1
|
Beaulieu M, Gaymard A, Massonnaud C, Peiffer-Smadja N, Bouscambert-Duchamp M, Carcelain G, Lingas G, Mentré F, Ader F, Hites M, Poignard P, Guedj J. Antiviral effect of Evusheld in COVID-19 hospitalized patients infected with pre-Omicron or Omicron variants: a modelling analysis of the randomized DisCoVeRy trial. J Antimicrob Chemother 2024; 79:2887-2895. [PMID: 39236218 PMCID: PMC11531825 DOI: 10.1093/jac/dkae301] [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: 07/10/2024] [Accepted: 08/09/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND The antiviral efficacy of Evusheld (AZD7442) in patients hospitalized for SARS-CoV-2 is unknown. METHODS We analysed the evolution of both the nasopharyngeal viral load and the serum neutralization activity against the variant of infection in 199 hospitalized patients (109 treated with Evusheld, 90 treated with placebo) infected with the SARS-CoV-2 virus and included in the randomized, double-blind, trial DisCoVeRy (NCT04315948). Using a mechanistic mathematical model, we reconstructed the trajectories of viral kinetics and how they are modulated by the increase in serum neutralization activity during Evusheld treatment. RESULTS Our model identified that the neutralization activity was associated with viral kinetics. Reflecting the variant-dependent neutralization activity of Evusheld, the antiviral activity of Evusheld was larger in patients infected with pre-Omicron or Omicron BA.2 variants than in patients infected with Omicron BA.1 variant. More specifically, the model predicted that Evusheld reduced the median time to viral clearance compared with placebo-treated patients by more than 5 days in patients infected by pre-Omicron (median: 5.9; 80% PI: 2.1-13.6) or Omicron BA.2 (median: 5.4; 80% PI: 2.0-12.4), respectively. The effect was more modest in patients infected by the Omicron BA.1 variant, reducing the median time to viral clearance by 2 days (median: 2.2; 80% PI: 0.4-8.9). CONCLUSIONS Hospitalized patients treated with Evusheld had a shorter median time to SARS-CoV-2 viral clearance. As Evusheld antiviral activity is mediated by the level of neutralization activity, its impact on viral clearance varies largely according to the variant of infection.
Collapse
Affiliation(s)
- Maxime Beaulieu
- Université Paris Cité et Université Sorbonne Paris Nord, Inserm, IAME, F-75018 Paris, France
| | - Alexandre Gaymard
- Hospices Civils de Lyon, Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des virus respiratoires France Sud, F-69317 Lyon, France
- Université Claude Bernard Lyon 1, Virpath, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, F69372 Lyon, France
| | - Clément Massonnaud
- Université Paris Cité et Université Sorbonne Paris Nord, Inserm, IAME, F-75018 Paris, France
- Département d’Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, F75018 Paris, France
| | - Nathan Peiffer-Smadja
- Université Paris Cité et Université Sorbonne Paris Nord, Inserm, IAME, F-75018 Paris, France
- AP-HP, Hôpital Bichat, Service de Maladies Infectieuses et Tropicales, F-75018 Paris, France
- National Institute for Health Research, Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Imperial College London, London, UK
| | - Maude Bouscambert-Duchamp
- Hospices Civils de Lyon, Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des virus respiratoires France Sud, F-69317 Lyon, France
- Université Claude Bernard Lyon 1, Virpath, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, F69372 Lyon, France
| | - Guislaine Carcelain
- Immunology Department, Robert Debré Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
- Université Paris Cité, INSERM U976, Paris, France
| | - Guillaume Lingas
- Université Paris Cité et Université Sorbonne Paris Nord, Inserm, IAME, F-75018 Paris, France
| | - France Mentré
- Université Paris Cité et Université Sorbonne Paris Nord, Inserm, IAME, F-75018 Paris, France
- Département d’Épidémiologie, Biostatistique et Recherche Clinique, AP-HP, Hôpital Bichat, F75018 Paris, France
| | - Florence Ader
- Département des Maladies Infectieuses et Tropicales, Hospices Civils de Lyon, Hôpital de la Croix-Rousse, F-69004 Lyon, France
- Université Claude Bernard Lyon 1, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, F-69372 Lyon, France
| | - Maya Hites
- Clinic of Infectious Diseases, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Pascal Poignard
- Groupe de Recherche en Infectiologie Clinique CIC-1406, Inserm—CHUGA—Université Grenoble Alpes, Grenoble, France
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
- Laboratoire de Virologie, Center Hospitalier Universitaire Grenoble-Alpes, Grenoble, France
| | - Jérémie Guedj
- Université Paris Cité et Université Sorbonne Paris Nord, Inserm, IAME, F-75018 Paris, France
| |
Collapse
|
2
|
Singh K, Rubenstein K, Callier V, Shaw-Saliba K, Rupert A, Dewar R, Laverdure S, Highbarger H, Lallemand P, Huang ML, Jerome KR, Sampoleo R, Mills MG, Greninger AL, Juneja K, Porter D, Benson CA, Dempsey W, El Sahly HM, Focht C, Jilg N, Paules CI, Rapaka RR, Uyeki TM, Clifford Lane H, Beigel J, Dodd LE. SARS-CoV-2 RNA and Nucleocapsid Antigen Are Blood Biomarkers Associated With Severe Disease Outcomes That Improve in Response to Remdesivir. J Infect Dis 2024; 230:624-634. [PMID: 38657001 PMCID: PMC11420797 DOI: 10.1093/infdis/jiae198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/09/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Although antivirals remain important for the treatment COVID-19, methods to assess treatment efficacy are lacking. Here, we investigated the impact of remdesivir on viral dynamics and their contribution to understanding antiviral efficacy in the multicenter Adaptive COVID-19 Treatment Trial 1, which randomized patients to remdesivir or placebo. METHODS Longitudinal specimens collected during hospitalization from a substudy of 642 patients with COVID-19 were measured for viral RNA (upper respiratory tract and plasma), viral nucleocapsid antigen (serum), and host immunologic markers. Associations with clinical outcomes and response to therapy were assessed. RESULTS Higher baseline plasma viral loads were associated with poorer clinical outcomes, and decreases in viral RNA and antigen in blood but not the upper respiratory tract correlated with enhanced benefit from remdesivir. The treatment effect of remdesivir was most pronounced in patients with elevated baseline nucleocapsid antigen levels: the recovery rate ratio was 1.95 (95% CI, 1.40-2.71) for levels >245 pg/mL vs 1.04 (95% CI, .76-1.42) for levels <245 pg/mL. Remdesivir also accelerated the rate of viral RNA and antigen clearance in blood, and patients whose blood levels decreased were more likely to recover and survive. CONCLUSIONS Reductions in SARS-CoV-2 RNA and antigen levels in blood correlated with clinical benefit from antiviral therapy. CLINICAL TRIAL REGISTRATION NCT04280705 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Kanal Singh
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - Kevin Rubenstein
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research
| | - Viviane Callier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research
| | | | - Adam Rupert
- National Laboratory for Cancer Research, Frederick, Maryland
| | - Robin Dewar
- National Laboratory for Cancer Research, Frederick, Maryland
| | | | | | | | - Meei-Li Huang
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | - Keith R Jerome
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
- Fred Hutchinson Cancer Center, Seattle, Washington
| | - Reigran Sampoleo
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | - Margaret G Mills
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | - Alexander L Greninger
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | | | | | | | - Walla Dempsey
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - Hana M El Sahly
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | | | - Nikolaus Jilg
- Massachusetts General Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston
| | - Catharine I Paules
- Division of Infectious Diseases, Milton S. Hershey Medical Center, Penn State Health, Hershey, Pennsylvania
| | - Rekha R Rapaka
- Center for Vaccine Development and Global Health, School of Medicine, University of Maryland, Baltimore
| | - Timothy M Uyeki
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - H Clifford Lane
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - John Beigel
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - Lori E Dodd
- National Institute of Allergy and Infectious Diseases, Bethesda
| |
Collapse
|
3
|
Balik M, Waldauf P, Jurisinova I, Svobodova E, Diblickova M, Tencer T, Zavora J, Smela G, Kupidlovska L, Adamkova V, Fridrichova M, Jerabkova K, Mikes J, Duska F, Dusek L. SARS-CoV-2 viral load is linked to remdesivir efficacy in severe Covid-19 admitted to intensive care. Sci Rep 2024; 14:20825. [PMID: 39242658 PMCID: PMC11379941 DOI: 10.1038/s41598-024-71588-9] [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: 04/11/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024] Open
Abstract
Remdesivir therapy has been declared as efficient in the early stages of Covid-19. Of the 339 patients (males 55.8%, age 71(59;77) years) with a detectable viral load, 140 were treated with remdesivir (of those 103 in the ICU and 57 immunosuppressed) and retrospectively compared with 199 patients (of those 82 in the ICU and 28 immunosuppressed) who were denied therapy due to advanced Covid-19. The viral load was estimated by detecting nucleocapsid antigen in serum (n = 155, median 217(28;1524)pg/ml), antigen in sputum (n = 18, COI 18(4.6;32)), nasopharyngeal antigen (n = 44, COI 17(8;35)) and the real-time PCR (n = 122, Ct 21(18;27)). After adjustment for confounders, patients on remdesivir had better 12-month survival (HR 0.66 (0.44;0.98), p = 0.039), particularly when admitted to the ICU (HR 0.49 (0.29;0.81), p = 0.006). For the immunocompromised patients, the difference did not reach statistical significance (HR 0.55 (0.18;1.69), p = 0.3). The other most significant confounders were age, ICU admission, mechanical ventilation, leukocyte/lymphocyte ratio, admission creatinine and immunosuppression. The impact of monoclonal antibodies or previous vaccinations was not significant. Despite frequent immune suppression including haemato-oncology diseases, lymphopenia, and higher inflammatory markers in the remdesivir group, the results support remdesivir administration with respect to widely available estimates of viral load in patients with high illness severity.
Collapse
Affiliation(s)
- M Balik
- Department of Anaesthesiology and Intensive Care, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, Prague 2, Prague, 12800, Czech Republic.
| | - P Waldauf
- Department of Anaesthesiology and Intensive Care, 3rd Faculty of Medicine, Charles University and Kralovske Vinohrady University Hospital in Prague, Prague, Czech Republic
| | - I Jurisinova
- Department of Anaesthesiology and Intensive Care, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, Prague 2, Prague, 12800, Czech Republic
| | - E Svobodova
- Department of Anaesthesiology and Intensive Care, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, Prague 2, Prague, 12800, Czech Republic
| | - M Diblickova
- Department of Anaesthesiology and Intensive Care, 1st Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 2, Prague 2, Prague, 12800, Czech Republic
| | - T Tencer
- Department of Anaesthesiology and Intensive Care, 3rd Faculty of Medicine, Charles University and Kralovske Vinohrady University Hospital in Prague, Prague, Czech Republic
| | - J Zavora
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic
- Department of Microbiology, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - G Smela
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic
| | - L Kupidlovska
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic
| | - V Adamkova
- Institute of Medical Biochemistry and Laboratory Diagnostics, 1st Faculty of Medicine, Charles University, and General University Hospital, Prague, Czech Republic
| | - M Fridrichova
- Department of Laboratory Diagnostics, 3rd Faculty of Medicine, Charles University and Kralovske Vinohrady University Hospital in Prague, Prague, Czech Republic
| | - K Jerabkova
- Department of Anaesthesiology and Intensive Care, 3rd Faculty of Medicine, Charles University and Kralovske Vinohrady University Hospital in Prague, Prague, Czech Republic
| | - J Mikes
- Department of Anaesthesiology and Intensive Care, 3rd Faculty of Medicine, Charles University and Kralovske Vinohrady University Hospital in Prague, Prague, Czech Republic
| | - F Duska
- Department of Anaesthesiology and Intensive Care, 3rd Faculty of Medicine, Charles University and Kralovske Vinohrady University Hospital in Prague, Prague, Czech Republic
| | - L Dusek
- Faculty of Medicine, Institute of Health Information and Statistics of the Czech Republic, Masaryk University, Brno, Czech Republic
| |
Collapse
|
4
|
Focosi D, Franchini M, Maggi F, Shoham S. COVID-19 therapeutics. Clin Microbiol Rev 2024; 37:e0011923. [PMID: 38771027 PMCID: PMC11237566 DOI: 10.1128/cmr.00119-23] [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] [Indexed: 05/22/2024] Open
Abstract
SUMMARYSince the emergence of COVID-19 in 2020, an unprecedented range of therapeutic options has been studied and deployed. Healthcare providers have multiple treatment approaches to choose from, but efficacy of those approaches often remains controversial or compromised by viral evolution. Uncertainties still persist regarding the best therapies for high-risk patients, and the drug pipeline is suffering fatigue and shortage of funding. In this article, we review the antiviral activity, mechanism of action, pharmacokinetics, and safety of COVID-19 antiviral therapies. Additionally, we summarize the evidence from randomized controlled trials on efficacy and safety of the various COVID-19 antivirals and discuss unmet needs which should be addressed.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Division of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Fabrizio Maggi
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome, Italy
| | - Shmuel Shoham
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Phan T, Zitzmann C, Chew KW, Smith DM, Daar ES, Wohl DA, Eron JJ, Currier JS, Hughes MD, Choudhary MC, Deo R, Li JZ, Ribeiro RM, Ke R, Perelson AS. Modeling the emergence of viral resistance for SARS-CoV-2 during treatment with an anti-spike monoclonal antibody. PLoS Pathog 2024; 20:e1011680. [PMID: 38635853 PMCID: PMC11060554 DOI: 10.1371/journal.ppat.1011680] [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: 09/13/2023] [Revised: 04/30/2024] [Accepted: 03/18/2024] [Indexed: 04/20/2024] Open
Abstract
To mitigate the loss of lives during the COVID-19 pandemic, emergency use authorization was given to several anti-SARS-CoV-2 monoclonal antibody (mAb) therapies for the treatment of mild-to-moderate COVID-19 in patients with a high risk of progressing to severe disease. Monoclonal antibodies used to treat SARS-CoV-2 target the spike protein of the virus and block its ability to enter and infect target cells. Monoclonal antibody therapy can thus accelerate the decline in viral load and lower hospitalization rates among high-risk patients with variants susceptible to mAb therapy. However, viral resistance has been observed, in some cases leading to a transient viral rebound that can be as large as 3-4 orders of magnitude. As mAbs represent a proven treatment choice for SARS-CoV-2 and other viral infections, evaluation of treatment-emergent mAb resistance can help uncover underlying pathobiology of SARS-CoV-2 infection and may also help in the development of the next generation of mAb therapies. Although resistance can be expected, the large rebounds observed are much more difficult to explain. We hypothesize replenishment of target cells is necessary to generate the high transient viral rebound. Thus, we formulated two models with different mechanisms for target cell replenishment (homeostatic proliferation and return from an innate immune response antiviral state) and fit them to data from persons with SARS-CoV-2 treated with a mAb. We showed that both models can explain the emergence of resistant virus associated with high transient viral rebounds. We found that variations in the target cell supply rate and adaptive immunity parameters have a strong impact on the magnitude or observability of the viral rebound associated with the emergence of resistant virus. Both variations in target cell supply rate and adaptive immunity parameters may explain why only some individuals develop observable transient resistant viral rebound. Our study highlights the conditions that can lead to resistance and subsequent viral rebound in mAb treatments during acute infection.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Carolin Zitzmann
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Kara W. Chew
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Davey M. Smith
- Department of Medicine, University of California, San Diego, California, United States of America
| | - Eric S. Daar
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, United States of America
| | - David A. Wohl
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Joseph J. Eron
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Judith S. Currier
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Michael D. Hughes
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Manish C. Choudhary
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Rinki Deo
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jonathan Z. Li
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ruy M. Ribeiro
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Ruian Ke
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Alan S. Perelson
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
- Santa Fe Institute, Santa Fe, New Mexico, United States of America
| | | |
Collapse
|
6
|
Hites M, Massonnaud CR, Lapique EL, Belhadi D, Jamard S, Goehringer F, Danion F, Reignier J, de Castro N, Garot D, Lacombe K, Tolsma V, Faure E, Malvy D, Staub T, Courjon J, Cazenave-Roblot F, Dyrhol Riise AM, Leturnier P, Martin-Blondel G, Roger C, Akinosoglou K, Moing VL, Piroth L, Sellier P, Lescure X, Trøseid M, Clevenbergh P, Dalgard O, Gallien S, Gousseff M, Loubet P, Vardon-Bounes F, Visée C, Belkhir L, Botelho-Nevers É, Cabié A, Kotanidou A, Lanternier F, Rouveix-Nordon E, Silva S, Thiery G, Poignard P, Carcelain G, Diallo A, Mercier N, Terzic V, Bouscambert-Duchamp M, Gaymard A, Trabaud MA, Destras G, Josset L, Billard N, Han THL, Guedj J, Couffin-Cadiergues S, Dechanet A, Delmas C, Esperou H, Fougerou-Leurent C, Mestre SL, Métois A, Noret M, Bally I, Dergan-Dylon S, Tubiana S, Kalif O, Bergaud N, Leveau B, Eustace J, Greil R, Hajdu E, Halanova M, Paiva JA, Piekarska A, Rodriguez Baño J, Tonby K, Trojánek M, Tsiodras S, Unal S, Burdet C, Costagliola D, Yazdanpanah Y, Peiffer-Smadja N, Mentré F, Ader F. Tixagevimab-cilgavimab (AZD7442) for the treatment of patients hospitalized with COVID-19 (DisCoVeRy): A phase 3, randomized, double-blind, placebo-controlled trial. J Infect 2024; 88:106120. [PMID: 38367705 DOI: 10.1016/j.jinf.2024.106120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024]
Affiliation(s)
- Maya Hites
- Clinic of Infectious Diseases, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles, Brussels, Belgium.
| | - Clément R Massonnaud
- Université Paris Cité, Inserm, IAME, F-75018 Paris, France; AP-HP, Hôpital Bichat, Département d'Épidémiologie, Biostatistique et Recherche Clinique, F-75018 Paris, France
| | - Eva Larranaga Lapique
- Clinic of Infectious Diseases, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Drifa Belhadi
- Université Paris Cité, Inserm, IAME, F-75018 Paris, France; AP-HP, Hôpital Bichat, Département d'Épidémiologie, Biostatistique et Recherche Clinique, F-75018 Paris, France
| | - Simon Jamard
- Service de Maladies Infectieuses Et Tropicales (SMIT), Centre Hospitalier Universitaire de Tours, 37044 Tours, France
| | - François Goehringer
- Université de Lorraine, CHRU de Nancy, Service des Maladies Infectieuses et Tropicales, F-54000 Nancy, France
| | - François Danion
- Hôpitaux Universitaires de Strasbourg, Département de maladies infectieuses et tropicales, F-67091 Strasbourg, France
| | - Jean Reignier
- CHU de Nantes, Service de Médecine Intensive et Réanimation, Université de Nantes, F-44093 Nantes, France
| | - Nathalie de Castro
- Département des Maladies Infectieuses et Tropicales, GH Saint-Louis/Lariboisière-Fernand Widal, Université de Paris Cité, INSERM U 944, Paris, France
| | - Denis Garot
- CHRU Tours, Service de Médecine Intensive Réanimation, F-37044 Tours, France
| | - Karine Lacombe
- Sorbonne Université, Inserm, Institut Pierre-Louis d'Épidémiologie et de Santé Publique, F-75013 Paris, France; APHP, Hôpital Saint-Antoine, Service de maladies infectieuses et tropicales, F-75012 Paris, France
| | - Violaine Tolsma
- Centre Hospitalier Annecy Genevois, Service des Maladies Infectieuses et Tropicales, F-74374 Annecy, France
| | - Emmanuel Faure
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Denis Malvy
- Department of Infectious Diseases and Tropical Medicine, CHU Bordeaux, Bordeaux, France
| | - Thérèse Staub
- Centre hospitalier de Luxembourg, Service des maladies infectieuses, L-1210 Luxembourg, Luxembourg
| | - Johan Courjon
- Université Côte d'Azur, CHU Nice, Nice, France, Infectious Disease Unit, Nice, France
| | - France Cazenave-Roblot
- Département des Maladies Infectieuses et Tropicales, CHU de Poitiers, INSERM U1070, Poitiers, France
| | | | - Paul Leturnier
- Department of Infectious Diseases, Hôtel-Dieu University Hospital, University Hospital of Nantes, Nantes, France
| | - Guillaume Martin-Blondel
- CHU de Toulouse, Service des maladies infectieuses et Tropicales, F-31320 Toulouse, France; Institut Toulousain des Maladies Infectieuses et Inflammatoires (Infinity) INSERM UMR1291 - CNRS UMR5051 - Université Toulouse III, F-31320 Toulouse, France
| | - Claire Roger
- Department of Anesthesiology, Critical Care Pain, and Emergency Medicine, Nimes University Hospital, Nimes, France
| | - Karolina Akinosoglou
- Department of Internal Medicine and Infectious Diseases, University General Hospital of Patras, Patras, Greece
| | - Vincent Le Moing
- CHU de Montpellier, Service des Maladies Infectieuses et Tropicales, F-34295 Montpellier, France
| | - Lionel Piroth
- CHU de Dijon, Département de Maladies Infectieuses, F-21000, Dijon, France; Université Bourgogne Franche-Comté, CIC 1432, INSERM, F-21000, Dijon, France
| | - Pierre Sellier
- Infectious Diseases Department, Lariboisière Hospital, AP-HP, Paris, France
| | - Xavier Lescure
- Université Paris Cité, Inserm, IAME, F-75018 Paris, France; AP-HP, Hôpital Bichat, Service de Maladies Infectieuses et Tropicales, F-75018 Paris, France
| | - Marius Trøseid
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Diseases and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | | | - Olav Dalgard
- Department of Infectious Diseases, Division of Medicine, Akershus University Hospital, Lørenskog, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sébastien Gallien
- APHP, Hôpital Henri Mondor, Département de maladies infectieuses, F-94000 Créteil, France; INSERM U955, Team 16, IMRB Créteil, Créteil, France
| | - Marie Gousseff
- Maladies infectieuses, Centre Hospitalier Bretagne-Atlantique, Vannes, France
| | - Paul Loubet
- Infectious and Tropical Diseases Department, Nimes University Hospital, Nimes, France; VBIC, INSERM U1047, University of Montpellier, Nimes, France
| | - Fanny Vardon-Bounes
- CHU de Toulouse, Département d'anesthésie et de soins intensifs, F-31300 Toulouse, France; Université Toulouse 3 Paul Sabatier, Inserm U1297, F-31300 Toulouse, France
| | - Clotilde Visée
- Department of Infectious Disease, Centre Hospitalier Régional Mons-Hainaut/Groupe Jolimont, Mons Belgium/Groupe Helora, Mons, Belgium
| | - Leila Belkhir
- Department of Internal Medicine and Infectious Diseases, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Élisabeth Botelho-Nevers
- CHU de Saint-Etienne, Service d'Infectiologie, F-42055 Saint-Etienne, France; Université Jean Monnet, Université Claude Bernard Lyon 1, GIMAP, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, F-42023 Saint-Etienne, France; CIC 1408, INSERM, F, 42055 Saint-Etienne, France
| | - André Cabié
- PCCEI, Univ Montpellier, Univ Antilles, Inserm, EFS, F-34394 Montpellier, France; CHU de Martinique, Service des maladies infectieuses et tropicales, Inserm CIC1424, F-97200 Fort de France, France
| | - Anastasia Kotanidou
- First Department of Critical Care Medicine and Pulmonary Services, Evangelismos Hospital, National and Kapodistrian University of Athens Medical School, 45-47 Ipsilantou Street, 10676 Athens, Greece
| | - Fanny Lanternier
- Infectious Diseases Unit, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
| | - Elisabeth Rouveix-Nordon
- AP-HP, Hôpital Ambroise-Paré, Service de Maladies Infectieuses et Tropicales, Boulogne-Billancourt, France
| | - Susana Silva
- EPIUnit - Instituto de Saúde Pública, Universidade do Porto, Rua das Taipas, no 135, 4050-600 Porto, Portugal
| | - Guillaume Thiery
- CHU Saint-Etienne, Hopital Nord, Medical Intensive Care Unit, Saint-Priest-En-Jarez, France
| | - Pascal Poignard
- Groupe de Recherche en Infectiologie Clinique CIC-1406, Inserm - CHUGA - Université Grenoble Alpes, Grenoble, France; Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France; Laboratoire de Virologie, Center Hospitalier Universitaire Grenoble-Alpes, Grenoble, France
| | - Guislaine Carcelain
- Immunology Department, Robert Debré Hospital, Assistance Publique Hôpitaux de Paris, Paris, France; Université Paris Cité, INSERM U976, Paris, France
| | - Alpha Diallo
- ANRS | Maladies Infectieuses Emergentes, Paris, France
| | | | - Vida Terzic
- ANRS | Maladies Infectieuses Emergentes, Paris, France
| | - Maude Bouscambert-Duchamp
- Hospices Civils de Lyon, Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des virus respiratoires France Sud, F-69317 Lyon, France; Université Claude Bernard Lyon 1, Virpath, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, F-69372 Lyon, France
| | - Alexandre Gaymard
- Hospices Civils de Lyon, Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des virus respiratoires France Sud, F-69317 Lyon, France; Université Claude Bernard Lyon 1, Virpath, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, F-69372 Lyon, France
| | | | - Grégory Destras
- Hospices Civils de Lyon, Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des virus respiratoires France Sud, F-69317 Lyon, France
| | - Laurence Josset
- Hospices Civils de Lyon, Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des virus respiratoires France Sud, F-69317 Lyon, France
| | - Nicolas Billard
- AP-HP, Hôpital Bichat, Département d'Épidémiologie, Biostatistique et Recherche Clinique, F-75018 Paris, France
| | - Thi-Hong-Lien Han
- AP-HP, Hôpital Bichat, Département d'Épidémiologie, Biostatistique et Recherche Clinique, F-75018 Paris, France
| | - Jérémie Guedj
- Université Paris Cité, Inserm, IAME, F-75018 Paris, France
| | | | - Aline Dechanet
- AP-HP, Hôpital Bichat, Unité de recherche clinique, F-75018 Paris, France
| | - Christelle Delmas
- Institut de santé publique, Pôle recherche clinique, INSERM, Paris, France
| | - Hélène Esperou
- Institut de santé publique, Pôle recherche clinique, INSERM, Paris, France
| | | | | | - Anabelle Métois
- AP-HP, Hôpital Bichat, Unité de recherche clinique, F-75018 Paris, France
| | - Marion Noret
- Renarci, Réseau National De Recherche Clinique En Infectiologie, Paris, France
| | - Isabelle Bally
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Sebastián Dergan-Dylon
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale (IBS), Grenoble, France
| | - Sarah Tubiana
- Université Paris Cité, Inserm, IAME, F-75018 Paris, France; AP-HP, Hôpital Bichat, Centre de ressources biologiques, F-75018 Paris, France
| | - Ouifiya Kalif
- AP-HP, Hôpital Bichat, Centre de ressources biologiques, F-75018 Paris, France
| | | | | | | | - Richard Greil
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Salzburg Cancer Research Institute - Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; Cancer Cluster Salzburg, 5020 Salzburg, Austria; AGMT, 5020 Salzburg, Austria
| | - Edit Hajdu
- Department of Internal Medicine Infectiology Unit, Albert Szent-Györgyi Health Centre, University of Szeged, Állomás Street 1-3, 6725 Szeged, Hungary
| | - Monika Halanova
- LF UPJŠ - Pavol Jozef Šafárik University in Košice Faculty of Medicine, Košice, Slovakia
| | - Jose-Artur Paiva
- Centro Hospitalar São João, Emergency and Intensive Care Department, Porto, Portugal; Universidade do Porto, Faculty of Medicine, Porto, Portugal
| | - Anna Piekarska
- Department of Infectious Diseases and Hepatology, Medical University of Łódź, Łódź, Poland
| | - Jesus Rodriguez Baño
- Infectious Diseases and Microbiology Division, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Kristian Tonby
- Department of Infectious Diseases, Oslo University Hospital, 0424 Oslo, Norway
| | - Milan Trojánek
- Department of Infectious Diseases, University Hospital Bulovka, Budínova 2, 180 81, Prague, Czech Republic
| | - Sotirios Tsiodras
- Fourth Department of Internal Medicine, Attikon University Hospital, Athens Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Serhat Unal
- Department of Infectious Diseases, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Charles Burdet
- Université Paris Cité, Inserm, IAME, F-75018 Paris, France; AP-HP, Hôpital Bichat, Département d'Épidémiologie, Biostatistique et Recherche Clinique, F-75018 Paris, France
| | - Dominique Costagliola
- Sorbonne Université, Inserm, Institut Pierre-Louis d'Épidémiologie et de Santé Publique, F-75013 Paris, France
| | - Yazdan Yazdanpanah
- Université Paris Cité, Inserm, IAME, F-75018 Paris, France; AP-HP, Hôpital Bichat, Service de Maladies Infectieuses et Tropicales, F-75018 Paris, France
| | - Nathan Peiffer-Smadja
- Université Paris Cité, Inserm, IAME, F-75018 Paris, France; AP-HP, Hôpital Bichat, Service de Maladies Infectieuses et Tropicales, F-75018 Paris, France; National Institute for Health Research, Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Imperial College London, London, UK
| | - France Mentré
- Université Paris Cité, Inserm, IAME, F-75018 Paris, France; AP-HP, Hôpital Bichat, Département d'Épidémiologie, Biostatistique et Recherche Clinique, F-75018 Paris, France
| | - Florence Ader
- Hospices Civils de Lyon, Hôpital de la Croix-Rousse, Département des Maladies Infectieuses et Tropicales, F-69004 Lyon, France; Université Claude Bernard Lyon 1, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, F-69372 Lyon, France
| |
Collapse
|
7
|
Néant N, Lingas G, Gaymard A, Belhadi D, Hites M, Staub T, Greil R, Paiva J, Poissy J, Peiffer‐Smadja N, Costagliola D, Yazdanpanah Y, Bouscambert‐Duchamp M, Gagneux‐Brunon A, Ader F, Mentré F, Wallet F, Burdet C, Guedj J. Association between SARS-CoV-2 viral kinetics and clinical score evolution in hospitalized patients. CPT Pharmacometrics Syst Pharmacol 2023; 12:2027-2037. [PMID: 37728045 PMCID: PMC10725266 DOI: 10.1002/psp4.13051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/27/2023] [Accepted: 09/05/2023] [Indexed: 09/21/2023] Open
Abstract
The role of antiviral treatment in coronavirus disease 2019 hospitalized patients is controversial. To address this question, we analyzed simultaneously nasopharyngeal viral load and the National Early Warning Score 2 (NEWS-2) using an effect compartment model to relate viral dynamics and the evolution of clinical severity. The model is applied to 664 hospitalized patients included in the DisCoVeRy trial (NCT04315948; EudraCT 2020-000936-23) randomly assigned to either standard of care (SoC) or SoC + remdesivir. Then we use the model to simulate the impact of antiviral treatments on the time to clinical improvement, defined by a NEWS-2 score lower than 3 (in patients with NEWS-2 <7 at hospitalization) or 5 (in patients with NEWS-2 ≥7 at hospitalization), distinguishing between patients with low or high viral load at hospitalization. The model can fit well the different observed patients trajectories, showing that clinical evolution is associated with viral dynamics, albeit with large interindividual variability. Remdesivir antiviral activity was 22% and 78% in patients with low or high viral loads, respectively, which is not sufficient to generate a meaningful effect on NEWS-2. However, simulations predicted that antiviral activity greater than 99% could reduce by 2 days the time to clinical improvement in patients with high viral load, irrespective of the NEWS-2 score at hospitalization, whereas no meaningful effect was predicted in patients with low viral loads. Our results demonstrate that time to clinical improvement is associated with time to viral clearance and that highly effective antiviral drugs could hasten clinical improvement in hospitalized patients with high viral loads.
Collapse
Affiliation(s)
- Nadège Néant
- IAMEUniversité Paris Cité, IAME, Inserm, F‐75018ParisFrance
| | | | - Alexandre Gaymard
- Laboratoire de Virologie, Institut des Agents Infectieux de LyonCentre National de Référence des Virus Respiratoires France Sud, Hospices Civils de LyonLyonFrance
- Laboratoire VirpathUniversité de Lyon, Virpath, CIRI, INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1LyonFrance
| | - Drifa Belhadi
- IAMEUniversité Paris Cité, IAME, Inserm, F‐75018ParisFrance
- Département d'ÉpidémiologieAP‐HP, Hôpital Bichat, Biostatistique et Recherche CliniqueParisFrance
| | - Maya Hites
- Hôpital de Bruxelles‐ÉrasmeUniversité Libre de Bruxelles, Clinique des Maladies InfectieusesBrusselsBelgium
| | - Thérèse Staub
- Centre Hospitalier de Luxembourg, Service des Maladies InfectieusesLuxembourgLuxembourg
| | - Richard Greil
- Department of Internal Medicine III with Haematology, Medical Oncology, Aemostaseology, Infectiology and Rheumatology, Oncologic CenterSalzburg Cancer Research Institute–Laboratory for Immunological and Molecular Cancer Research, Paracelsus Medical University SalzburgSalzburgAustria
- Cancer Cluster SalzburgSalzburgAustria
- AGMTSalzburgAustria
| | - Jose‐Artur Paiva
- Emergency and Intensive Care Department, Centro Hospitalar São JoãoPortoPortugal
- Faculty of MedicineUniversidade do PortoPortoPortugal
| | - Julien Poissy
- Intensive Care DepartmentUniversité de Lille, Inserm U1285, CHU Lille, Pôle de Réanimation, CNRS, UMR 8576–UGSF–Unité de Glycobiologie Structurale et FonctionnelleLilleFrance
| | - Nathan Peiffer‐Smadja
- IAMEUniversité Paris Cité, IAME, Inserm, F‐75018ParisFrance
- AP‐HP, Hôpital Bichat, Service de Maladies Infectieuses et TropicalesParisFrance
- National Institute for Health Research, Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, Imperial College LondonLondonUK
| | - Dominique Costagliola
- Sorbonne Université, Inserm, Institut Pierre‐Louis d'Épidémiologie et de Santé PubliqueParisFrance
| | - Yazdan Yazdanpanah
- IAMEUniversité Paris Cité, IAME, Inserm, F‐75018ParisFrance
- AP‐HP, Hôpital Bichat, Service de Maladies Infectieuses et TropicalesParisFrance
| | - Maude Bouscambert‐Duchamp
- Laboratoire de Virologie, Institut des Agents Infectieux de LyonCentre National de Référence des Virus Respiratoires France Sud, Hospices Civils de LyonLyonFrance
| | - Amandine Gagneux‐Brunon
- CHU de Saint‐Etienne, Service d'InfectiologieSaint‐EtienneFrance
- Université Jean Monnet, Université Claude Bernard Lyon 1, GIMAP, CIRI, INSERM U1111, CNRS UMR5308, ENS LyonSaint‐EtienneFrance
- CIC 1408, INSERMSaint‐EtienneFrance
| | - Florence Ader
- Département des Maladies Infectieuses et TropicalesHospices Civils de LyonLyonFrance
- Département des Maladies Infectieuses et TropicalesUniversité Claude Bernard Lyon 1, CIRI, INSERM U1111, CNRS UMR5308, ENS LyonLyonFrance
| | - France Mentré
- IAMEUniversité Paris Cité, IAME, Inserm, F‐75018ParisFrance
- Département d'ÉpidémiologieAP‐HP, Hôpital Bichat, Biostatistique et Recherche CliniqueParisFrance
| | - Florent Wallet
- Service de Médecine Intensive Réanimation Anesthésie, Centre Hospitalier Lyon SudHospices Civils de LyonPierre‐BeniteFrance
| | - Charles Burdet
- IAMEUniversité Paris Cité, IAME, Inserm, F‐75018ParisFrance
- Département d'ÉpidémiologieAP‐HP, Hôpital Bichat, Biostatistique et Recherche CliniqueParisFrance
| | - Jérémie Guedj
- IAMEUniversité Paris Cité, IAME, Inserm, F‐75018ParisFrance
| | | |
Collapse
|
8
|
Hagman K, Hedenstierna M, Widaeus J, Arvidsson E, Hammas B, Grillner L, Jakobsson J, Gille-Johnson P, Ursing J. Effects of remdesivir on SARS-CoV-2 viral dynamics and mortality in viraemic patients hospitalized for COVID-19. J Antimicrob Chemother 2023; 78:2735-2742. [PMID: 37757451 PMCID: PMC10631829 DOI: 10.1093/jac/dkad295] [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: 04/23/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Studies on the antiviral effects of remdesivir have shown conflicting results. SARS-CoV-2 viraemia could identify patients in whom antiviral treatment may be particularly beneficial. OBJECTIVES To investigate antiviral effects and clinical outcomes of remdesivir treatment in viraemic patients. METHODS Viraemic patients hospitalized for COVID-19 with ratio of arterial oxygen partial pressure to fractional inspired oxygen of ≤300, symptom duration ≤10 days, and estimated glomerular filtration rate ≥30 mL/min were included in a cohort. The rate of serum viral clearance and serum viral load decline, 60 day mortality and in-hospital outcomes were estimated. A subgroup analysis including patients with symptom duration ≤7 days was performed. RESULTS A total of 318 viraemic patients were included. Thirty-three percent (105/318) received remdesivir. The rate of serum viral clearance [subhazard risk ratio (SHR) 1.4 (95% CI 0.9-2.0), P = 0.11] and serum viral load decline (P = 0.11) were not significantly different between remdesivir-treated patients and controls. However, the rate of serum viral clearance was non-significantly higher [SHR 1.6 (95% CI 1.0-2.7), P = 0.051] and the viral load decline was faster (P = 0.03) in remdesivir-treated patients with symptom duration ≤7 days at admission. The 60 day mortality [HR 1.0 (95% CI 0.6-1.8), P = 0.97] and adverse in-hospital outcomes [OR 1.4 (95% CI 0.8-2.4), P = 0.31] were not significantly different between remdesivir-treated patients and controls. CONCLUSIONS Remdesivir treatment did not significantly change the duration of SARS-CoV-2 viraemia, decline of serum viral load, 60 day mortality or in-hospital adverse outcomes in patients with ≤10 days of symptoms at admission. Remdesivir appeared to reduce the duration of viraemia in a subgroup of patients with ≤7 days of symptoms at admission.
Collapse
Affiliation(s)
- Karl Hagman
- Department of Infectious Diseases, Sahlgrenska University Hospital, Diagnosvagen 21, 416 50 Gothenburg, Sweden
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | - Jacob Widaeus
- Department of Infectious Diseases, Danderyd Hospital, Stockholm, Sweden
| | - Emelie Arvidsson
- Department of Infectious Diseases, Danderyd Hospital, Stockholm, Sweden
| | - Berit Hammas
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm Sweden
| | - Lena Grillner
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm Sweden
| | - Jan Jakobsson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Department of Anaesthesia and Intensive Care, Danderyd Hospital, Stockholm, Sweden
| | | | - Johan Ursing
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Danderyd Hospital, Stockholm, Sweden
| |
Collapse
|
9
|
Giannitsioti E, Mavroudis P, Speggos I, Katsoulidou A, Pantazis N, Loupis T, Daniil I, Rekleiti N, Damianidou S, Louka C, Sidiropoulou C, Kranidiotis G, Velentza L, Stamati A, Kasidiaraki M, Efstratiadi E, Linardaki G, Chrysos G, Zarkotou O, Zoi K, Tryfinopoulou K, Gerakari S. Real life treatment experience and outcome of consecutively hospitalised patients with SARS-CoV-2 pneumonia by Omicron-1 vs Delta variants. Infect Dis (Lond) 2023; 55:706-715. [PMID: 37427461 DOI: 10.1080/23744235.2023.2232445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/11/2023] Open
Abstract
BACKGROUND Omicron-1 COVID-19 is less invasive in the general population than previous viral variants. However, clinical course and outcome of hospitalised patients with SARS-CoV-2 pneumonia during the shift of the predominance from Delta to Omicron variants are not fully explored. METHODS During January 2022 consecutively hospitalised patients with SARS-CoV-2 pneumonia were analysed. SARS-CoV-2 variants were identified by a 2-step pre-screening protocol and randomly confirmed by whole genome sequencing analysis. Clinical, laboratory and treatment data split by type of variant were analysed along with logistic regression of factors associated to mortality. RESULTS 150 patients [mean age (SD) 67.2(15.8) years, male 54%] were analysed. Compared to Delta (n = 46), Omicron-1 patients (n = 104) were older [mean age (SD): 69.5(15.4) vs 61.9(15.8) years, p = 0.007], with more comorbidities (89.4% vs 65.2%, p = 0.001), less obesity (BMI >30Kg/m2 in 24% vs 43.5%, p = 0.034) but higher vaccination rates for COVID-19 (52.9% vs 8.7%, p < 0.001). Severe pneumonia (48.7%), pulmonary embolism (4.7%), need for invasive mechanical ventilation (8%), administration of dexamethasone (76%) and 60-day mortality (22.6%) did not significantly differ. Severe SARS-CoV-2 pneumonia independently predicted mortality [OR 8.297 (CI95% 2.080-33.095), p = 0.003]. Remdesivir administration (n = 135) was protective from death both in unadjusted and adjusted models [OR 0.157 (CI95% 0.026-0.945), p = 0.043. CONCLUSIONS In a COVID-19 department the severity of pneumonia that did not differ between Omicron-1 and Delta variants predicted mortality whilst remdesivir remained protective in all analyses. Death rates did not differ between SARS-CoV-2 variants. Vigilance and consistency with prevention and treatment guidelines for COVID-19 is mandatory regardless of the predominant SARS-CoV-2 variant.
Collapse
Affiliation(s)
- Efthymia Giannitsioti
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Fourth Department of Internal Medicine, National and Kapodistrian University of Athens, ATTIKON University General Hospital, Athens, Greece
| | - Panagiotis Mavroudis
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Second Department of Internal Medicine, Tzaneio General Hospital, Piraeus, Greece
| | - Ioannis Speggos
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Second Department of Internal Medicine, Tzaneio General Hospital, Piraeus, Greece
| | - Antigoni Katsoulidou
- Central Public Health Laboratory, National Public Health Organization, Athens, Greece
| | - Nikos Pantazis
- Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Theodoros Loupis
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
- Haematology Research Laboratory, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Ioannis Daniil
- Department of Microbiology, Tzaneio General Hospital, Piraeus, Greece
| | - Nektaria Rekleiti
- Department of Microbiology, Tzaneio General Hospital, Piraeus, Greece
| | - Sofia Damianidou
- Central Public Health Laboratory, National Public Health Organization, Athens, Greece
| | - Christina Louka
- Department of Microbiology, Tzaneio General Hospital, Piraeus, Greece
| | - Chrysanthi Sidiropoulou
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Second Department of Internal Medicine, Tzaneio General Hospital, Piraeus, Greece
| | - Georgios Kranidiotis
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Second Department of Internal Medicine, Tzaneio General Hospital, Piraeus, Greece
| | - Lemonia Velentza
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Emergency Department, Tzaneio General Hospital, Piraeus, Greece
| | - Alexandra Stamati
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Second Department of Internal Medicine, Tzaneio General Hospital, Piraeus, Greece
| | - Maria Kasidiaraki
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Emergency Department, Tzaneio General Hospital, Piraeus, Greece
| | - Efrosini Efstratiadi
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Emergency Department, Tzaneio General Hospital, Piraeus, Greece
| | - Garyfallia Linardaki
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Second Department of Internal Medicine, Tzaneio General Hospital, Piraeus, Greece
| | - Georgios Chrysos
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Second Department of Internal Medicine, Tzaneio General Hospital, Piraeus, Greece
| | - Olympia Zarkotou
- Department of Microbiology, Tzaneio General Hospital, Piraeus, Greece
| | - Katerina Zoi
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
- Haematology Research Laboratory, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Kyriaki Tryfinopoulou
- Central Public Health Laboratory, National Public Health Organization, Athens, Greece
| | - Styliani Gerakari
- COVID-19 Department, Tzaneio General Hospital, Piraeus, Greece
- Emergency Department, Tzaneio General Hospital, Piraeus, Greece
| |
Collapse
|
10
|
Phan T, Zitzmann C, Chew KW, Smith DM, Daar ES, Wohl DA, Eron JJ, Currier JS, Hughes MD, Choudhary MC, Deo R, Li JZ, Ribeiro RM, Ke R, Perelson AS. Modeling the emergence of viral resistance for SARS-CoV-2 during treatment with an anti-spike monoclonal antibody. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.14.557679. [PMID: 37745410 PMCID: PMC10515893 DOI: 10.1101/2023.09.14.557679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The COVID-19 pandemic has led to over 760 million cases and 6.9 million deaths worldwide. To mitigate the loss of lives, emergency use authorization was given to several anti-SARS-CoV-2 monoclonal antibody (mAb) therapies for the treatment of mild-to-moderate COVID-19 in patients with a high risk of progressing to severe disease. Monoclonal antibodies used to treat SARS-CoV-2 target the spike protein of the virus and block its ability to enter and infect target cells. Monoclonal antibody therapy can thus accelerate the decline in viral load and lower hospitalization rates among high-risk patients with susceptible variants. However, viral resistance has been observed, in some cases leading to a transient viral rebound that can be as large as 3-4 orders of magnitude. As mAbs represent a proven treatment choice for SARS-CoV-2 and other viral infections, evaluation of treatment-emergent mAb resistance can help uncover underlying pathobiology of SARS-CoV-2 infection and may also help in the development of the next generation of mAb therapies. Although resistance can be expected, the large rebounds observed are much more difficult to explain. We hypothesize replenishment of target cells is necessary to generate the high transient viral rebound. Thus, we formulated two models with different mechanisms for target cell replenishment (homeostatic proliferation and return from an innate immune response anti-viral state) and fit them to data from persons with SARS-CoV-2 treated with a mAb. We showed that both models can explain the emergence of resistant virus associated with high transient viral rebounds. We found that variations in the target cell supply rate and adaptive immunity parameters have a strong impact on the magnitude or observability of the viral rebound associated with the emergence of resistant virus. Both variations in target cell supply rate and adaptive immunity parameters may explain why only some individuals develop observable transient resistant viral rebound. Our study highlights the conditions that can lead to resistance and subsequent viral rebound in mAb treatments during acute infection.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Carolin Zitzmann
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Kara W. Chew
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Davey M. Smith
- Department of Medicine, University of California, San Diego, CA, USA
| | - Eric S. Daar
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - David A. Wohl
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Joseph J. Eron
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Judith S. Currier
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | | - Manish C. Choudhary
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Rinki Deo
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan Z. Li
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruy M. Ribeiro
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Ruian Ke
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Alan S. Perelson
- Theoretical Biology & Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
- Santa Fe Institute, Santa Fe, NM, USA
| | | |
Collapse
|
11
|
Chang TH, Wu CH, Chen PY, Ho SY, Chung MY, Sheng WH, Lu CY, Yen TY, Chen JM, Lee PI, Tang HJ, Ho CH, Chang LY, Chen YC, Huang LM. Viral dynamics of SARS-CoV-2 Omicron infections in a previously low COVID-19 prevalence region: Effects of vaccination status, antiviral agents, and age. J Formos Med Assoc 2023; 122:872-879. [PMID: 37179128 PMCID: PMC10151453 DOI: 10.1016/j.jfma.2023.04.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/28/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND In Taiwan, the prevalence of COVID-19 was low before April 2022. The low SARS-CoV-2 seroprevalence in the population of Taiwan provides an opportunity for comparison with fewer confounding factors than other populations globally. Cycle threshold (Ct) value is an easily accessible method for modeling SARS-CoV-2 dynamics. In this study, we used clinical samples collected from hospitalized patients to explore the Ct value dynamics of the Omicron variant infection. METHODS From Jan 2022 to May 2022, we retrospectively included hospitalized patients tested positive by nasopharyngeal SARS-CoV-2 PCR. We categorized the test-positive subjects into different groups according to age, vaccination status, and use of antiviral agents. To investigate the nonlinear relationship between symptom onset days and Ct value, a fractional polynomial model was applied to draw a regression line. RESULTS We collected 1718 SARS-CoV-2 viral samples from 812 individuals. The Ct values of unvaccinated individuals were lower than those of vaccinated persons from Day 4 to Day 10 after symptom onset. The Ct value increased more rapidly in those individuals with antiviral drug treatment from Day 2 to Day 7. In elderly individuals, the Ct values increased slowly from Day 5 to Day 10, and the increasing trend was unique compared with that in children and adults. CONCLUSION Our study demonstrated the primary viral infection dynamics of the Omicron variant in hospitalized patients. Vaccination significantly affected viral dynamics, and antiviral agents modified viral dynamics irrespective of vaccination status. In elderly individuals, viral clearance is slower than that in adults and children.
Collapse
Affiliation(s)
- Tu-Hsuan Chang
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan
| | - Chi-Hsien Wu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, Cardinal Tien Hospital, Taipei, Taiwan
| | - Po-Yu Chen
- Department of Nursing, Min-Hwei College of Health Care Management, Tainan, Taiwan
| | - Shu-Yuan Ho
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Yi Chung
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wang-Huei Sheng
- Division of Infectious Diseases, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chun-Yi Lu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Ting-Yu Yen
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Jong-Min Chen
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ping-Ing Lee
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Hung-Jen Tang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Chung-Han Ho
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan; Department of Information Management, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Luan-Yin Chang
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.
| | - Yee-Chun Chen
- Division of Infectious Diseases, Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Li-Min Huang
- Division of Pediatric Infectious Diseases, Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
12
|
Blair HA. Remdesivir: A Review in COVID-19. Drugs 2023; 83:1215-1237. [PMID: 37589788 PMCID: PMC10474216 DOI: 10.1007/s40265-023-01926-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 08/18/2023]
Abstract
Remdesivir (Veklury®), a nucleotide analogue prodrug with broad-spectrum antiviral activity, is approved for the treatment of coronavirus disease 2019 (COVID-19), the illness caused by severe acute respiratory syndrome coronavirus 2 infection. Unlike some antivirals, remdesivir has a low potential for drug-drug interactions. In the pivotal ACTT-1 trial in hospitalized patients with COVID-19, daily intravenous infusions of remdesivir significantly reduced time to recovery relative to placebo. Subsequent trials provided additional support for the efficacy of remdesivir in hospitalized patients with moderate or severe COVID-19, with a greater benefit seen in patients with minimal oxygen requirements at baseline. Clinical trials also demonstrated the efficacy of remdesivir in other patient populations, including outpatients at high risk for progression to severe COVID-19, as well as hospitalized paediatric patients. In terms of mortality, results were equivocal. However, remdesivir appeared to have a small mortality benefit in hospitalized patients who were not already being ventilated at baseline. Remdesivir was generally well tolerated in clinical trials, but pharmacovigilance data found an increased risk of hepatic, renal and cardiovascular adverse drug reactions in the real-world setting. In conclusion, remdesivir represents a useful treatment option for patients with COVID-19, particularly those who require supplemental oxygen.
Collapse
Affiliation(s)
- Hannah A Blair
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
13
|
Phan T, Brozak S, Pell B, Oghuan J, Gitter A, Hu T, Ribeiro RM, Ke R, Mena KD, Perelson AS, Kuang Y, Wu F. Making waves: Integrating wastewater surveillance with dynamic modeling to track and predict viral outbreaks. WATER RESEARCH 2023; 243:120372. [PMID: 37494742 DOI: 10.1016/j.watres.2023.120372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/10/2023] [Accepted: 07/15/2023] [Indexed: 07/28/2023]
Abstract
Wastewater surveillance has proved to be a valuable tool to track the COVID-19 pandemic. However, most studies using wastewater surveillance data revolve around establishing correlations and lead time relative to reported case data. In this perspective, we advocate for the integration of wastewater surveillance data with dynamic within-host and between-host models to better understand, monitor, and predict viral disease outbreaks. Dynamic models overcome emblematic difficulties of using wastewater surveillance data such as establishing the temporal viral shedding profile. Complementarily, wastewater surveillance data bypasses the issues of time lag and underreporting in clinical case report data, thus enhancing the utility and applicability of dynamic models. The integration of wastewater surveillance data with dynamic models can enhance real-time tracking and prevalence estimation, forecast viral transmission and intervention effectiveness, and most importantly, provide a mechanistic understanding of infectious disease dynamics and the driving factors. Dynamic modeling of wastewater surveillance data will advance the development of a predictive and responsive monitoring system to improve pandemic preparedness and population health.
Collapse
Affiliation(s)
- Tin Phan
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, NM 87544, USA
| | - Samantha Brozak
- School of Mathematical and Statistical Sciences, Arizona State University, AZ 85281, USA
| | - Bruce Pell
- Department of Mathematics and Computer Science, Lawrence Technological University, MI 48075, USA
| | - Jeremiah Oghuan
- School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Anna Gitter
- School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Tao Hu
- Department of Geography, Oklahoma State University, Stillwater, OK 74078, USA
| | - Ruy M Ribeiro
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, NM 87544, USA
| | - Ruian Ke
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, NM 87544, USA
| | - Kristina D Mena
- School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Texas Epidemic Public Health Institute, Houston, TX 77030, USA
| | - Alan S Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, NM 87544, USA; Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Yang Kuang
- School of Mathematical and Statistical Sciences, Arizona State University, AZ 85281, USA
| | - Fuqing Wu
- School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Texas Epidemic Public Health Institute, Houston, TX 77030, USA.
| |
Collapse
|
14
|
Okugawa S, Ikeda M, Kashiwabara K, Moritoyo T, Kohsaka T, Shimizu T, Hagiya H, Hasegawa K, Otsuka F, Miwa A, Kisimoto N, Mizoguchi A, Imamura A, Ikeuchi K, Tsutsumi T, Jubishi D, Hashimoto H, Okamoto K, Harada S, Inoue JI, Seto Y, Moriya K. Antiviral effect and safety of nafamostat mesilate in patients with mild early-onset COVID-19: An exploratory multicentre randomized controlled clinical trial. Int J Antimicrob Agents 2023; 62:106922. [PMID: 37429450 DOI: 10.1016/j.ijantimicag.2023.106922] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/03/2023] [Accepted: 07/05/2023] [Indexed: 07/12/2023]
Abstract
OBJECTIVES This study aimed to evaluate the antiviral effects and safety of nafamostat in early-onset patients with coronavirus disease 2019 (COVID-19). METHODS In this exploratory multicentre randomized controlled trial, patients were assigned to three groups within 5 days of symptom onset, with 10 participants in each group: nafamostat at either 0.2 mg/kg/h or 0.1 mg/kg/h or a standard-of-care group. The primary endpoint was area under the curve for decrease in SARS-CoV-2 viral load in nasopharyngeal samples from baseline to day 6. RESULTS Of the 30 randomized patients, 19 received nafamostat. Overall, 10 patients received low-dose nafamostat, 9 patients received high-dose nafamostat, and 10 received standard-of-care. The detected viruses were Omicron strains. The regression coefficient for area under the curve for decrease in viral load as the response variable and nafamostat dose per body weight as the explanatory variable showed a significant relationship of -40.1 (95% confidence interval, -74.1 to -6.2; P = 0.022). Serious adverse events were not observed in either group. Phlebitis occurred in ca. 50% of patients treated with nafamostat. CONCLUSIONS Nafamostat exerts virus load-reducing effects in patients with early-onset COVID-19.
Collapse
Affiliation(s)
- Shu Okugawa
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan
| | - Mahoko Ikeda
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan; Department of Infection Control and Prevention, The University of Tokyo Hospital, Tokyo, Japan
| | - Kosuke Kashiwabara
- Clinical Research Promotion Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Takashi Moritoyo
- Clinical Research Promotion Center, The University of Tokyo Hospital, Tokyo, Japan
| | - Takao Kohsaka
- Department of Surgery, Horinouchi Hospital, Saitama, Japan
| | - Toshio Shimizu
- Department of Surgery, Horinouchi Hospital, Saitama, Japan
| | - Hideharu Hagiya
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kou Hasegawa
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Fumio Otsuka
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ayumi Miwa
- Pharmaceutical Department, Takamatsu Municipal Hospital, Kagawa, Japan
| | - Nobuhito Kisimoto
- Department of Pulmonary Medicine, Takamatsu Municipal Hospital, Kagawa, Japan
| | - Ayako Mizoguchi
- Pharmaceutical Department, Moriya Keiyu Hospital, Ibaraki, Japan
| | - Akira Imamura
- Department of Internal medicine, Moriya Keiyu Hospital, Ibaraki, Japan
| | - Kazuhiko Ikeuchi
- Division of Infectious Diseases, Advanced Clinical Research Center, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takeya Tsutsumi
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan; Division of Infectious Diseases, Advanced Clinical Research Center, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Daisuke Jubishi
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan
| | - Hideki Hashimoto
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan
| | - Koh Okamoto
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan
| | - Sohei Harada
- Department of Infection Control and Prevention, The University of Tokyo Hospital, Tokyo, Japan
| | - Jun-Ichiro Inoue
- The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), Tokyo, Japan
| | - Yasuyuki Seto
- Department of Stomach and Esophageal Surgery, The University of Tokyo Hospital, Tokyo, Japan
| | - Kyoji Moriya
- Department of Infectious Diseases, The University of Tokyo Hospital, Tokyo, Japan; Department of Infection Control and Prevention, The University of Tokyo Hospital, Tokyo, Japan.
| |
Collapse
|
15
|
Chevret S, Bouadma L, Dupuis C, Burdet C, Timsit JF. Which severe COVID-19 patients could benefit from high dose dexamethasone? A Bayesian post-hoc reanalysis of the COVIDICUS randomized clinical trial. Ann Intensive Care 2023; 13:75. [PMID: 37634234 PMCID: PMC10460760 DOI: 10.1186/s13613-023-01168-z] [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: 05/08/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND The respective benefits of high and low doses of dexamethasone (DXM) in patients with severe acute respiratory syndrome coronavirus 2 (SARS-Cov2) and acute respiratory failure (ARF) are controversial, with two large triple-blind RCTs reaching very important difference in the effect-size. In the COVIDICUS trial, no evidence of additional benefit of high-dose dexamethasone (DXM20) was found. We aimed to explore whether some specific patient phenotypes could benefit from DXM20 compared to the standard of care 6 mg dose of DXM (DXMSoC). METHODS We performed a post hoc exploratory Bayesian analysis of 473 patients who received either DXMSoc or DXM20 in the COVIDICUS trial. The outcome was the 60 day mortality rate of DXM20 over DXMSoC, with treatment effect measured on the hazard ratio (HR) estimated from Cox model. Bayesian analyses allowed to compute the posterior probability of a more than trivial benefit (HR < 0.95), and that of a potential harm (HR > 1.05). Bayesian measures of interaction then quantified the probability of interaction (Pr Interact) that the HR of death differed across the subsets by 20%. Primary analyses used noninformative priors, centred on HR = 1.00. Sensitivity analyses used sceptical and enthusiastic priors, based on null (HR = 1.00) or benefit (HR = 0.95) effects. RESULTS Overall, the posterior probability of a more than trivial benefit and potential harm was 29.0 and 51.1%, respectively. There was some evidence of treatment by subset interaction (i) according to age (Pr Interact, 84%), with a 86.5% probability of benefit in patients aged below 70 compared to 22% in those aged above 70; (ii) according to the time since symptoms onset (Pr Interact, 99%), with a 99.9% probability of a more than trivial benefit when lower than 7 days compared to a < 0.1% probability when delayed by 7 days or more; and (iii) according to use of remdesivir (Pr Interact, 91%), with a 90.1% probability of benefit in patients receiving remdesivir compared to 19.1% in those who did not. CONCLUSIONS In this exploratory post hoc Bayesian analysis, compared with standard-of-care DXM, high-dose DXM may benefit patients aged less than 70 years with severe ARF that occurred less than 7 days after symptoms onset. The use of remdesivir may also favour the benefit of DXM20. Further analysis is needed to confirm these findings. TRIAL REGISTRATION NCT04344730, date of registration April 14, 2020 ( https://clinicaltrials.gov/ct2/show/NCT04344730?term=NCT04344730&draw=2&rank=1 ); EudraCT: 2020-001457-43 ( https://www.clinicaltrialsregister.eu/ctr-search/search?query=2020-001457-43 ).
Collapse
Affiliation(s)
- Sylvie Chevret
- ECSTRRA, UMR 1153, Saint Louis Hospital, University Paris Cité, Paris, France
| | - Lila Bouadma
- Medical and Infectious Diseases ICU, APHP Bichat Hospital, 75018, Paris, France
- Université Paris Cité, IAME, INSERM, UMR 1137, 75018, Paris, France
| | - Claire Dupuis
- Université Paris Cité, IAME, INSERM, UMR 1137, 75018, Paris, France
- Intensive Care Unit, Gabriel Montpied Hospital, CHU de Clermont-Ferrand, 63000, Clermont-Ferrand, France
| | - Charles Burdet
- Université Paris Cité, IAME, INSERM, UMR 1137, 75018, Paris, France
- Epidemiology, Biostatistics and Clinical Research Department, AP-HP, Bichat Hospital, 75018, Paris, France
| | - Jean-François Timsit
- Medical and Infectious Diseases ICU, APHP Bichat Hospital, 75018, Paris, France.
- Université Paris Cité, IAME, INSERM, UMR 1137, 75018, Paris, France.
| |
Collapse
|
16
|
McCormack CP, Yan AWC, Brown JC, Sukhova K, Peacock TP, Barclay WS, Dorigatti I. Modelling the viral dynamics of the SARS-CoV-2 Delta and Omicron variants in different cell types. J R Soc Interface 2023; 20:20230187. [PMID: 37553993 PMCID: PMC10410224 DOI: 10.1098/rsif.2023.0187] [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: 03/30/2023] [Accepted: 07/18/2023] [Indexed: 08/10/2023] Open
Abstract
We use viral kinetic models fitted to viral load data from in vitro studies to explain why the SARS-CoV-2 Omicron variant replicates faster than the Delta variant in nasal cells, but slower than Delta in lung cells, which could explain Omicron's higher transmission potential and lower severity. We find that in both nasal and lung cells, viral infectivity is higher for Omicron but the virus production rate is higher for Delta, with an estimated approximately 200-fold increase in infectivity and 100-fold decrease in virus production when comparing Omicron with Delta in nasal cells. However, the differences are unequal between cell types, and ultimately lead to the basic reproduction number and growth rate being higher for Omicron in nasal cells, and higher for Delta in lung cells. In nasal cells, Omicron alone can enter via a TMPRSS2-independent pathway, but it is primarily increased efficiency of TMPRSS2-dependent entry which accounts for Omicron's increased activity. This work paves the way for using within-host mathematical models to understand the transmission potential and severity of future variants.
Collapse
Affiliation(s)
- Clare P. McCormack
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Ada W. C. Yan
- Department of Infectious Disease, Imperial College London, London, UK
| | - Jonathan C. Brown
- Department of Infectious Disease, Imperial College London, London, UK
| | - Ksenia Sukhova
- Department of Infectious Disease, Imperial College London, London, UK
| | - Thomas P. Peacock
- Department of Infectious Disease, Imperial College London, London, UK
| | - Wendy S. Barclay
- Department of Infectious Disease, Imperial College London, London, UK
| | - Ilaria Dorigatti
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
17
|
Th-1, Th-2, Th-9, Th-17, Th-22 type cytokine concentrations of critical COVID-19 patients after treatment with Remdesivir. Immunobiology 2023; 228:152378. [PMID: 37058846 PMCID: PMC10036294 DOI: 10.1016/j.imbio.2023.152378] [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: 11/23/2022] [Revised: 02/22/2023] [Accepted: 03/20/2023] [Indexed: 03/26/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) rapidly spread around the world causing a pandemic known as coronavirus disease 2019 (COVID-19). Cytokine storm was directly correlated with severity of COVID-19 syndromes. We evaluated the levels of 13 cytokines in ICU hospitalized COVID-19 patients (n=29) before, and after treatment with Remdesivir as well as in healthy controls (n=29). Blood samples were obtained from ICU patients during ICU admission (before treatment) and 5 days after treatment with Remdesivir. A group of 29 age- and gender-matched healthy controls was also studied. Cytokine levels were evaluated by multiplex immunoassay method using a fluorescence labeled cytokine panel. In comparison to cytokine levels measured at ICU admission, serum levels were reduced of IL-6 (134.75 pg/mL vs. 20.73 pg/mL, P< 0.0001), TNF-α (121.67 pg/mL vs. 10.15 pg/mL, P< 0.0001) and IFN-γ (29.69 pg/mL vs. 22.27 pg/mL, P= 0.005), whereas serum level was increased of IL-4 (8.47 pg/mL vs. 12.44 pg/mL, P= 0.002) within 5 days after Remdesivir treatment. Comparing with before treatment, Remdesivir significantly reduced the levels of inflammatory (258.98 pg/mL vs. 37.43 pg/mL, P< 0.0001), Th1-type (31.24 pg/mL vs. 24.46 pg/mL, P= 0.007), and Th17-type (36.79 pg/mL vs. 26.22 pg/mL, P< 0.0001) cytokines in critical COVID-19 patients. However, after Remdesivir treatment, the concentrations of Th2-type cytokines were significantly higher than before treatment (52.69 pg/mL vs. 37.09 pg/mL, P< 0.0001). In conclusion, Remdesivir led to decrease levels of Th1-type and Th17-type cytokines and increase Th2-type cytokines in critical COVID-19 patients 5 days after treatment.
Collapse
|
18
|
Amstutz A, Speich B, Mentré F, Rueegg CS, Belhadi D, Assoumou L, Burdet C, Murthy S, Dodd LE, Wang Y, Tikkinen KAO, Ader F, Hites M, Bouscambert M, Trabaud MA, Fralick M, Lee TC, Pinto R, Barratt-Due A, Lund-Johansen F, Müller F, Nevalainen OPO, Cao B, Bonnett T, Griessbach A, Taji Heravi A, Schönenberger C, Janiaud P, Werlen L, Aghlmandi S, Schandelmaier S, Yazdanpanah Y, Costagliola D, Olsen IC, Briel M. Effects of remdesivir in patients hospitalised with COVID-19: a systematic review and individual patient data meta-analysis of randomised controlled trials. THE LANCET. RESPIRATORY MEDICINE 2023; 11:453-464. [PMID: 36828006 PMCID: PMC10156140 DOI: 10.1016/s2213-2600(22)00528-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 02/24/2023]
Abstract
BACKGROUND Interpretation of the evidence from randomised controlled trials (RCTs) of remdesivir in patients treated in hospital for COVID-19 is conflicting. We aimed to assess the benefits and harms of remdesivir compared with placebo or usual care in these patients, and whether treatment effects differed between prespecified patient subgroups. METHODS For this systematic review and meta-analysis, we searched PubMed, Embase, the Cochrane COVID-19 trial registry, ClinicalTrials.gov, the International Clinical Trials Registry Platform, and preprint servers from Jan 1, 2020, until April 11, 2022, for RCTs of remdesivir in adult patients hospitalised with COVID-19, and contacted the authors of eligible trials to request individual patient data. The primary outcome was all-cause mortality at day 28 after randomisation. We used multivariable hierarchical regression-adjusting for respiratory support, age, and enrollment period-to investigate effect modifiers. This study was registered with PROSPERO, CRD42021257134. FINDINGS Our search identified 857 records, yielding nine RCTs eligible for inclusion. Of these nine eligible RCTs, individual data were provided for eight, covering 10 480 patients hospitalised with COVID-19 (99% of such patients included in such RCTs worldwide) recruited between Feb 6, 2020, and April 1, 2021. Within 28 days of randomisation, 662 (12·5%) of 5317 patients assigned to remdesivir and 706 (14·1%) of 5005 patients assigned to no remdesivir died (adjusted odds ratio [aOR] 0·88, 95% CI 0·78-1·00, p=0·045). We found evidence for a credible subgroup effect according to respiratory support at baseline (pinteraction=0·019). Of patients who were ventilated-including those who received high-flow oxygen-253 (30·0%) of 844 patients assigned to remdesivir died compared with 241 (28·5%) of 846 patients assigned to no remdesivir (aOR 1·10 [0·88-1·38]; low-certainty evidence). Of patients who received no oxygen or low-flow oxygen, 409 (9·1%) of 4473 patients assigned to remdesivir died compared with 465 (11·2%) of 4159 patients assigned to no remdesivir (0·80 [0·70-0·93]; high-certainty evidence). No credible subgroup effect was found for time to start of remdesivir after symptom onset, age, presence of comorbidities, enrolment period, or corticosteroid use. Remdesivir did not increase the frequency of severe or serious adverse events. INTERPRETATION This individual patient data meta-analysis showed that remdesivir reduced mortality in patients hospitalised with COVID-19 who required no or conventional oxygen support, but was underpowered to evaluate patients who were ventilated when receiving remdesivir. The effect size of remdesivir in patients with more respiratory support or acquired immunity and the cost-effectiveness of remdesivir remain to be further elucidated. FUNDING EU-RESPONSE.
Collapse
Affiliation(s)
- Alain Amstutz
- CLEAR Methods Center, Division of Clinical Epidemiology, University Hospital Basel, Basel, Switzerland; Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Benjamin Speich
- CLEAR Methods Center, Division of Clinical Epidemiology, University Hospital Basel, Basel, Switzerland; Department of Clinical Research, University Hospital Basel, Basel, Switzerland.
| | - France Mentré
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, Paris, France; Département d'Épidémiologie, Biostatistique et Recherche Clinique, Hôpital Bichat, AP-HP, Paris, France
| | - Corina Silvia Rueegg
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Drifa Belhadi
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, Paris, France; Département d'Épidémiologie, Biostatistique et Recherche Clinique, Hôpital Bichat, AP-HP, Paris, France
| | - Lambert Assoumou
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, Paris, France
| | - Charles Burdet
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, Paris, France; Département d'Épidémiologie, Biostatistique et Recherche Clinique, Hôpital Bichat, AP-HP, Paris, France
| | - Srinivas Murthy
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Lori Elizabeth Dodd
- Clinical Trials Research Section, Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Yeming Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Kari A O Tikkinen
- Department of Urology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Surgery, South Karelian Central Hospital, Lappeenranta, Finland
| | - Florence Ader
- Département des Maladies Infectieuses et Tropicales, Hospices Civils de Lyon, Lyon, France; Legiopath, Centre International de Recherche en Infectiologie, Inserm 1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, Lyon, France
| | - Maya Hites
- Cliniques Universitaires de Bruxelles Hôpital Érasme, Université Libre de Bruxelles, Clinique des Maladies Infectieuses, Brussels, Belgium
| | - Maude Bouscambert
- Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des Virus Respiratoires France Sud, Hospices Civils de Lyon, Lyon, France
| | - Mary Anne Trabaud
- Laboratoire de Virologie, Institut des Agents Infectieux de Lyon, Centre National de Référence des Virus Respiratoires France Sud, Hospices Civils de Lyon, Lyon, France
| | - Mike Fralick
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Todd C Lee
- Clinical Practice Assessment Unit, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Ruxandra Pinto
- Department of Critical Care Medicine, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Andreas Barratt-Due
- Department of Anesthesia and Intensive Care Medicine, Oslo University Hospital, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Anesthesia and Intensive Care Medicine, Oslo University Hospital, Oslo, Norway
| | - Fredrik Müller
- Department of Anesthesia and Intensive Care Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Bin Cao
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China; National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Tyler Bonnett
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD, USA
| | - Alexandra Griessbach
- CLEAR Methods Center, Division of Clinical Epidemiology, University Hospital Basel, Basel, Switzerland; Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Ala Taji Heravi
- CLEAR Methods Center, Division of Clinical Epidemiology, University Hospital Basel, Basel, Switzerland; Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Christof Schönenberger
- CLEAR Methods Center, Division of Clinical Epidemiology, University Hospital Basel, Basel, Switzerland; Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Perrine Janiaud
- Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Laura Werlen
- Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Soheila Aghlmandi
- CLEAR Methods Center, Division of Clinical Epidemiology, University Hospital Basel, Basel, Switzerland; Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Stefan Schandelmaier
- CLEAR Methods Center, Division of Clinical Epidemiology, University Hospital Basel, Basel, Switzerland; Department of Clinical Research, University Hospital Basel, Basel, Switzerland
| | - Yazdan Yazdanpanah
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, IAME, Paris, France; Service de Maladies Infectieuses et Tropicales, Hôpital Bichat, AP-HP, Paris, France
| | - Dominique Costagliola
- Sorbonne Université, Inserm, Institut Pierre-Louis d'Épidémiologie et de Santé Publique, Paris, France
| | | | - Matthias Briel
- CLEAR Methods Center, Division of Clinical Epidemiology, University Hospital Basel, Basel, Switzerland; Department of Clinical Research, University Hospital Basel, Basel, Switzerland; Department of Health Research Methodology, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
19
|
Grundeis F, Ansems K, Dahms K, Thieme V, Metzendorf MI, Skoetz N, Benstoem C, Mikolajewska A, Griesel M, Fichtner F, Stegemann M. Remdesivir for the treatment of COVID-19. Cochrane Database Syst Rev 2023; 1:CD014962. [PMID: 36695483 PMCID: PMC9875553 DOI: 10.1002/14651858.cd014962.pub2] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Remdesivir is an antiviral medicine approved for the treatment of mild-to-moderate coronavirus disease 2019 (COVID-19). This led to widespread implementation, although the available evidence remains inconsistent. This update aims to fill current knowledge gaps by identifying, describing, evaluating, and synthesising all evidence from randomised controlled trials (RCTs) on the effects of remdesivir on clinical outcomes in COVID-19. OBJECTIVES To assess the effects of remdesivir and standard care compared to standard care plus/minus placebo on clinical outcomes in patients treated for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. SEARCH METHODS We searched the Cochrane COVID-19 Study Register (which comprises the Cochrane Central Register of Controlled Trials (CENTRAL), PubMed, Embase, ClinicalTrials.gov, World Health Organization (WHO) International Clinical Trials Registry Platform, and medRxiv) as well as Web of Science (Science Citation Index Expanded and Emerging Sources Citation Index) and WHO COVID-19 Global literature on coronavirus disease to identify completed and ongoing studies, without language restrictions. We conducted the searches on 31 May 2022. SELECTION CRITERIA We followed standard Cochrane methodology. We included RCTs evaluating remdesivir and standard care for the treatment of SARS-CoV-2 infection compared to standard care plus/minus placebo irrespective of disease severity, gender, ethnicity, or setting. We excluded studies that evaluated remdesivir for the treatment of other coronavirus diseases. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. To assess risk of bias in included studies, we used the Cochrane RoB 2 tool for RCTs. We rated the certainty of evidence using the GRADE (Grading of Recommendations, Assessment, Development and Evaluation) approach for outcomes that were reported according to our prioritised categories: all-cause mortality, in-hospital mortality, clinical improvement (being alive and ready for discharge up to day 28) or worsening (new need for invasive mechanical ventilation or death up to day 28), quality of life, serious adverse events, and adverse events (any grade). We differentiated between non-hospitalised individuals with asymptomatic SARS-CoV-2 infection or mild COVID-19 and hospitalised individuals with moderate to severe COVID-19. MAIN RESULTS We included nine RCTs with 11,218 participants diagnosed with SARS-CoV-2 infection and a mean age of 53.6 years, of whom 5982 participants were randomised to receive remdesivir. Most participants required low-flow oxygen at baseline. Studies were mainly conducted in high- and upper-middle-income countries. We identified two studies that are awaiting classification and five ongoing studies. Effects of remdesivir in hospitalised individuals with moderate to severe COVID-19 With moderate-certainty evidence, remdesivir probably makes little or no difference to all-cause mortality at up to day 28 (risk ratio (RR) 0.93, 95% confidence interval (CI) 0.81 to 1.06; risk difference (RD) 8 fewer per 1000, 95% CI 21 fewer to 6 more; 4 studies, 7142 participants), day 60 (RR 0.85, 95% CI 0.69 to 1.05; RD 35 fewer per 1000, 95% CI 73 fewer to 12 more; 1 study, 1281 participants), or in-hospital mortality at up to day 150 (RR 0.93, 95% CI 0.84 to 1.03; RD 11 fewer per 1000, 95% CI 25 fewer to 5 more; 1 study, 8275 participants). Remdesivir probably increases the chance of clinical improvement at up to day 28 slightly (RR 1.11, 95% CI 1.06 to 1.17; RD 68 more per 1000, 95% CI 37 more to 105 more; 4 studies, 2514 participants; moderate-certainty evidence). It probably decreases the risk of clinical worsening within 28 days (hazard ratio (HR) 0.67, 95% CI 0.54 to 0.82; RD 135 fewer per 1000, 95% CI 198 fewer to 69 fewer; 2 studies, 1734 participants, moderate-certainty evidence). Remdesivir may make little or no difference to the rate of adverse events of any grade (RR 1.04, 95% CI 0.92 to 1.18; RD 23 more per 1000, 95% CI 46 fewer to 104 more; 4 studies, 2498 participants; low-certainty evidence), or serious adverse events (RR 0.84, 95% CI 0.65 to 1.07; RD 44 fewer per 1000, 95% CI 96 fewer to 19 more; 4 studies, 2498 participants; low-certainty evidence). We considered risk of bias to be low, with some concerns for mortality and clinical course. We had some concerns for safety outcomes because participants who had died did not contribute information. Without adjustment, this leads to an uncertain amount of missing values and the potential for bias due to missing data. Effects of remdesivir in non-hospitalised individuals with mild COVID-19 One of the nine RCTs was conducted in the outpatient setting and included symptomatic people with a risk of progression. No deaths occurred within the 28 days observation period. We are uncertain about clinical improvement due to very low-certainty evidence. Remdesivir probably decreases the risk of clinical worsening (hospitalisation) at up to day 28 (RR 0.28, 95% CI 0.11 to 0.75; RD 46 fewer per 1000, 95% CI 57 fewer to 16 fewer; 562 participants; moderate-certainty evidence). We did not find any data for quality of life. Remdesivir may decrease the rate of serious adverse events at up to 28 days (RR 0.27, 95% CI 0.10 to 0.70; RD 49 fewer per 1000, 95% CI 60 fewer to 20 fewer; 562 participants; low-certainty evidence), but it probably makes little or no difference to the risk of adverse events of any grade (RR 0.91, 95% CI 0.76 to 1.10; RD 42 fewer per 1000, 95% CI 111 fewer to 46 more; 562 participants; moderate-certainty evidence). We considered risk of bias to be low for mortality, clinical improvement, and safety outcomes. We identified a high risk of bias for clinical worsening. AUTHORS' CONCLUSIONS Based on the available evidence up to 31 May 2022, remdesivir probably has little or no effect on all-cause mortality or in-hospital mortality of individuals with moderate to severe COVID-19. The hospitalisation rate was reduced with remdesivir in one study including participants with mild to moderate COVID-19. It may be beneficial in the clinical course for both hospitalised and non-hospitalised patients, but certainty remains limited. The applicability of the evidence to current practice may be limited by the recruitment of participants from mostly unvaccinated populations exposed to early variants of the SARS-CoV-2 virus at the time the studies were undertaken. Future studies should provide additional data on the efficacy and safety of remdesivir for defined core outcomes in COVID-19 research, especially for different population subgroups.
Collapse
Affiliation(s)
- Felicitas Grundeis
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Kelly Ansems
- Department of Intensive Care Medicine and Intermediate Care, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Karolina Dahms
- Department of Intensive Care Medicine and Intermediate Care, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Volker Thieme
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Maria-Inti Metzendorf
- Institute of General Practice, Medical Faculty of the Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Nicole Skoetz
- Cochrane Haematology, Department I of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Carina Benstoem
- Department of Intensive Care Medicine and Intermediate Care, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Agata Mikolajewska
- Centre for Biological Threats and Special Pathogens (ZBS), Strategy and Incident Response, Clinical Management and Infection Control, Robert Koch Institute, Berlin, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Mirko Griesel
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Falk Fichtner
- Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany
| | - Miriam Stegemann
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
20
|
Kim MH, Elbaz J, Jilg N, Gustafson JL, Xu M, Hatipoglu D, Nohelty E, Kim AY, Chung RT. Peginterferon lambda for the treatment of hospitalized patients with mild COVID-19: A pilot phase 2 randomized placebo-controlled trial. Front Med (Lausanne) 2023; 10:1095828. [PMID: 36910479 PMCID: PMC10002416 DOI: 10.3389/fmed.2023.1095828] [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: 11/11/2022] [Accepted: 02/08/2023] [Indexed: 02/26/2023] Open
Abstract
Background This study aimed to investigate the efficacy and safety of subcutaneous injection of peginterferon lambda in patients hospitalized with COVID-19. Methods In this study (NCT04343976), patients admitted to hospital with COVID-19 confirmed by RT-PCR from nasopharyngeal swab were randomly assigned within 48 h to receive peginterferon lambda or placebo in a 1:1 ratio. Participants were subcutaneously injected with a peginterferon lambda or saline placebo at baseline and day 7 and were followed up until day 14. Results We enrolled 14 participants; 6 participants (85.7%) in the peginterferon lambda group and 1 participant (14.3%) in the placebo group were treated with remdesivir prior to enrollment. Fifty percent of participants were SARS-CoV-2 RNA negative at baseline although they tested SARS-CoV-2 RNA positive within 48 h of randomization. Among participants who were SARS-CoV-2 positive at baseline, 2 out of 5 participants (40%) in the peginterferon lambda group became negative at day 14, while 0 out of 2 participants (0%) in the placebo group achieved negativity for SARS-CoV-2 by day 14 (p > 0.05). The median change in viral load (log copies per ml) was +1.72 (IQR -2.78 to 3.19) in the placebo group and -2.22 (IQR -3.24 to 0.55) in the peginterferon lambda group at day 14 (p = 0.24). Symptomatic changes did not differ between the two groups. Peginterferon lambda was well tolerated with a few treatment-related adverse effects. Conclusion Peginterferon lambda appears to accelerate SARS-CoV-2 viral load decline and improve plasma disease progression markers in hospitalized patients with COVID-19.
Collapse
Affiliation(s)
- Myung-Ho Kim
- Liver Center, Gastrointestinal Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Department of Internal Korean Medicine, Woosuk University Medical Center, Jeonju, Republic of Korea
| | - Josh Elbaz
- Liver Center, Gastrointestinal Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Department of Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Nikolaus Jilg
- Division of Infectious Disease, Massachusetts General Hospital, Harvard Medical School Boston, Boston, MA, United States.,Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Jenna L Gustafson
- Liver Center, Gastrointestinal Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Min Xu
- Liver Center, Gastrointestinal Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Dilara Hatipoglu
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Eric Nohelty
- Division of Infectious Disease, Massachusetts General Hospital, Harvard Medical School Boston, Boston, MA, United States.,Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Arthur Y Kim
- Division of Infectious Disease, Massachusetts General Hospital, Harvard Medical School Boston, Boston, MA, United States.,Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Raymond T Chung
- Liver Center, Gastrointestinal Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
21
|
McMahon JH, Lau JS, Coldham A, Roney J, Hagenauer M, Price S, Bryant M, Garlick J, Paterson A, Lee SJ, O'Bryan J, Hearps A, Tachedjian G, Pinskier H, Phillips C, Garrow S, Pinskier N, Melvin R, Blakeway L, Wisniewski JA, Byers S, Badoordeen GZ, Pereira S, Pragastis K, Trubiano JA, Chua KY, Kainer M, Molton JS, Gardiner BJ, Pierce AB, Cheng A, Rogers BA, Peleg AY. Favipiravir in early symptomatic COVID-19, a randomised placebo-controlled trial. EClinicalMedicine 2022; 54:101703. [PMID: 36284645 PMCID: PMC9583769 DOI: 10.1016/j.eclinm.2022.101703] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/28/2022] [Accepted: 09/28/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Well tolerated antivirals administered early in the course of COVID-19 infection when the viremia is highest could prevent progression to severe disease. Favipiravir inhibits SARS-CoV-2 viral replication in vitro with evidence of clinical benefit in open label trials. Placebo controlled studies of people with early symptomatic COVID-19 with regular assessments of SARS-CoV-2 viral load can determine if it has an antiviral effect and improves clinical outcomes. METHODS People with PCR-confirmed COVID-19 and 5 days or less of symptoms were randomised 1:1 to favipiravir 1800 mg on day 1, then 800 mg twice daily or matched placebo for 14 days. SARS-CoV-2 viral load was quantitated from second daily self-collected nose-throat swabs while receiving study drug. The primary endpoint was time to virological cure defined as 2 successive swabs negative for SARS-CoV-2 by PCR and secondary outcomes were progression of disease severity, symptom resolution and safety. FINDINGS Between 31 July 2020 and 19 September 2021, 200 people were enrolled (199 in the community, 1 in hospital) with 190 receiving one or more doses of drug (modified intention to treat [mITT] population). There was no difference in time to virological cure (Log-rank p=0.6 comparing Kaplan Meier curves), progression to hospitalisation (14 favipiravir, 9 placebo; p=0.38), time to symptom resolution (cough, fever, sore throat) and there were no deaths. 51 people related an adverse event that was possibly drug related, but these were evenly distributed (n=24 favipiravir, n=27 placebo). Sensitivity analyses where the definition of virological cure was changed to: a single negative PCR, exclude datapoints based on the presence or absence of human DNA in the swab, a SARS-CoV-2 viral load < 300 copies/mL being considered negative all demonstrated no difference between arms. INTERPRETATION Favipiravir does not improve the time to virological cure or clinical outcomes and shows no evidence of an antiviral effect when treating early symptomatic COVID-19 infection. FUNDING The study was supported in part by grants from the Commonwealth Bank Australia, the Lord Mayor's Charitable Foundation, Melbourne Australia and the Orloff Family Charitable Trust, Melbourne, Australia. JHM is supported by the Medical Research Future Fund, AYP, JT are supported by the Australian National Health and Medical Research Council.
Collapse
Affiliation(s)
- James H. McMahon
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
- Department of Infectious Diseases, Monash Medical Centre, Melbourne, Australia
- Corresponding author at: Department of Infectious Diseases, The Alfred, Level 2, Burnet Institute, 85 Commercial Rd, Melbourne, Victoria 3004, Australia.
| | - Jillian S.Y. Lau
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
- Department of Infectious Diseases, Monash Medical Centre, Melbourne, Australia
- Department of Infectious Diseases, Eastern Health, Melbourne, Australia
| | - Anna Coldham
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Janine Roney
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Michelle Hagenauer
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Sally Price
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Mellissa Bryant
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Jill Garlick
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Anne Paterson
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Sue J. Lee
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Jess O'Bryan
- Department of Infectious Diseases, Monash Medical Centre, Melbourne, Australia
| | - Anna Hearps
- Life Sciences Discipline, Burnet Institute, Melbourne, Australia
| | - Gilda Tachedjian
- Life Sciences Discipline, Burnet Institute, Melbourne, Australia
- Department of Microbiology, Monash University, Clayton, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Australia
| | | | | | | | | | - Robert Melvin
- Department of Emergency Medicine, Alfred Hospital, Melbourne, Australia
| | - Luke Blakeway
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Jessica A. Wisniewski
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Sally Byers
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Gnei Z. Badoordeen
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Stephanie Pereira
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Katherine Pragastis
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Jason A. Trubiano
- Department of Infectious Diseases, Austin Hospital, Melbourne, Australia & Department of Medicine, University of Melbourne
| | - Kyra Y.L. Chua
- Department of Infectious Diseases, Austin Hospital, Melbourne, Australia & Department of Medicine, University of Melbourne
| | - Marion Kainer
- Department of Infectious Diseases, Western Health, Melbourne, Australia
| | - James S. Molton
- Department of Infectious Diseases, Western Health, Melbourne, Australia
| | - Bradley J. Gardiner
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
- Epworth Healthcare, Melbourne, Australia
| | - Anna B. Pierce
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
- Department of Infectious Diseases, Monash Medical Centre, Melbourne, Australia
- South East Public Health Unit, Monash Health, Melbourne, Australia
| | - Allen Cheng
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Benjamin A. Rogers
- Department of Infectious Diseases, Monash Medical Centre, Melbourne, Australia
- Monash University School of Clinical Sciences at Monash Health, Clayton, Australia
| | - Anton Y. Peleg
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
- Infection Theme, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, Australia
| |
Collapse
|
22
|
Spagnuolo V, Voarino M, Tonelli M, Galli L, Poli A, Bruzzesi E, Racca S, Clementi N, Oltolini C, Tresoldi M, Rovere Querini P, Dagna L, Zangrillo A, Ciceri F, Clementi M, Castagna A. Impact of Remdesivir on SARS-CoV-2 Clearance in a Real-Life Setting: A Matched-Cohort Study. Drug Des Devel Ther 2022; 16:3645-3654. [PMID: 36268521 PMCID: PMC9578770 DOI: 10.2147/dddt.s369473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/26/2022] [Indexed: 11/23/2022] Open
Abstract
Background Evidence regarding the impact of remdesivir (RDV) on SARS-CoV-2 viral clearance (VC) is scarce. The aim of this study was to compare VC timing in hospitalized COVID-19 patients who did or did not receive RDV. Methods This was a matched-cohort study of patients hospitalized with pneumonia, a SARS-CoV-2-positive nasopharyngeal swab (NPS) at admission, and at least one NPS during follow-up. Patients who received RDV (cases) and those who did not (controls) were matched in a 1:2 ratio by age, sex, and PaO2/FiO2 (P/F) values at admission. NPSs were analyzed using real-time polymerase chain reaction. Time to VC (within 30 days after hospital discharge) was estimated using the Kaplan-Meier curve. A multivariable Cox proportional hazard model was fitted to determine factors associated with VC. Results There were 648 patients enrolled in the study (216 cases and 432 controls). VC was observed in 490 patients (75.6%), with a median time of 25 (IQR 16-34) days. Overall, time to VC was similar between cases and controls (p = 0.519). However, time to VC was different when considering both RDV treatment status and age (p = 0.007). A significant finding was also observed when considering both RDV treatment status and P/F values at admission (p = 0.007). A multivariate analysis showed that VC was associated with a younger age (aHR = 0.990, 95% CI 0.983-0.998 per every 10-year increase in age; p = 0.009) and a higher baseline P/F ratio (aHR=1.275, 95% CI 1.029-1.579; p=0.026), but not with RDV treatment status. Conclusion Time to VC was similar in cases and controls. However, there was a benefit associated with using RDV in regard to time to VC in younger patients and in those with a P/F ratio ≤200 mmHg at hospital admission.
Collapse
Affiliation(s)
- Vincenzo Spagnuolo
- Unit of Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Scientific Institute, Milan, Italy,Correspondence: Vincenzo Spagnuolo, Unit of Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Scientific Institute, Milan, Italy, Tel +390226437907, Fax +390226437903, Email
| | - Marta Voarino
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Marco Tonelli
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy,Unit of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Galli
- Unit of Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Poli
- Unit of Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Scientific Institute, Milan, Italy
| | - Elena Bruzzesi
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Sara Racca
- Unit of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nicola Clementi
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy,Unit of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Oltolini
- Unit of Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Scientific Institute, Milan, Italy
| | - Moreno Tresoldi
- General Medicine and Advanced Care Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Patrizia Rovere Querini
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy,Internal Medicine, Diabetes, and Endocrinology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy,Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Zangrillo
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy,Anesthesia and Intensive Care Department, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy,Hematology and Bone Marrow Transplant Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Clementi
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy,Unit of Microbiology and Virology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonella Castagna
- Unit of Infectious Diseases, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), San Raffaele Scientific Institute, Milan, Italy,Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | | |
Collapse
|
23
|
Battaglini D, Cruz F, Robba C, Pelosi P, Rocco PRM. Failed clinical trials on COVID-19 acute respiratory distress syndrome in hospitalized patients: common oversights and streamlining the development of clinically effective therapeutics. Expert Opin Investig Drugs 2022; 31:995-1015. [PMID: 36047644 DOI: 10.1080/13543784.2022.2120801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION The coronavirus disease 2019 (COVID-19) pandemic has put a strain on global healthcare systems. Despite admirable efforts to develop rapidly new pharmacotherapies, supportive treatments remain the standard of care. Multiple clinical trials have failed due to design issues, biased patient enrollment, small sample sizes, inadequate control groups, and lack of long-term outcomes monitoring. AREAS COVERED This narrative review depicts the current situation around failed and success COVID-19 clinical trials and recommendations in hospitalized patients with COVID-19, oversights and streamlining of clinically effective therapeutics. PubMed, EBSCO, Cochrane Library, and WHO and NIH guidelines were searched for relevant literature up to 5 August 2022. EXPERT OPINION The WHO, NIH, and IDSA have issued recommendations to better clarify which drugs should be used during the different phases of the disease. Given the biases and high heterogeneity of published studies, interpretation of the current literature is difficult. Future clinical trials should be designed to standardize clinical approaches, with appropriate organization, patient selection, addition of control groups, and careful identification of disease phase to reduce heterogeneity and bias and should rely on the integration of scientific societies to promote a consensus on interpretation of the data and recommendations for optimal COVID-19 therapies.
Collapse
Affiliation(s)
- Denise Battaglini
- Dipartimento di Anestesia e Rianimazione, Policlinico San Martino, IRCCS per l'Oncologia e le Neuroscienze, Genoa, Italy
| | - Fernanda Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Chiara Robba
- Policlinico San Martino, IRCCS per l'Oncologia e Neuroscienze, Dipartimento di Scienze Chirurgiche e Diagnostiche Integrate, Università degli Studi di Genova, Genoa, Italy
| | - Paolo Pelosi
- Dipartimento di Anestesia e Rianimazione, Policlinico San Martino, IRCCS per l'Oncologia e le Neuroscienze, Genoa, Italy.,Policlinico San Martino, IRCCS per l'Oncologia e Neuroscienze, Dipartimento di Scienze Chirurgiche e Diagnostiche Integrate, Università degli Studi di Genova, Genoa, Italy
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,COVID-19 Virus Network from Ministry of Science, Technology, and Innovation, Brazilian Council for Scientific and Technological Development, and Foundation Carlos Chagas Filho Research Support of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Risk Factors for Slow Viral Decline in COVID-19 Patients during the 2022 Omicron Wave. Viruses 2022; 14:v14081714. [PMID: 36016336 PMCID: PMC9412339 DOI: 10.3390/v14081714] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 02/01/2023] Open
Abstract
Formulating termination of isolation (de-isolation) policies requires up-to-date knowledge about viral shedding dynamics. However, current de-isolation policies are largely based on viral load data obtained before the emergence of Omicron variant. In this retrospective cohort study involving adult patients hospitalised for COVID-19 between January and February 2022, we sought to determine SARS-CoV-2 viral shedding kinetics and to investigate the risk factors associated with slow viral decline during the 2022 Omicron wave. A total of 104 patients were included. The viral load was highest (Ct value was lowest) on days 1 post-symptom-onset (PSO) and gradually declined. Older age, hypertension, hyperlipidaemia and chronic kidney disease were associated with slow viral decline in the univariate analysis on both day 7 and day 10 PSO, while incomplete or no vaccination was associated with slow viral decline on day 7 PSO only. However, older age was the only risk factor that remained statistically significant in the multivariate analysis. In conclusion, older age is an independent risk factor associated with slow viral decline in this study conducted during the Omicron-dominant 2022 COVID-19 wave. Transmission-based precaution guidelines should take age into consideration when determining the timing of de-isolation.
Collapse
|
25
|
Battaglini D, Robba C, Pelosi P, Rocco PRM. Treatment for acute respiratory distress syndrome in adults: A narrative review of phase 2 and 3 trials. Expert Opin Emerg Drugs 2022; 27:187-209. [PMID: 35868654 DOI: 10.1080/14728214.2022.2105833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Ventilatory management and general supportive care of acute respiratory distress syndrome (ARDS) in the adult population have led to significant clinical improvements, but morbidity and mortality remain high. Pharmacologic strategies acting on the coagulation cascade, inflammation, oxidative stress, and endothelial cell injury have been targeted in the last decade for patients with ARDS, but only a few of these have shown potential benefits with a meaningful clinical response and improved patient outcomes. The lack of availability of specific pharmacologic treatments for ARDS can be attributed to its complex pathophysiology, different risk factors, huge heterogeneity, and difficult classification into specific biological phenotypes and genotypes. AREAS COVERED In this narrative review, we briefly discuss the relevance and current advances in pharmacologic treatments for ARDS in adults and the need for the development of new pharmacological strategies. EXPERT OPINION Identification of ARDS phenotypes, risk factors, heterogeneity, and pathophysiology may help to design clinical trials personalized according to ARDS-specific features, thus hopefully decreasing the rate of failed clinical pharmacologic trials. This concept is still under clinical investigation and needs further development.
Collapse
Affiliation(s)
- Denise Battaglini
- Dipartimento di Anestesia e Rianimazione, Policlinico San Martino, IRCCS per l'Oncologia e le Neuroscienze, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Chiara Robba
- Dipartimento di Anestesia e Rianimazione, Policlinico San Martino, IRCCS per l'Oncologia e le Neuroscienze, Largo Rosanna Benzi, 10, 16132, Genoa, Italy.,Dipartimento di Scienze Chirurgiche e Diagnostiche Integrate, Università degli Studi di Genova, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Paolo Pelosi
- Dipartimento di Anestesia e Rianimazione, Policlinico San Martino, IRCCS per l'Oncologia e le Neuroscienze, Largo Rosanna Benzi, 10, 16132, Genoa, Italy.,Dipartimento di Scienze Chirurgiche e Diagnostiche Integrate, Università degli Studi di Genova, Largo Rosanna Benzi, 10, 16132, Genoa, Italy
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, 373, Bloco G1-014, Ilha do Fundão, Rio de Janeiro, RJ 21941-902, Brazil.,COVID-19 Virus Network from Ministry of Science, Technology, and Innovation, Brazilian Council for Scientific and Technological Development, and Foundation Carlos Chagas Filho Research Support of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
26
|
Characterizing SARS-CoV-2 Viral Clearance Kinetics to Improve the Design of Antiviral Pharmacometric Studies. Antimicrob Agents Chemother 2022; 66:e0019222. [PMID: 35736134 PMCID: PMC9295592 DOI: 10.1128/aac.00192-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A consensus methodology for the pharmacometric assessment of candidate SARS-CoV-2 antiviral drugs would be useful for comparing trial results and improving trial design. The time to viral clearance, assessed by serial qPCR of nasopharyngeal swab samples, has been the most widely reported measure of virological response in clinical trials, but it has not been compared formally with other metrics, notably model-based estimates of the rate of viral clearance. We analyzed prospectively gathered viral clearance profiles from 280 infection episodes in vaccinated and unvaccinated individuals. We fitted different phenomenological pharmacodynamic models (single exponential decay, bi-exponential, penalized splines) and found that the clearance rate, estimated from a mixed effects single exponential decay model, is a robust pharmacodynamic summary of viral clearance. The rate of viral clearance, estimated from viral densities during the first week following peak viral load, provides increased statistical power (reduced type 2 error) compared with time to clearance. Antiviral effects approximately equivalent to those with currently used and recommended SARS-CoV-2 antiviral treatments, notably nirmatrelvir and molnupiravir, can be detected from randomized trials with sample sizes of only 35 to 65 patients per arm. We recommend that pharmacometric antiviral assessments should be conducted in early COVID-19 illness with serial qPCR samples taken over 1 week.
Collapse
|
27
|
Merches K, Breunig L, Fender J, Brand T, Bätz V, Idel S, Kollipara L, Reinders Y, Sickmann A, Mally A, Lorenz K. The potential of remdesivir to affect function, metabolism and proliferation of cardiac and kidney cells in vitro. Arch Toxicol 2022; 96:2341-2360. [PMID: 35579693 PMCID: PMC9110936 DOI: 10.1007/s00204-022-03306-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/27/2022] [Indexed: 12/13/2022]
Abstract
Remdesivir is a prodrug of a nucleoside analog and the first antiviral therapeutic approved for coronavirus disease. Recent cardiac safety concerns and reports on remdesivir-related acute kidney injury call for a better characterization of remdesivir toxicity and understanding of the underlying mechanisms. Here, we performed an in vitro toxicity assessment of remdesivir around clinically relevant concentrations (Cmax 9 µM) using H9c2 rat cardiomyoblasts, neonatal mouse cardiomyocytes (NMCM), rat NRK-52E and human RPTEC/TERT1 cells as cell models for the assessment of cardiotoxicity or nephrotoxicity, respectively. Due to the known potential of nucleoside analogs for the induction of mitochondrial toxicity, we assessed mitochondrial function in response to remdesivir treatment, early proteomic changes in NMCM and RPTEC/TERT1 cells and the contractile function of NMCM. Short-term treatments (24 h) of H9c2 and NRK-52E cells with remdesivir adversely affected cell viability by inhibition of proliferation as determined by significantly decreased 3H-thymidine uptake. Mitochondrial toxicity of remdesivir (1.6–3.1 µM) in cardiac cells was evident by a significant decrease in oxygen consumption, a collapse of mitochondrial membrane potential and an increase in lactate secretion after a 24–48-h treatment. This was supported by early proteomic changes of respiratory chain proteins and intermediate filaments that are typically involved in mitochondrial reorganization. Functionally, an impedance-based analysis showed that remdesivir (6.25 µM) affected the beat rate and contractility of NMCM. In conclusion, we identified adverse effects of remdesivir in cardiac and kidney cells at clinically relevant concentrations, suggesting a careful evaluation of therapeutic use in patients at risk for cardiovascular or kidney disease.
Collapse
Affiliation(s)
- Katja Merches
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany.,Landesamt für Gesundheit und Lebensmittelsicherheit (LGL), Erlangen, Germany
| | - Leonie Breunig
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Julia Fender
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Theresa Brand
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Vanessa Bätz
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Svenja Idel
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | | | - Yvonne Reinders
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany.,Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, Scotland, UK.,Medizinische Fakultät, Medizinisches Proteom-Center (MPC), Ruhr-Universität Bochum, Bochum, Germany
| | - Angela Mally
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany. .,Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany. .,PGS Toxicology and Environmental Protection, University of Leipzig, Johannisallee 28, Leipzig, Germany.
| |
Collapse
|