toulouse, France
- Department of Medical and Clinical Pharmacology, Centre of PharmacoVigilance and Pharmacoepidemiology, Faculty of Medicine, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| |
Collapse
|
141
|
Verra C, Mohammad S, Alves GF, Porchietto E, Coldewey SM, Collino M, Thiemermann C. Baricitinib protects mice from sepsis-induced cardiac dysfunction and multiple-organ failure. Front Immunol 2023; 14:1223014. [PMID: 37781388 PMCID: PMC10536262 DOI: 10.3389/fimmu.2023.1223014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/07/2023] [Indexed: 10/03/2023] Open
Abstract
Sepsis is one of the major complications of surgery resulting in high morbidity and mortality, but there are no specific therapies for sepsis-induced organ dysfunction. Data obtained under Gene Expression Omnibus accession GSE131761 were re-analyzed and showed an increased gene expression of Janus Kinase 2 (JAK2) and Signal Transducer and Activator of Transcription 3 (STAT3) in the whole blood of post-operative septic patients. Based on these results, we hypothesized that JAK/STAT activation may contribute to the pathophysiology of septic shock and, hence, investigated the effects of baricitinib (JAK1/JAK2 inhibitor) on sepsis-induced cardiac dysfunction and multiple-organ failure (MOF). In a mouse model of post-trauma sepsis induced by midline laparotomy and cecal ligation and puncture (CLP), 10-week-old male (n=32) and female (n=32) C57BL/6 mice received baricitinib (1mg/kg; i.p.) or vehicle at 1h or 3h post-surgery. Cardiac function was assessed at 24h post-CLP by echocardiography in vivo, and the degree of MOF was analyzed by determination of biomarkers in the serum. The potential mechanism underlying both the cardiac dysfunction and the effect of baricitinib was analyzed by western blot analysis in the heart. Trauma and subsequent sepsis significantly depressed the cardiac function and induced multiple-organ failure, associated with an increase in the activation of JAK2/STAT3, NLRP3 inflammasome and NF- κβ pathways in the heart of both male and female animals. These pathways were inhibited by the administration of baricitinib post the onset of sepsis. Moreover, treatment with baricitinib at 1h or 3h post-CLP protected mice from sepsis-induced cardiac injury and multiple-organ failure. Thus, baricitinib may be repurposed for trauma-associated sepsis.
Collapse
Affiliation(s)
- Chiara Verra
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Shireen Mohammad
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | - Elisa Porchietto
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Sina Maren Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Massimo Collino
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Christoph Thiemermann
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
142
|
Gonzaga A, Andreu E, Hernández-Blasco LM, Meseguer R, Al-Akioui-Sanz K, Soria-Juan B, Sanjuan-Gimenez JC, Ferreras C, Tejedo JR, Lopez-Lluch G, Goterris R, Maciá L, Sempere-Ortells JM, Hmadcha A, Borobia A, Vicario JL, Bonora A, Aguilar-Gallardo C, Poveda JL, Arbona C, Alenda C, Tarín F, Marco FM, Merino E, Jaime F, Ferreres J, Figueira JC, Cañada-Illana C, Querol S, Guerreiro M, Eguizabal C, Martín-Quirós A, Robles-Marhuenda Á, Pérez-Martínez A, Solano C, Soria B. Rationale for combined therapies in severe-to-critical COVID-19 patients. Front Immunol 2023; 14:1232472. [PMID: 37767093 PMCID: PMC10520558 DOI: 10.3389/fimmu.2023.1232472] [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: 05/31/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
An unprecedented global social and economic impact as well as a significant number of fatalities have been brought on by the coronavirus disease 2019 (COVID-19), produced by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Acute SARS-CoV-2 infection can, in certain situations, cause immunological abnormalities, leading to an anomalous innate and adaptive immune response. While most patients only experience mild symptoms and recover without the need for mechanical ventilation, a substantial percentage of those who are affected develop severe respiratory illness, which can be fatal. The absence of effective therapies when disease progresses to a very severe condition coupled with the incomplete understanding of COVID-19's pathogenesis triggers the need to develop innovative therapeutic approaches for patients at high risk of mortality. As a result, we investigate the potential contribution of promising combinatorial cell therapy to prevent death in critical patients.
Collapse
Affiliation(s)
- Aitor Gonzaga
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Institute of Bioengineering, Miguel Hernández University, Elche, Spain
| | - Etelvina Andreu
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Applied Physics Department, Miguel Hernández University, Elche, Spain
| | | | - Rut Meseguer
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Clinic University Hospital, Fundación para la Investigación del Hospital Clínico de la Comunidad Valenciana (INCLIVA) Health Research Institute, Valencia, Spain
| | - Karima Al-Akioui-Sanz
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Bárbara Soria-Juan
- Réseau Hospitalier Neuchâtelois, Hôpital Pourtalès, Neuchâtel, Switzerland
| | | | - Cristina Ferreras
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
| | - Juan R. Tejedo
- Department of Molecular Biology and Biochemical Engineering, University Pablo de Olavide, Seville, Spain
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) of the Carlos III Health Institute (ISCIII), Madrid, Spain
| | - Guillermo Lopez-Lluch
- University Pablo de Olavide, Centro Andaluz de Biología del Desarrollo - Consejo Superior de Investigaciones Científicas (CABD-CSIC), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Sevilla, Spain
| | - Rosa Goterris
- Clinic University Hospital, Fundación para la Investigación del Hospital Clínico de la Comunidad Valenciana (INCLIVA) Health Research Institute, Valencia, Spain
| | - Loreto Maciá
- Nursing Department, University of Alicante, Alicante, Spain
| | - Jose M. Sempere-Ortells
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Biotechnology Department, University of Alicante, Alicante, Spain
| | - Abdelkrim Hmadcha
- Department of Molecular Biology and Biochemical Engineering, University Pablo de Olavide, Seville, Spain
- Biosanitary Research Institute (IIB-VIU), Valencian International University (VIU), Valencia, Spain
| | - Alberto Borobia
- Clinical Pharmacology Department, La Paz University Hospital, School of Medicine, Universidad Autónoma de Madrid, IdiPAz, Madrid, Spain
| | - Jose L. Vicario
- Transfusion Center of the Autonomous Community of Madrid, Madrid, Spain
| | - Ana Bonora
- Health Research Institute Hospital La Fe, Valencia, Spain
| | | | - Jose L. Poveda
- Health Research Institute Hospital La Fe, Valencia, Spain
| | - Cristina Arbona
- Valencian Community Blood Transfusion Center, Valencia, Spain
| | - Cristina Alenda
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Fabian Tarín
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Francisco M. Marco
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Immunology Department, Dr. Balmis General University Hospital, Alicante, Spain
| | - Esperanza Merino
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Department of Clinical Medicine, Miguel Hernández University, Elche, Spain
- Infectious Diseases Unit, Dr. Balmis General University Hospital, Alicante, Spain
| | - Francisco Jaime
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - José Ferreres
- Intensive Care Service, Hospital Clinico Universitario, Fundación para la Investigación del Hospital Clínico de la Comunidad Valenciana (INCLIVA), Valencia, Spain
| | | | | | | | - Manuel Guerreiro
- Department of Hematology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Cristina Eguizabal
- Research Unit, Basque Center for Blood Transfusion and Human Tissues, Galdakao, Spain
- Cell Therapy, Stem Cells and Tissues Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | | | | | - Antonio Pérez-Martínez
- Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, Madrid, Spain
- Department of Pediatrics, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Carlos Solano
- Hematology Service, Hospital Clínico Universitario, Fundación para la Investigación del Hospital Clínico de la Comunidad Valenciana (INCLIVA), Valencia, Spain
| | - Bernat Soria
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Institute of Bioengineering, Miguel Hernández University, Elche, Spain
- Biomedical Research Network for Diabetes and Related Metabolic Diseases-Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) of the Carlos III Health Institute (ISCIII), Madrid, Spain
| |
Collapse
|
143
|
Miot HA, Criado PR, de Castro CCS, Ianhez M, Talhari C, Ramos PM. JAK-STAT pathway inhibitors in dermatology. An Bras Dermatol 2023; 98:656-677. [PMID: 37230920 PMCID: PMC10404561 DOI: 10.1016/j.abd.2023.03.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/26/2023] [Accepted: 03/02/2023] [Indexed: 05/27/2023] Open
Abstract
The JAK-STAT signaling pathway mediates important cellular processes such as immune response, carcinogenesis, cell differentiation, division and death. Therefore, drugs that interfere with different JAK-STAT signaling patterns have potential indications for various medical conditions. The main dermatological targets of JAK-STAT pathway inhibitors are inflammatory or autoimmune diseases such as psoriasis, vitiligo, atopic dermatitis and alopecia areata; however, several dermatoses are under investigation to expand this list of indications. As JAK-STAT pathway inhibitors should gradually occupy a relevant space in dermatological prescriptions, this review presents the main available drugs, their immunological effects, and their pharmacological characteristics, related to clinical efficacy and safety, aiming to validate the best dermatological practice.
Collapse
Affiliation(s)
- Hélio Amante Miot
- Department of Dermatology, Faculty of Medicine, Universidade Estadual Paulista, Botucatu, SP, Brazil.
| | - Paulo Ricardo Criado
- Centro Universitário Faculdade de Medicina do ABC, Santo André, SP, Brazil; Faculdade de Ciências Médicas de Santos, Santos, SP, Brazil
| | - Caio César Silva de Castro
- Hospital de Dermatologia Sanitária do Paraná, Curitiba, PR, Brazil; Escola de Medicina, Pontifícia Universidade Católica do Paraná, Curitiba, PR, Brazil
| | - Mayra Ianhez
- Department of Tropical Medicine and Dermatology, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Carolina Talhari
- Department of Dermatology, Universidade do Estado do Amazonas, Manaus, AM, Brazil
| | - Paulo Müller Ramos
- Department of Dermatology, Faculty of Medicine, Universidade Estadual Paulista, Botucatu, SP, Brazil
| |
Collapse
|
144
|
López V, Mazuecos A, Villanego F, López-Oliva M, Alonso A, Beneyto I, Crespo M, Díaz-Corte C, Franco A, González-Roncero F, Guirado L, Jiménez C, Juega J, Llorente S, Paul J, Rodríguez-Benot A, Ruiz JC, Sánchez-Fructuoso A, Torregrosa V, Zárraga S, Rodrigo E, Hernández D. Update of the recommendations on the management of the SARS-CoV-2 coronavirus pandemic (COVID-19) in kidney transplant patients. Nefrologia 2023; 43:531-545. [PMID: 37957107 DOI: 10.1016/j.nefroe.2023.10.007] [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: 10/10/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2023] Open
Abstract
SARS-CoV-2 infection (COVID-19) has had a significant impact on transplant activity in our country. Mortality and the risk of complications associated with COVID-19 in kidney transplant recipients (KT) were expected to be higher due to their immunosuppressed condition and the frequent associated comorbidities. Since the beginning of the pandemic in March 2020 we have rapidly improved our knowledge about the epidemiology, clinical features and management of COVID-19 post-transplant, resulting in a better prognosis for our patients. KT units have been able to adapt their programs to this new reality, normalizing both donation and transplantation activity in our country. This manuscript presents a proposal to update the general recommendations for the prevention and treatment of infection in this highly vulnerable population such as KT.
Collapse
Affiliation(s)
- Verónica López
- Unidad de Gestión Clínica de Nefrología, Hospital Regional Universitario de Málaga, Universidad de Málaga, Instituto Biomédico de Investigación de Málaga (IBIMA), RICORS2040 (RD21/0005/0012), Málaga, Spain.
| | | | | | | | - Angel Alonso
- Servicio de Nefrología, Complejo Hospitalario A Coruña, A Coruña, Spain
| | - Isabel Beneyto
- Servicio de Nefrología, Hospital Universitario Politécnico La Fe, Valencia, Spain
| | - Marta Crespo
- Servicio de Nefrología, Hospital del Mar, Hospital del Mar Medical Research Institute (IMIM), RD16/0009/0013 (ISCIII FEDER REDinREN), Barcelona, Spain
| | - Carmen Díaz-Corte
- Servicio de Nefrología, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Antonio Franco
- Servicio de Nefrología, Hospital de Alicante, Alicante, Spain
| | | | - Luis Guirado
- Servicio de Nefrología, Fundación Puigvert, REDinREN RD16/0009/0019, Barcelona, Spain
| | | | - Javier Juega
- Servicio de Nefrología, Hospital Trias i Pujol, REDinREN RD16/0009/0032, Barcelona, Spain
| | - Santiago Llorente
- Servicio de Nefrología, Hospital Virgen de la Arrixaca, Murcia, Spain
| | - Javier Paul
- Servicio de Nefrología, Hospital Miguel Servet, Zaragoza, Spain
| | - Alberto Rodríguez-Benot
- Servicio de Nefrología, Hospital Reina Sofía, Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain
| | - Juan Carlos Ruiz
- Servicio de Nefrología, Hospital Marqués de Valdecilla, IDIVAL, REDinREN RD16/0009/0027, Santander, Cantabria, Spain
| | - Ana Sánchez-Fructuoso
- Servicio de Nefrología, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Sofía Zárraga
- Servicio de Nefrología, Hospital de Cruces, Bilbao, Vizcaya, Spain
| | - Emilio Rodrigo
- Servicio de Nefrología, Hospital Marqués de Valdecilla, IDIVAL, REDinREN RD16/0009/0027, Santander, Cantabria, Spain
| | - Domingo Hernández
- Unidad de Gestión Clínica de Nefrología, Hospital Regional Universitario de Málaga, Universidad de Málaga, Instituto Biomédico de Investigación de Málaga (IBIMA), RICORS2040 (RD21/0005/0012), Málaga, Spain
| |
Collapse
|
145
|
DE Vito A, Saderi L, Fiore V, Geremia N, Princic E, Fanelli C, Muredda AA, Panu Napodano C, Moi G, Maida I, Fois AG, Sotgiu G, Madeddu G, Babudieri S. Early treatment with low-molecular-weight heparin reduces mortality rate in SARS-CoV-2 patients. Panminerva Med 2023; 65:286-291. [PMID: 35622392 DOI: 10.23736/s0031-0808.22.04572-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Since the beginning of the SARS-CoV-2 pandemic, millions of people have been infected and died. Different therapeutic approaches have been recommended, but only a few have shown clinical advantages. Low-molecular-weight heparin (LMWH) has been recommended to prevent COVID-19-related thrombo-embolic events. We aimed to evaluate the impact of early treatment with LMWH on hospital admission and death in patients with SARS-CoV-2 infection. METHODS We conducted an observational monocentric retrospective study to evaluate the preventive role of LMWH on the mortality rate of COVID-19 patients. SARS-CoV-2 infected patients were recruited from the beginning of the Italian epidemic to March 31, 2021. We excluded patients with missing data and those chronically exposed to LMWH. Treatment prescription was based on international and national guidelines and modified depending on clinical presentation and drug-drug interactions. RESULTS Seven hundred thirty-four SARS-CoV-2 infected patients were recruited, with 357 (48.6%) males and a median (IQR) age of 77.9 (65-85.7) years. 77.5% of people developed SARS-CoV-2-related symptoms and 62.8% were admitted to the hospital, and 20.2% died. Four hundred ninety-two (67%) started LMWH. In particular, 296 (40.3%) were treated within five days since symptoms onset. At logistic regression, early LMWH therapy was associated with lower mortality. Furthermore, remdesivir treatment showed a lower risk of death. On the contrary, age, BMI>30 kg/m2, neurological diseases, fever or dyspnea were associated with an increased risk of death. CONCLUSIONS Early treatment with LMWH was associated with lower mortality in our cohort. Further studies are needed to better assess the role of wider LMWH administration in terms of timing and regimen dose.
Collapse
Affiliation(s)
- Andrea DE Vito
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy -
| | - Laura Saderi
- Unit of Clinical Epidemiology and Medical Statistics, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Vito Fiore
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Nicholas Geremia
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Elija Princic
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Chiara Fanelli
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Alberto A Muredda
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Catello Panu Napodano
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giulia Moi
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Ivana Maida
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Alessandro G Fois
- Unit of Respiratory Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giovanni Sotgiu
- Unit of Clinical Epidemiology and Medical Statistics, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giordano Madeddu
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Sergio Babudieri
- Unit of Infectious Diseases, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
146
|
Leaf DE, Gordon AC, Lawler PR. Adverse Effects of Tocilizumab Versus Baricitinib in Severe COVID-19. Crit Care Med 2023; 51:e184-e185. [PMID: 37589523 DOI: 10.1097/ccm.0000000000005933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Affiliation(s)
- David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA
| | - Anthony C Gordon
- Division of Anaesthetics, Pain Medicine and Intensive Care, Imperial College London, London, United Kingdom
| | - Patrick R Lawler
- Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
- Department of Medicine, Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, ON, Canada
| |
Collapse
|
147
|
Palmer ME, Belcher RM, Engeleit A, Wenzler E, Bulman ZP, Benken ST. Pharmacokinetics and dialytic clearance of baricitinib during in vivo continuous venovenous haemodialysis in a patient with COVID-19. Int J Antimicrob Agents 2023; 62:106920. [PMID: 37442487 DOI: 10.1016/j.ijantimicag.2023.106920] [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: 04/16/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023]
Abstract
OBJECTIVES To date, there are no published pharmacokinetic (PK) data for baricitinib in critically ill patients requiring continuous renal replacement therapy. This paper describes in detail the plasma PK and dialytic clearance of baricitinib in a patient infected with coronavirus disease 2019 (COVID-19) requiring continuous renal replacement therapy in order to suggest dosing strategies in this population. METHODS Baricitinib 2 mg daily was used for the treatment of COVID-19 in a critically ill patient on continuous venovenous haemodialysis (CVVHD). Prefiltration plasma drug concentrations of baricitinib were measured at hours 1, 2, 12, and 24 after drug administration. Postfiltration and ultrafiltrate concentrations were collected at hour 24. RESULTS Plasma PK parameters of baricitinib in this patient were as follows: maximum plasma concentration (Cmax), 20.98 ng/mL; minimum plasma concentration (Cmin), 9.84 ng/mL; half-life (t1/2), 23.85 h; apparent volume of distribution at the steady state (Vss), 99.42 L; total clearance at the steady state (CLss), 2.89 L/h; and area under the concentration-time curve (AUC0-∞), 692.14 ng · h/mL. The saturation coefficient for baricitinib at 24 h after administration was 0.607. The transmembrane clearance of baricitinib by CVVHD running at a flow rate of 2 L/h was 1.21 L/h, representing 41.9% of the total clearance of baricitinib. CONCLUSIONS In a critically ill COVID-19 patient on CVVHD, a 2-mg dose of baricitinib achieves a Cmax comparable with healthy subjects, but total clearance was reduced to about 20%. Larger studies exploring multiple patients and dialysis modes are needed to determine the optimal dosing strategy for baricitinib in this patient population.
Collapse
Affiliation(s)
- Mary E Palmer
- Department of Pharmacy Practice, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois; University of Cincinnati Health, Department of Pharmacy Services, Cincinnati, Ohio
| | - Rachel M Belcher
- Department of Pharmacy Practice, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois
| | - Anastasia Engeleit
- Department of Pharmacy Practice, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois; University of Florida Shands Hospital, Department of Pharmacy, Gainesville, Florida
| | - Eric Wenzler
- Department of Pharmacy Practice, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois; Roche Pharma, Little Falls, New Jersey
| | - Zackery P Bulman
- Department of Pharmacy Practice, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois
| | - Scott T Benken
- Department of Pharmacy Practice, University of Illinois at Chicago College of Pharmacy, Chicago, Illinois.
| |
Collapse
|
148
|
Ahmed IS, Tapponi SL, Widatallah ME, Alakkad YM, Haider M. Unmasking the enigma: An in-depth analysis of COVID-19 impact on the pediatric population. J Infect Public Health 2023; 16:1346-1360. [PMID: 37433256 PMCID: PMC10299956 DOI: 10.1016/j.jiph.2023.06.017] [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: 05/26/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/13/2023] Open
Abstract
OBJECTIVES COVID-19, caused by the novel coronavirus, has had a profound and wide-reaching impact on individuals of all age groups across the globe, including children. This review article aims to provide a comprehensive analysis of COVID-19 in children, covering essential topics such as epidemiology, transmission, pathogenesis, clinical features, risk factors, diagnosis, treatment, vaccination, and others. By delving into the current understanding of the disease and addressing the challenges that lie ahead, this article seeks to shed light on the unique considerations surrounding COVID-19 in children and contribute to a deeper comprehension of this global health crisis affecting our youngest population. METHODS A comprehensive literature search was conducted to gather the most recent and relevant information regarding COVID-19 in children. Multiple renowned databases, including MEDLINE, PubMed, Scopus, as well as authoritative sources such as the World Health Organization (WHO), the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMA), and the National Institutes of Health (NIH) websites and others were thoroughly searched. The search included articles, guidelines, reports, clinical trials results and expert opinions published within the past three years, ensuring the inclusion of the latest research findings on COVID-19 in children. Several relevant keywords, including "COVID-19," "SARS-CoV-2," "children," "pediatrics," and related terms were used to maximize the scope of the search and retrieve a comprehensive set of articles. RESULTS AND CONCLUSION Three years since the onset of the COVID-19 pandemic, our understanding of its impact on children has evolved, but many questions remain unanswered. While SAR-CoV-2 generally leads to mild illness in children, the occurrence of severe cases and the potential for long-term effects cannot be overlooked. Efforts to comprehensively study COVID-19 in children must continue to improve preventive strategies, identify high-risk populations, and ensure optimal management. By unraveling the enigma surrounding COVID-19 in children, we can strive towards safeguarding their health and well-being in the face of future global health challenges.
Collapse
Affiliation(s)
- Iman Saad Ahmed
- Department of Pharmaceutics & Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates.
| | - Sara Luay Tapponi
- Department of Pharmaceutics & Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Marwa Eltahir Widatallah
- Department of Pharmaceutics & Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Yumna Mohamed Alakkad
- Department of Pharmaceutics & Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Mohamed Haider
- Department of Pharmaceutics & Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
149
|
Fukihara J, Kondoh Y. COVID-19 and interstitial lung diseases: A multifaceted look at the relationship between the two diseases. Respir Investig 2023; 61:601-617. [PMID: 37429073 PMCID: PMC10281233 DOI: 10.1016/j.resinv.2023.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/09/2023] [Accepted: 05/22/2023] [Indexed: 07/12/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although it has been a fatal disease for many patients, the development of treatment strategies and vaccines have progressed over the past 3 years, and our society has become able to accept COVID-19 as a manageable common disease. However, as COVID-19 sometimes causes pneumonia, post-COVID pulmonary fibrosis (PCPF), and worsening of preexisting interstitial lung diseases (ILDs), it is still a concern for pulmonary physicians. In this review, we have selected several topics regarding the relationships between ILDs and COVID-19. The pathogenesis of COVID-19-induced ILD is currently assumed based mainly on the evidence of other ILDs and has not been well elucidated specifically in the context of COVID-19. We have summarized what has been clarified to date and constructed a coherent story about the establishment and progress of the disease. We have also reviewed clinical information regarding ILDs newly induced or worsened by COVID-19 or anti-SARS-CoV-2 vaccines. Inflammatory and profibrotic responses induced by COVID-19 or vaccines have been thought to be a risk for de novo induction or worsening of ILDs, and this has been supported by the evidence obtained through clinical experience over the past 3 years. Although COVID-19 has become a mild disease in most cases, it is still worth looking back on the above-reviewed information to broaden our perspectives regarding the relationship between viral infection and ILD. As a representative etiology for severe viral pneumonia, further studies in this area are expected.
Collapse
Affiliation(s)
- Jun Fukihara
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, 160 Nishioiwake-cho, Seto, Aichi, 489-8642, Japan
| | - Yasuhiro Kondoh
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, 160 Nishioiwake-cho, Seto, Aichi, 489-8642, Japan.
| |
Collapse
|
150
|
de Brabander J, Boers LS, Kullberg RFJ, Zhang S, Nossent EJ, Heunks LMA, Vlaar APJ, Bonta PI, Schultz MJ, van der Poll T, Duitman J, Bos LDJ. Persistent alveolar inflammatory response in critically ill patients with COVID-19 is associated with mortality. Thorax 2023; 78:912-921. [PMID: 37142421 DOI: 10.1136/thorax-2023-219989] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/29/2023] [Indexed: 05/06/2023]
Abstract
INTRODUCTION Patients with COVID-19-related acute respiratory distress syndrome (ARDS) show limited systemic hyperinflammation, but immunomodulatory treatments are effective. Little is known about the inflammatory response in the lungs and if this could be targeted using high-dose steroids (HDS). We aimed to characterise the alveolar immune response in patients with COVID-19-related ARDS, to determine its association with mortality, and to explore the association between HDS treatment and the alveolar immune response. METHODS In this observational cohort study, a comprehensive panel of 63 biomarkers was measured in repeated bronchoalveolar lavage (BAL) fluid and plasma samples of patients with COVID-19 ARDS. Differences in alveolar-plasma concentrations were determined to characterise the alveolar inflammatory response. Joint modelling was performed to assess the longitudinal changes in alveolar biomarker concentrations, and the association between changes in alveolar biomarker concentrations and mortality. Changes in alveolar biomarker concentrations were compared between HDS-treated and matched untreated patients. RESULTS 284 BAL fluid and paired plasma samples of 154 patients with COVID-19 were analysed. 13 biomarkers indicative of innate immune activation showed alveolar rather than systemic inflammation. A longitudinal increase in the alveolar concentration of several innate immune markers, including CC motif ligand (CCL)20 and CXC motif ligand (CXCL)1, was associated with increased mortality. Treatment with HDS was associated with a subsequent decrease in alveolar CCL20 and CXCL1 levels. CONCLUSIONS Patients with COVID-19-related ARDS showed an alveolar inflammatory state related to the innate host response, which was associated with a higher mortality. HDS treatment was associated with decreasing alveolar concentrations of CCL20 and CXCL1.
Collapse
Affiliation(s)
- Justin de Brabander
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Leonoor S Boers
- Intensive Care Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology (LEICA), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Robert F J Kullberg
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Shiqi Zhang
- Intensive Care Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Esther J Nossent
- Pulmonary Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Leo M A Heunks
- Intensive Care Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Alexander P J Vlaar
- Intensive Care Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology (LEICA), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Peter I Bonta
- Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Marcus J Schultz
- Intensive Care Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology (LEICA), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Infection & Immunity, Inflammatory Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - JanWillem Duitman
- Pulmonary Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Infection & Immunity, Inflammatory Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Experimental Immunology (EXIM), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Lieuwe D J Bos
- Intensive Care Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Intensive Care and Anesthesiology (LEICA), Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|