1
|
Çelik G, Karaoğlu ŞA, Suyabatmaz Ş, Bozdeveci A, Yılmaz GT, Yaylı N, Akpınar R, Çiçek AÇ. Synthesis, biological evaluation and molecular docking studies of flavonol-3-O-β-D-glycoside as a potential inhibitor of SARS-CoV-2 main protease (3CLpro) in drug development for COVID-19. Int J Biol Macromol 2025; 298:139621. [PMID: 39818399 DOI: 10.1016/j.ijbiomac.2025.139621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 01/18/2025]
Abstract
The COVID-19 pandemic began in March 2020 and has affected many countries and infected over a million people. It has had a serious impact on people's physical and mental health, daily life and the global economy. Today, many drugs show limited efficacy in the treatment of COVID-19 and studies to develop effective drugs continue. Here, we aim to the synthesise and characterise of the flavonol-3-O-glycoside derivatives, the following and evaluated molecular docking studies with antimicrobial activity, inhibition of SARS-CoV-2 main protease enzyme (3CLpro) and nuclease activity. Molecular docking simulations of the synthesized flavonol-3-O-glycoside derivatives, especially compounds 5a, 5d, 5h, 5i and 5m, showed a stronger interaction with SARS-CoV-2 3CLpro in the active site. Two compounds from the target compounds, 5h and 5m, were found to be specifically effective against M. smegmatis and yeasts. In particular, compounds 5a, 5d, 5h, 5i and 5m, which exhibited high activity against the SARS-CoV-2 main protease enzyme, were found to be effective at low concentrations. We determined the IC50 values for the compounds that showed an inhibitory effect as well as their nuclease activities, which further emphasising the potential of our results. Among these, compound 5d showed a significant competitive inhibitor of 3CLpro. Furthermore, nuclease activity studies identified compound 5d as the most potent. The above results suggest that the flavonol-3-O-glycoside derivatives could be promising new antiviral agents for the development of 3CLpro inhibitors to combat COVID-19.
Collapse
Affiliation(s)
- Gonca Çelik
- Department of Chemistry, Faculty of Science, Karadeniz Technical University, Trabzon 61080, Türkiye.
| | - Şengül Alpay Karaoğlu
- Department of Biology, Faculty of Science, Recep Tayyip Erdoğan University, Rize 53100, Türkiye
| | - Şeyma Suyabatmaz
- Department of Biology, Faculty of Science, Recep Tayyip Erdoğan University, Rize 53100, Türkiye
| | - Arif Bozdeveci
- Department of Biology, Faculty of Science, Recep Tayyip Erdoğan University, Rize 53100, Türkiye
| | - Gizem Tatar Yılmaz
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Karadeniz Technical University, Trabzon 61080, Türkiye; Karadeniz Technical University, Institute of Health Sciences, Department of Bioinformatics, 61080 Trabzon, Türkiye; Yılmaz Bilişim R&D Consulting Software Engineering and Services Trade Limited Company, 61081 Trabzon, Türkiye
| | - Nurettin Yaylı
- Department of Pharmacognosy, Faculty of Pharmacy, Karadeniz Technical University, Trabzon 61080, Türkiye
| | - Rahşan Akpınar
- Laboratory of Bee Diseases, Samsun Veterinary Control Institute, Samsun 55200, Türkiye
| | - Ayşegül Çopur Çiçek
- Department of Basic Medical Sciences, Faculty of Medicine, Istanbul Medipol University, Istanbul 34815, Türkiye
| |
Collapse
|
2
|
Petcherski A, Tingley BM, Martin A, Adams S, Brownstein AJ, Steinberg RA, Shabane B, Ngo J, Osto C, Garcia G, Veliova M, Arumugaswami V, Colby AH, Shirihai OS, Grinstaff MW. Endolysosome-targeted nanoparticle delivery of antiviral therapy for coronavirus infections. Life Sci Alliance 2025; 8:e202403182. [PMID: 39900438 PMCID: PMC11790838 DOI: 10.26508/lsa.202403182] [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: 12/18/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/05/2025] Open
Abstract
SARS-CoV-2 can infect cells through endocytic uptake, a process that is targeted by inhibition of lysosomal proteases. However, clinically this approach to treat viral infections has afforded mixed results, with some studies detailing an oral regimen of hydroxychloroquine accompanied by significant off-target toxicities. We rationalized that an organelle-targeted approach will avoid toxicity while increasing the concentration of the drug at the target. Here, we describe a lysosome-targeted, mefloquine-loaded poly(glycerol monostearate-co-ε-caprolactone) nanoparticle (MFQ-NP) for pulmonary delivery via inhalation. Mefloquine is a more effective inhibitor of viral endocytosis than hydroxychloroquine in cellular models of COVID-19. MFQ-NPs are less toxic than molecular mefloquine, are 100-150 nm in diameter, and possess a negative surface charge, which facilitates uptake via endocytosis allowing inhibition of lysosomal proteases. MFQ-NPs inhibit coronavirus infection in mouse MHV-A59 and human OC43 coronavirus model systems and inhibit SARS-CoV-2 WA1 and its Omicron variant in a human lung epithelium model. Organelle-targeted delivery is an effective means to inhibit viral infection.
Collapse
Affiliation(s)
- Anton Petcherski
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Brett M Tingley
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Andrew Martin
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sarah Adams
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Alexandra J Brownstein
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Molecular Cellular Integrative Physiology, University of California Los Angeles, Los Angeles, CA, USA
| | - Ross A Steinberg
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Byourak Shabane
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer Ngo
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Corey Osto
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Michaela Veliova
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Aaron H Colby
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Orian S Shirihai
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Department of Chemistry, Boston University, Boston, MA, USA
| |
Collapse
|
3
|
Pacnejer AM, Negru MC, Arseniu AM, Trandafirescu C, Oancea C, Gligor FG, Morgovan C, Butuca A, Dehelean CA. Comparative Analysis of Neuropsychiatric Adverse Reactions Associated with Remdesivir and Nirmatrelvir/Ritonavir in COVID-19 Treatment: Insights from EudraVigilance Data. J Clin Med 2025; 14:1886. [PMID: 40142695 PMCID: PMC11942844 DOI: 10.3390/jcm14061886] [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: 02/07/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Remdesivir (RDV) and nirmatrelvir/ritonavir (NMVr) are among the most widely used antivirals in the treatment of COVID-19, aiming to reduce disease severity and progression. Adverse neuropsychiatric effects, such as anxiety, sleep disturbances, and movement disorders, have emerged as significant concerns associated with these treatments. To better understand the safety profiles of RDV and NMVr, this study performs a pharmacovigilance analysis of individual case safety reports (ICSRs) from the EudraVigilance (EV) database. Objectives: This study evaluates the risk of neuropsychiatric adverse events associated with RDV and NMVr. Comparisons with other antiviral drugs, including darunavir, sofosbuvir, ribavirin, tenofovir, ritonavir, and sotrovimab, are also performed to develop a comprehensive understanding of the safety profiles. Methods: A retrospective analysis of ICSRs submitted to EV until 7 July 2024, with data extraction on 12 July 2024, was conducted. Demographic characteristics (age, sex, geographic region, and reporter type) and case severity were included in the descriptive analysis. Disproportionality analysis using reporting odds ratio (ROR) and 95% confidence intervals (CI) was performed to compare adverse drug reaction (ADRs) frequencies across 27 system organ classes (SOCs), with emphasis on "Nervous system disorders" and "Psychiatric disorders. Results: The total number of ICSRs was significantly higher for NMVr (n = 8078) compared to RDV (n = 3934). Nervous system disorders accounted for 3.07% of the total RDV reports and for 17.31% of NMVr reports, while psychiatric disorders represented 0.92% of the total ADRs reported for RDV (n = 60) and 3.61% for NMVr (n = 672). On the other hand, RDV showed a significantly lower frequency of reporting headache compared to NMVr (ROR: 0.1057; 95% CI: 0.0676-0.1653). Conclusions: NMVr presents a higher risk of neuropsychiatric ADRs than RDV, underscoring the need for enhanced monitoring, particularly in patients with preexisting central nervous system (CNS) conditions. These findings contribute to optimizing antiviral safety and informing clinical decision making.
Collapse
Affiliation(s)
- Aliteia-Maria Pacnejer
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timișoara, Romania; (A.-M.P.); (C.A.D.)
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (F.G.G.); (C.M.); (A.B.)
| | - Mihaela Cristina Negru
- Department of ENT, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
| | - Anca Maria Arseniu
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (F.G.G.); (C.M.); (A.B.)
| | - Cristina Trandafirescu
- Discipline of Pharmaceutical Chemistry, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timișoara, Romania;
| | - Cristian Oancea
- Department of Pulmonology, Center for Research and Innovation in Personalized Medicine of Respiratory Diseases, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania;
| | - Felicia Gabriela Gligor
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (F.G.G.); (C.M.); (A.B.)
| | - Claudiu Morgovan
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (F.G.G.); (C.M.); (A.B.)
| | - Anca Butuca
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (F.G.G.); (C.M.); (A.B.)
| | - Cristina Adriana Dehelean
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, 2nd Eftimie Murgu Square, 300041 Timișoara, Romania; (A.-M.P.); (C.A.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
| |
Collapse
|
4
|
Nieves DJ, Tran MT, Singh J, Ashouri N, Morphew T, Lusk JG, Adler-Shohet FC, Marano R, Osborne S, Strickland J, Arrieta AC. Remdesivir Use in Pediatric Patients with Acute SARS-CoV-2 Infection Is Safe and Well Tolerated. CHILDREN (BASEL, SWITZERLAND) 2025; 12:331. [PMID: 40150613 PMCID: PMC11941674 DOI: 10.3390/children12030331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 02/26/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025]
Abstract
Background/Objective: Millions of children were infected with SARS-CoV-2, and a small proportion progressed to severe disease, especially those with underlying risk factors. Adult COVID-19 studies showed mortality benefits with Remdesivir. Data on Remdesivir use in pediatrics are limited. We report on the safety and tolerability of Remdesivir in pediatric patients seen at our institution. Methods: This was a retrospective cohort study of patients <19 years old with acute SARS-CoV-2 infection who received at least one dose of Remdesivir. Patients followed strict institutional guidelines for safety monitoring including standard clinical and laboratory daily observations. Demographics and underlying conditions were reported as averages; for laboratory values, linear regression was applied within a generalized linear mixed-effects model framework to evaluate the significance of changes in average levels over time. Results: We enrolled 318 patients with acute SARS-CoV2 infection from May 2020 to December 2022. In total, 53% were male, and the age range was distributed broadly. In total, 61% were school-aged children (28% 5-11 and 33% 12-18 years of age). In total, 62% of cases were Hispanic. The most common reasons for Remdesivir treatment included respiratory distress (201; 63%) and having high-risk underlying conditions (109; 34%). Therapy was completed as planned in 91% and discontinued early in 9%. Mean baseline, peak, and end of treatment values for AST were 57 (95% CI 53, 61), 79 (95% CI 73, 84) (p < 0.001), and 55 (51, 59) (p = 0.479); for ALT, they were 42 (38, 47), 59 (95% CI 52, 66) (p < 0.001), and 46 (95% CI 41, 52) (p = 0.054); and for bilirubin, they were 0.56 (95% CI 0.50, 0.62), 0.67 (95% CI 0.61, 0.74) (p < 0.001), and 0.44 (95% CI 0.40, 0.48) (p < 0.001), respectively. During Remdesivir treatment, we did not observe marrow suppression or renal toxicity. Conclusions: No clinically significant hematological or renal toxicity was noted. Mean liver enzymes increased modestly and returned to baseline without interrupting treatment. Remdesivir was well tolerated in patients <19 years old.
Collapse
Affiliation(s)
- Delma J. Nieves
- Division of Infectious Diseases, Department of Pediatrics, CHOC Children’s Hospital, University of California, Orange, CA 92868, USA; (J.S.); (N.A.); (A.C.A.)
| | - M. Tuan Tran
- Division of Infectious Diseases, Department of Pharmacy, CHOC Children’s Hospital, Orange, CA 92868, USA;
| | - Jasjit Singh
- Division of Infectious Diseases, Department of Pediatrics, CHOC Children’s Hospital, University of California, Orange, CA 92868, USA; (J.S.); (N.A.); (A.C.A.)
| | - Negar Ashouri
- Division of Infectious Diseases, Department of Pediatrics, CHOC Children’s Hospital, University of California, Orange, CA 92868, USA; (J.S.); (N.A.); (A.C.A.)
| | - Tricia Morphew
- CHOC Research Institute, Orange, CA 92868, USA; (T.M.); (S.O.)
| | - Jennifer G. Lusk
- Division of Hospitalist Medicine, Department of Pediatrics, CHOC Children’s Hospital, University of California, Orange, CA 92868, USA;
| | | | - Rachel Marano
- Rady Children’s Hospital, University of California, San Diego, CA 92123, USA;
| | | | - Jennifer Strickland
- Division of Infectious Diseases, CHOC Children’s Hospital, Orange, CA 92868, USA;
| | - Antonio C. Arrieta
- Division of Infectious Diseases, Department of Pediatrics, CHOC Children’s Hospital, University of California, Orange, CA 92868, USA; (J.S.); (N.A.); (A.C.A.)
| |
Collapse
|
5
|
Zhao T, Wang Z, Tong M, Fei Y. The development of therapeutics and vaccines against COVID-19. Diagn Microbiol Infect Dis 2025; 111:116643. [PMID: 39637679 DOI: 10.1016/j.diagmicrobio.2024.116643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/24/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Since the COVID-19 pandemic, it has caused a great threat to the global economy and public health, initiatives have been launched to control the spread of the virus. To explore the efficacy of drugs, a large number of clinical trials have been carried out, with the purpose of providing guidelines based on high-quality evidence for clinicians. We mainly discuss therapeutic agents for COVID-19 and explain the mechanism, including antiviral agents, tocilizumab, Janus kinase (JAK) inhibitors, neutralizing antibody therapies and corticosteroids. In addition, the COVID-19 vaccine has been proven to be efficacious in preventing SARS-CoV-2 infection. We systematically analyzed four mainstream vaccine platforms: messenger RNA (mRNA) vaccines, viral vector vaccines, inactivated vaccines and protein subunit vaccines. We evaluated the therapeutic effects of drugs and vaccines through enumerating the most typical clinical trials. However, the emergence of novel variants has further complicated the interpretation of the available clinical data, especially vaccines and antibody therapies. In the post-epidemic era, therapeutic agents are still the first choice for controlling the progression of disease, whereas the protective effect of vaccines against different strains should be assessed comprehensively.
Collapse
Affiliation(s)
- Tianyu Zhao
- The Affiliated Hospital of Shao Xing University/The Affiliated Hospital of Shao Xing University(Shao Xing Municipal Hospital), China
| | - Zhiwei Wang
- The Affiliated Hospital of Shao Xing University/The Affiliated Hospital of Shao Xing University(Shao Xing Municipal Hospital), China
| | - Mingjiong Tong
- The Affiliated Hospital of Shao Xing University/The Affiliated Hospital of Shao Xing University(Shao Xing Municipal Hospital), China
| | - Yingming Fei
- The Affiliated Hospital of Shao Xing University/The Affiliated Hospital of Shao Xing University(Shao Xing Municipal Hospital), China.
| |
Collapse
|
6
|
Garcia Vidal C, González J, Lumbreras C, Salavert M, Castro A, Rubio-Rodríguez D, Rubio-Terrés C. Effectiveness of pharmacological treatments for COVID-19 due to SARS-CoV-2: a systematic literature review. Front Pharmacol 2025; 16:1469681. [PMID: 40093322 PMCID: PMC11907208 DOI: 10.3389/fphar.2025.1469681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
Aim Since the first cases of the COVID-19 pandemic, caused by the SARS-CoV-2 virus, described in 2019, numerous drugs have been proposed for the treatment of the disease. However, studies have given contradictory or inconclusive results, making it difficult to determine which treatments are truly effective. The objective was to carry out a systematic review of the literature analyzing the effectiveness (mortality, hospitalization and clinical improvement) of COVID-19 treatments initially proposed and finally authorized in the European Union. Methods PubMed and other electronic databases were systematically searched for meta-analyses published between January 2020 and December 2022, as well as two additional searches: one of individual clinical studies published until October 2023 and another of those drugs that were considered at the beginning and that were discarded early because the clinical results were unfavorable. Results In the synthesis, 85 meta-analyses and 19 additional clinical studies were included (base case). All medications indicated in the treatment of COVID-19 have favorable efficacy results (mortality, hospitalization rate, clinical improvement) but these results were not confirmed in all studies carried out, being frequently contradictory (confirming or not confirming the impact of treatment on mortality). According to meta-analysis with the largest sample size, the drugs with the greatest evidence of effectiveness in reducing mortality are remdesivir (HR= 0.79; 95% CI 0.73-0.85) and tocilizumab (OR= 0.73; 95% CI 0.56-0.93). Regarding the composite of Covid-19-related hospitalization or death from any cause, the drugs with the greatest evidence of efficacy are remdesivir, nirmatrelvir/ritonavir and sotrovimab (although, currently the effectiveness of monoclonal antibodies against the new variants of the virus has not been demonstrated). Conclusion According to this systematic review, the treatments with the greatest evidence of reducing mortality in patients with COVID-19 are remdesivir and tocilizumab.
Collapse
Affiliation(s)
| | - Jonathan González
- Pharmacy Department, Complejo Hospitalario Universitario de Canarias, Santa Cruz de Tenerife, Spain
| | - Carlos Lumbreras
- Internal Medicine Department, Hospital Universitario “12 de Octubre”, Madrid, Spain
| | - Miguel Salavert
- Infectious Diseases Unit, Hospital Universitario y Politécnico “La Fe”, Valencia, Spain
| | | | | | | |
Collapse
|
7
|
Nyström K, Trybala E, Said J, Roth A, Patzi Churqui M, Kärmander A, Cihlar T, Bilello JP, Bergström T, Lagging M. Remdesivir is active in vitro against tick-borne encephalitis virus and selects for resistance mutations in the viral RNA-dependent RNA polymerase. Infect Dis (Lond) 2025:1-8. [PMID: 39973341 DOI: 10.1080/23744235.2025.2468510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Tick-borne encephalitis (TBE) is a neurological disease caused by the tick-borne encephalitis virus (TBEV). Despite available vaccines, breakthrough infections occur, some fatal. OBJECTIVES As no antiviral therapy for TBE is currently approved, this study evaluated the in vitro activity of already licenced remdesivir (RDV) and sofosbuvir (SOF) for possible drug repurposing against TBEV. METHODS TBEV was cultured in A549 cells, and the inhibitory effects of RDV (GS-5734), its parent nucleotide GS-441524, and SOF (GS-7977) were assessed. RESULTS After 78 h, RDV demonstrated significantly lower EC50 values than SOF (0.14 vs. 11 µM) based on TBEV RNA levels measured by RT-qPCR. RDV also had a lower mean EC50 (0.55 µM) compared to GS-441524 and SOF (>8.9 and 13.1 µM, respectively) using crystal violet staining after 5 days. After 11 passages of TBEV in the presence of RDV, emergence of virus with a higher EC50 (1.32 vs. 0.55 µM) was detected with two mutations (L3122F and Y3278F) in NS5, the viral RNA-dependent RNA polymerase (RdRp), and one substitution in envelope (E) protein (E402G). Similarly, SOF resistance appeared after 20 passages, increasing EC50 values (35.5 vs. 10 µM). CONCLUSION RDV exhibits potent in vitro antiviral activity against TBEV via specific targeting of the viral RdRp as confirmed by the emergence of resistance-associated double NS5 substitutions in vitro in the presence of RDV. While the potential in vivo implications of the observed RDV resistance remain to be determined, these in vitro data support further assessment of RDV for the treatment of TBEV infection.
Collapse
Affiliation(s)
- Kristina Nyström
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Edward Trybala
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joanna Said
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anette Roth
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marianela Patzi Churqui
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ambjörn Kärmander
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Tomas Bergström
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Martin Lagging
- Department of Infectious Diseases/Virology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Microbiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
8
|
Saha A, Ganguly A, Kumar A, Srivastava N, Pathak R. Harnessing Epigenetics: Innovative Approaches in Diagnosing and Combating Viral Acute Respiratory Infections. Pathogens 2025; 14:129. [PMID: 40005506 PMCID: PMC11858160 DOI: 10.3390/pathogens14020129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Acute respiratory infections (ARIs) caused by viruses such as SARS-CoV-2, influenza viruses, and respiratory syncytial virus (RSV), pose significant global health challenges, particularly for the elderly and immunocompromised individuals. Substantial evidence indicates that acute viral infections can manipulate the host's epigenome through mechanisms like DNA methylation and histone modifications as part of the immune response. These epigenetic alterations can persist beyond the acute phase, influencing long-term immunity and susceptibility to subsequent infections. Post-infection modulation of the host epigenome may help distinguish infected from uninfected individuals and predict disease severity. Understanding these interactions is crucial for developing effective treatments and preventive strategies for viral ARIs. This review highlights the critical role of epigenetic modifications following viral ARIs in regulating the host's innate immune defense mechanisms. We discuss the implications of these modifications for diagnosing, preventing, and treating viral infections, contributing to the advancement of precision medicine. Recent studies have identified specific epigenetic changes, such as hypermethylation of interferon-stimulated genes in severe COVID-19 cases, which could serve as biomarkers for early detection and disease progression. Additionally, epigenetic therapies, including inhibitors of DNA methyltransferases and histone deacetylases, show promise in modulating the immune response and improving patient outcomes. Overall, this review provides valuable insights into the epigenetic landscape of viral ARIs, extending beyond traditional genetic perspectives. These insights are essential for advancing diagnostic techniques and developing innovative treatments to address the growing threat of emerging viruses causing ARIs globally.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar 814152, India;
| | - Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, India;
| | - Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
9
|
Ling-Hu T, Simons LM, Rios-Guzman E, Carvalho AM, Agnes MFR, Alisoltanidehkordi A, Ozer EA, Lorenzo-Redondo R, Hultquist JF. The impact of remdesivir on SARS-CoV-2 evolution in vivo. JCI Insight 2025; 10:e182376. [PMID: 39836474 PMCID: PMC11949014 DOI: 10.1172/jci.insight.182376] [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: 04/24/2024] [Accepted: 01/13/2025] [Indexed: 01/23/2025] Open
Abstract
The impact of remdesivir on SARS-CoV-2 diversity and evolution in vivo has remained unclear. In this single-center, retrospective cohort study, we assessed SARS-CoV-2 diversification and diversity over time in a cohort of hospitalized patients who did or did not receive remdesivir. Whole-genome sequencing was performed on 98 paired specimens collected from 49 patients before and after remdesivir administration. The genetic divergence between paired specimens was not significantly different in this cohort compared with that in a control group of patients who did not receive the drug. However, when we focused on minority variants, several positions showed preferential diversification after remdesivir treatment, some of which were associated with specific variants of concern. Most notably, remdesivir administration resulted in strong selection for a nonsynonymous mutation in nsp12, G671S, previously associated with enhanced viral fitness. This same mutation was found to be enriched in a second cohort of 143 inpatients with specimens collected after remdesivir administration compared with controls. Only one other mutation previously implicated in remdesivir resistance (nsp12:V792I) was found to be preferentially selected for after remdesivir administration. These data suggest that SARS-CoV-2 variants with enhanced replicative fitness may be selected for in the presence of antiviral therapy as an indirect means to overcome this selective pressure.
Collapse
Affiliation(s)
- Ted Ling-Hu
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Lacy M. Simons
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Estefany Rios-Guzman
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Alexandre Machado Carvalho
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Maria Francesca R. Agnes
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Arghavan Alisoltanidehkordi
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Egon A. Ozer
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Ramon Lorenzo-Redondo
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| | - Judd F. Hultquist
- Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, Illinois, USA
| |
Collapse
|
10
|
Pranić SM, Estevão MD, Vasanthan LT, Pérez-Neri I, Pulumati A, de Lima Junior FAS, Malih N, Mishra V, Thompson J, Nnate D. Reporting of participant race and ethnicity from COVID-19 randomized controlled drug and biologicals trials: a scoping review. Epidemiol Rev 2025; 47:1-14. [PMID: 39673248 DOI: 10.1093/epirev/mxae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 07/10/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024] Open
Abstract
Racial and ethnic minorities have been disproportionally burdened by hospitalization and death due to COVID-19. Participation of individuals of diverse races and ethnicities in clinical trials, according to study-level characteristics of randomized controlled trials (RCTs) that test effectiveness of COVID-19 drugs, could be insightful for future researchers. Our objective for this scoping review was to describe the frequency of race and ethnicity reported as demographic variables and specific reporting of race and ethnicity according to COVID-19 RCT characteristics. We conducted comprehensive searches in PubMed, ProQuest, World Health Organization Database, and Cochrane Central Register of Controlled Trials, and gray literature via preprint servers from January 1, 2020, to May 4, 2022. We included RCTs on emergency- or conditionally approved COVID-19 drug interventions (remdesivir, baricitinib, and molnupiravir) with or without comparators. Self-reported race as American Indian/Pacific Islander, Asian, Black/African American, or White, ethnicity as Hispanic/Latinx, study design characteristics, and participant-relevant data were collected. In total, 17 RCTs with 17 935 participants were included. Most (n = 13; 76%) reported at least 1 race and ethnicity and were US-based, industry-funded RCTs. Asian, Black, Latinx, and White participants were mostly enrolled in RCTs that studied remdesivir. Native American and Hawaiian participants were mostly assessed for progression to high-flow oxygen/noninvasive ventilation. Time to recovery was assessed predominantly in Black and White participants, whereas hospitalization or death was mostly assessed in Asian, Latinx, and multirace participants. Trialists should be aware of RCT-level factors and characteristics that may be associated with low participation of racial and ethnic minorities, which could inform evidence-based interventions to increase minority participation.
Collapse
Affiliation(s)
- Shelly Melissa Pranić
- Department of Public Health, University of Split School of Medicine, 21000 Split, Croatia
- Cochrane Croatia, 21000 Split, Croatia
| | - Maria Dulce Estevão
- School of Health, University of Algarve, Faro, Faro District, 8005-139, Portugal
| | - Lenny T Vasanthan
- Physiotherapy Unit, Physical Medicine and Rehabilitation Department, Christian Medical College, Vellore, 632004, India
| | - Iván Pérez-Neri
- Department of Neurochemistry, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Insurgentes Sur 3877, La Fama, Tlalpan, 14269, Ciudad de México, Mexico
| | - Anika Pulumati
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| | - Fábio Antonio Serra de Lima Junior
- Centro de Ciências Médicas, Universidade Federal da Paraíba (Federal University of Paraíba), João Pessoa, Castelo Branco, PB, 58051-900, Brazil
| | - Narges Malih
- Global Health Research Group, University of the Balearic Islands, 07122 Palma, Spain
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, 1983969411 Tehran, Iran
| | - Vinayak Mishra
- University of Liverpool, Liverpool, L69 7ZX, United Kingdom
| | | | - Daniel Nnate
- University of Liverpool, Liverpool, L69 7ZX, United Kingdom
| |
Collapse
|
11
|
Mozaffari E, Chandak A, Gottlieb RL, Kalil AC, Jiang H, Oppelt T, Berry M, Chima-Melton C, Amin AN. Remdesivir Effectiveness in Reducing the Risk of 30-Day Readmission in Vulnerable Patients Hospitalized for COVID-19: A Retrospective US Cohort Study Using Propensity Scores. Clin Infect Dis 2024; 79:S167-S177. [PMID: 39405450 PMCID: PMC11638780 DOI: 10.1093/cid/ciae511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Reducing hospital readmission offer potential benefits for patients, providers, payers, and policymakers to improve quality of healthcare, reduce cost, and improve patient experience. We investigated effectiveness of remdesivir in reducing 30-day coronavirus disease 2019 (COVID-19)-related readmission during the Omicron era, including older adults and those with underlying immunocompromising conditions. METHODS This retrospective study utilized the US PINC AI Healthcare Database to identify adult patients discharged alive from an index COVID-19 hospitalization between December 2021 and February 2024. Odds of 30-day COVID-19-related readmission to the same hospital were compared between patients who received remdesivir vs those who did not, after balancing characteristics of the two groups using inverse probability of treatment weighting (IPTW). Analyses were stratified by maximum supplemental oxygen requirement during index hospitalization. RESULTS Of 326 033 patients hospitalized for COVID-19 during study period, 210 586 patients met the eligibility criteria. Of these, 109 551 (52%) patients were treated with remdesivir. After IPTW, lower odds of 30-day COVID-19-related readmission were observed in patients who received remdesivir vs those who did not, in the overall population (3.3% vs 4.2%, respectively; odds ratio [95% confidence interval {CI}]: 0.78 [.75-.80]), elderly population (3.7% vs 4.7%, respectively; 0.78 [.75-.81]), and those with underlying immunocompromising conditions (5.3% vs 6.2%, respectively; 0.86 [.80-.92]). These results were consistent irrespective of supplemental oxygen requirements. CONCLUSIONS Treating patients hospitalized for COVID-19 with remdesivir was associated with a significantly lower likelihood of 30-day COVID-19-related readmission across all patients discharged alive from the initial COVID-19 hospitalization, including older adults and those with underlying immunocompromising conditions.
Collapse
Affiliation(s)
- Essy Mozaffari
- Medical Affairs, Gilead Sciences, Foster City, California, USA
| | | | - Robert L Gottlieb
- Department of Internal Medicine, Baylor University Medical Center, Dallas, Texas, USA
- Baylor Scott & White Heart and Vascular Hospital, Dallas, Texas, USA
- Baylor Scott & White The Heart Hospital, Plano, Texas, USA
- Baylor Scott & White Research Institute, Dallas, Texas, USA
| | - Andre C Kalil
- Division of Infectious Diseases, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Heng Jiang
- Evidence and Access, Certara, Paris, France
| | - Thomas Oppelt
- Medical Affairs, Gilead Sciences, Foster City, California, USA
| | - Mark Berry
- Real World Evidence, Gilead Sciences, Foster City, California, USA
| | | | - Alpesh N Amin
- Division of Hospital Medicine & Palliative Medicine, Department of Medicine, University of California Irvine, Orange, California, USA
| |
Collapse
|
12
|
Kuritzkes DR. Remdesivir for Patients Hospitalized With COVID-19: Evidence of Effectiveness From Cohort Studies in the Omicron Era. Clin Infect Dis 2024; 79:S127-S130. [PMID: 39445631 PMCID: PMC11638770 DOI: 10.1093/cid/ciae515] [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: 08/21/2024] [Revised: 10/11/2024] [Accepted: 10/22/2024] [Indexed: 10/25/2024] Open
Abstract
Remdesivir is the only antiviral approved for treatment of persons hospitalized for coronavirus disease 2019 (COVID-19). This supplement presents new information from real-world cohort studies that report reduced mortality in at-risk populations and reduction in readmission for COVID-19 in the Omicron era.
Collapse
Affiliation(s)
- Daniel R Kuritzkes
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Liu WL, Kampouri E, Bui JK, Sekhon MK, Tercero A, Finlay D, Asghedom LH, Romasanta GR, Rice NT, Ranjbaran F, Stoltzman C, Cook J, Blake J, Delaney CS, Hill JA. Off-the-shelf allogeneic natural killer cells for the treatment of COVID-19. Mol Ther Methods Clin Dev 2024; 32:101361. [PMID: 39624798 PMCID: PMC11609367 DOI: 10.1016/j.omtm.2024.101361] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/24/2024] [Indexed: 02/14/2025]
Abstract
Low levels and function of natural killer (NK) cells are associated with increased coronavirus disease 2019 (COVID-19) severity. NK cell immunotherapy may improve immune function to reduce infection severity. We conducted a first-in-human, open-label, phase 1, dose-escalating (100 × 106, 300 × 106, or 900 × 106 cells) study of a single dose of DVX201, a cord-blood-derived allogeneic NK cell therapy, in hospitalized patients with COVID-19. Participants were followed for 28 days. The maximum allowed steroid dose for eligibility was up to 0.5 mg/kg prednisone (or equivalent) daily. We enrolled nine participants, 3 per dose level. Eight participants had ≥1 comorbidity associated with increased COVID-19 severity, three of whom had a hematologic malignancy. Infusions were well tolerated, with no treatment-related adverse events. There was no evidence of inflammatory complications related to infusions. Peripheral blood NK cells generally increased after infusion, peaking by day 7. The median time from infusion to discharge was 2 days (range: 1-13). Two patients (both with acute lymphoblastic leukemia) were readmitted with recurrent COVID-19. This trial demonstrates the safety of allogeneic NK cell immunotherapy as a potential antiviral. Larger controlled trials are needed to establish efficacy.
Collapse
Affiliation(s)
| | | | - John K. Bui
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington School of Medicine, Seattle, WA, USA
| | | | | | - Dan Finlay
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | | | | | | | | | - Jody Cook
- Deverra Therapeutics, Seattle, WA, USA
| | - Joe Blake
- Deverra Therapeutics, Seattle, WA, USA
| | | | - Joshua A. Hill
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
14
|
Neilsen G, Mathew AM, Castro JM, McFadden WM, Wen X, Ong YT, Tedbury PR, Lan S, Sarafianos SG. Dimming the corona: studying SARS-coronavirus-2 at reduced biocontainment level using replicons and virus-like particles. mBio 2024; 15:e0336823. [PMID: 39530689 PMCID: PMC11633226 DOI: 10.1128/mbio.03368-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
The coronavirus-induced disease 19 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infections, has had a devastating impact on millions of lives globally, with severe mortality rates and catastrophic social implications. Developing tools for effective vaccine strategies and platforms is essential for controlling and preventing the recurrence of such pandemics. Moreover, molecular virology tools that facilitate the study of viral pathogens, impact of viral mutations, and interactions with various host proteins are essential. Viral replicon- and virus-like particle (VLP)-based systems are excellent examples of such tools. This review outlines the importance, advantages, and disadvantages of both the replicon- and VLP-based systems that have been developed for SARS-CoV-2 and have helped the scientific community in dimming the intensity of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Grace Neilsen
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Asha Maria Mathew
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Jose M. Castro
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - William M. McFadden
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Xin Wen
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Yee T. Ong
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Philip R. Tedbury
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Shuiyun Lan
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Stefan G. Sarafianos
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Center for ViroScience and Cure, Emory University School of Medicine, Atlanta, Georgia, USA
- Children’s Healthcare of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|
15
|
Satoh T, Akata K, Yamasaki K, Tsuruta A, Yatera K. Successful Sequential Treatment From Remdesivir to Molnupiravir for Prolonged COVID-19 in a Patient With Follicular Lymphoma and Renal Pelvic Carcinoma: A Case Report. Cureus 2024; 16:e75722. [PMID: 39816292 PMCID: PMC11732614 DOI: 10.7759/cureus.75722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 01/18/2025] Open
Abstract
A 69-year-old Japanese male with follicular lymphoma and renal pelvic carcinoma presented with fever and cough, testing positive for SARS-CoV-2 via PCR. Chest CT revealed ground-glass opacities. Initially, his symptoms improved with a 10-day course of remdesivir (RDV), but they recurred. On day 42, a second 10-day course of RDV combined with dexamethasone was initiated; however, symptoms persisted, and his SARS-CoV-2 PCR test remained positive on day 72. Subsequently, a successful treatment regimen of 10 days of RDV followed by five days of molnupiravir (MOL) was administered. This study represents the first reported case of persistent SARS-CoV-2 infection successfully treated with sequential therapy transitioning from RDV to MOL, without extending the treatment duration.
Collapse
Affiliation(s)
- Tomoki Satoh
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, JPN
| | - Kentaro Akata
- Division of Infection Control and Prevention, University of Occupational and Environmental Health, Japan, Kitakyushu, JPN
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, JPN
| | - Kei Yamasaki
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, JPN
| | - Akimasa Tsuruta
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, JPN
| | - Kazuhiro Yatera
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, JPN
| |
Collapse
|
16
|
Sise ME, Santos JR, Goldman JD, Tuttle KR, Teixeira JP, Seibert AF, Koullias Y, Llewellyn J, Regan S, Zhao Y, Huang H, Hyland RH, Osinusi A, Winter H, Humeniuk R, Hulter HN, Gottlieb RL, Fusco DN, Birne R, Stancampiano FF, Libertin CR, Small CB, Plate M, McPhail MJ. Efficacy and Safety of Remdesivir in People With Impaired Kidney Function Hospitalized for COVID-19 Pneumonia: A Randomized Clinical Trial. Clin Infect Dis 2024; 79:1172-1181. [PMID: 38913574 PMCID: PMC11581693 DOI: 10.1093/cid/ciae333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/06/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Few antiviral therapies have been studied in patients with coronavirus disease 2019 (COVID-19) and kidney impairment. Herein, the efficacy, safety, and pharmacokinetics of remdesivir, its metabolites, and sulfobutylether-β-cyclodextrin excipient were evaluated in hospitalized patients with COVID-19 and severe kidney impairment. METHODS In REDPINE, a phase 3, randomized, double-blind, placebo-controlled study, participants aged ≥12 years hospitalized for COVID-19 pneumonia with acute kidney injury, chronic kidney disease, or kidney failure were randomized 2:1 to receive intravenous remdesivir (200 mg on day 1; 100 mg daily up to day 5) or placebo (enrollment from March 2021 to March 2022). The primary efficacy end point was the composite of the all-cause mortality rate or invasive mechanical ventilation rate through day 29. Safety was evaluated through day 60. RESULTS Although enrollment concluded early, 243 participants were enrolled and treated (remdesivir, n = 163; placebo, n = 80). At baseline, 90 participants (37.0%) had acute kidney injury (remdesivir, n = 60; placebo, n = 30), 64 (26.3%) had chronic kidney disease (remdesivir, n = 44; placebo, n = 20), and 89 (36.6%) had kidney failure (remdesivir, n = 59; placebo, n = 30); and 31 (12.8%) were vaccinated against COVID-19. Composite all-cause mortality or invasive mechanical ventilation rates through day 29 were 29.4% and 32.5% in the remdesivir and placebo group, respectively (P = .61). Treatment-emergent adverse events were reported in 80.4% for remdesivir versus 77.5% for placebo, and serious adverse events in 50.3% versus 50.0%, respectively. Pharmacokinetic plasma exposure to remdesivir was not affected by kidney function. CONCLUSIONS Although the study was underpowered, no significant difference in efficacy was observed between treatment groups. REDPINE demonstrated that remdesivir is safe in patients with COVID-19 and severe kidney impairment. CLINICAL TRIALS REGISTRATION EudraCT 2020-005416-22; Clinical Trials.gov NCT04745351.
Collapse
Affiliation(s)
- Meghan E Sise
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jose Ramon Santos
- Fight Infections Foundation, Service of Infectious Diseases, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Jason D Goldman
- Swedish Center for Research and Innovation, Providence Swedish Medical Center, Seattle, Washington, USA
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Katherine R Tuttle
- Providence Medical Research Center, Providence Inland Northwest Health, Spokane, Washington, USA
| | - J Pedro Teixeira
- Divisions of Nephrology and Pulmonary, Critical Care, and Sleep Medicine, University of New Mexico Hospital, Albuquerque, New Mexico, USA
| | - Allan F Seibert
- Pulmonary Associates Research, Ascension Providence, Mobile, Alabama, USA
| | | | | | - Sean Regan
- Gilead Sciences, Foster City, California, USA
| | - Yang Zhao
- Gilead Sciences, Foster City, California, USA
| | | | | | - Anu Osinusi
- Gilead Sciences, Foster City, California, USA
| | | | | | - Henry N Hulter
- Department of Medicine, University of California, San Francisco, California, USA
| | - Robert L Gottlieb
- Department of Internal Medicine, Baylor University Medical Center, Dallas, Texas, USA
- Baylor Scott & White Research Institute, Dallas, Texas, USA
| | - Dahlene N Fusco
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Rita Birne
- Department of Nephrology, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- NOVA Medical School, Lisbon, Portugal
| | - Fernando F Stancampiano
- Department of Internal Medicine, Mayo Clinic College of Medicine and Science, Jacksonville, Florida, USA
| | - Claudia R Libertin
- Department of Internal Medicine, Mayo Clinic College of Medicine and Science, Jacksonville, Florida, USA
| | - Catherine B Small
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, USA
| | - Markus Plate
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, USA
| | - Mark J McPhail
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| |
Collapse
|
17
|
Fusco D, Malenica I, Günthard HF, Gupta SK, Kurbegov D, Balani B, Olender S, Aberg JA, Telep LE, Tian Y, Blair C, Wu G, Haubrich RH, Wang CY, Chokkalingam AP, Osinusi AO, Wendtner CM, Gottlieb RL. Remdesivir for Treatment of COVID-19 Requiring Oxygen Support: A Cross-Study Comparison From 2 Large, Open-Label Studies. Clin Infect Dis 2024; 79:1182-1189. [PMID: 38920297 DOI: 10.1093/cid/ciae336] [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: 03/01/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Remdesivir, an RNA-polymerase prodrug inhibitor approved for treatment of coronavirus disease 2019 (COVID-19), shortens recovery time and improves clinical outcomes. This prespecified analysis compared remdesivir plus standard of care (SOC) with SOC alone in adults hospitalized with COVID-19 requiring oxygen support in the early stage of the pandemic. METHODS Data for 10-day remdesivir treatment plus SOC from the extension phase of an open-label study (NCT04292899) were compared with real-world, retrospective data on SOC alone (EUPAS34303). Both studies included patients aged ≥18 years hospitalized with severe acute respiratory syndrome coronavirus 2 up to 30 May 2020, with oxygen saturation ≤94% on room air or supplemental oxygen (all forms), and with pulmonary infiltrates. Propensity score weighting was used to balance patient demographics and clinical characteristics across treatment groups. The primary endpoint was time to all-cause mortality or end of study (day 28). Time to discharge, with a 10-day landmark to account for duration of remdesivir treatment, was a secondary endpoint. RESULTS A total of 1974 patients treated with remdesivir plus SOC, and 1426 with SOC alone, were included after weighting. Remdesivir significantly reduced mortality versus SOC (hazard ratio [HR], 0.46; 95% confidence interval, .39-.54). This association was observed at each oxygen support level, with the lowest HR for patients on low-flow oxygen. Remdesivir significantly increased the likelihood of discharge at day 28 versus SOC in the 10-day landmark analysis (HR, 1.64; 95% confidence interval: 1.43-1.87). CONCLUSIONS Remdesivir plus early-2020 SOC was associated with a 54% lower mortality risk and shorter hospital stays compared with SOC alone in patients hospitalized with COVID-19 requiring oxygen support. Clinical Trials Registration. ClinicalTrials.gov NCT04292899 and EUPAS34303.
Collapse
Affiliation(s)
- Dahlene Fusco
- Department of Medicine, Tulane Medical Center, New Orleans, Louisiana, USA
| | - Ivana Malenica
- Real World Evidence, Gilead Sciences, Inc., Foster City, California, USA
- Department of Biostatistics, University of California, Berkeley, Berkeley, California, USA
| | - Huldrych F Günthard
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
| | - Samir K Gupta
- Division of Infectious Diseases, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Bindu Balani
- Hackensack School of Medicine, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Susan Olender
- Department of Internal Medicine, Columbia University in the City of New York, New York, New York, USA
| | - Judith A Aberg
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Laura E Telep
- Real World Evidence, Gilead Sciences, Inc., Foster City, California, USA
| | - Yuan Tian
- Clinical Bioinformatics and Exploratory Analytics, Gilead Sciences, Inc., Foster City, California, USA
| | - Christiana Blair
- Biostatistics, Gilead Sciences, Inc., Foster City, California, USA
| | - George Wu
- Biostatistics, Gilead Sciences, Inc., Foster City, California, USA
| | - Richard H Haubrich
- Global Medical Affairs, Gilead Sciences, Inc., Foster City, California, USA
| | - Chen-Yu Wang
- Global Medical Affairs, Gilead Sciences, Inc., Foster City, California, USA
| | - Anand P Chokkalingam
- Real World Evidence, Gilead Sciences, Inc., Foster City, California, USA
- Division of Epidemiology, School of Public Health, University of California, Berkeley, California, USA
| | - Anu O Osinusi
- Clinical Development, Gilead Sciences, Inc., Foster City, California, USA
| | - Clemens-Martin Wendtner
- Munich Clinic Schwabing, Academic Teaching Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Robert L Gottlieb
- Center for Advanced Heart and Lung Disease, Baylor University Medical Center, Dallas, Texas, USA
- Department of Internal Medicine, Texas A&M Health Science Center, Dallas, Texas, USA
- Department of Internal Medicine, Burnett School of Medicine at TCU, Fort Worth, Texas, USA
- Baylor Scott & White Research Institute, Dallas, Texas, USA
| |
Collapse
|
18
|
Seethamraju H, Yang OO, Loftus R, Ogbuagu O, Sammartino D, Mansour A, Sacha JB, Ojha S, Hansen SG, Arman AC, Lalezari JP. A Randomized Placebo-Controlled Trial of Leronlimab in Mild-To-Moderate COVID-19. Clin Ther 2024; 46:891-899. [PMID: 39353749 DOI: 10.1016/j.clinthera.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 10/04/2024]
Abstract
PURPOSE Early in the course of the SARS-CoV-2 pandemic it was hypothesised that host genetics played a role in the pathophysiology of COVID-19 including a suggestion that the CCR5-Δ32 mutation may be protective in SARS-CoV-2 infection. Leronlimab is an investigational CCR5-specific humanized IgG4 monoclonal antibody currently in development for HIV-1 infection. We aimed to explore the impact of leronlimab on the severity of disease symptoms among participants with mild-to-moderate COVID-19. METHODS The TEMPEST trial was a randomized, double-blind, placebo-controlled study in participants with mild-to-moderate COVID-19. Participants were randomly assigned in a 2:1 ratio to receive subcutaneous leronlimab (700 mg) or placebo on days 0 and 7. The primary efficacy endpoint was assessed by change in total symptom score based on fever, myalgia, dyspnea, and cough, at end of treatment (day 14). FINDINGS Overall, 84 participants were randomized and treated with leronlimab (n = 56) or placebo (n = 28). No difference was observed in change in total symptom score (P = 0.8184) or other pre-specified secondary endpoints between treatments. However, in a post hoc analysis, 50.0% of participants treated with leronlimab demonstrated improvements from baseline in National Early Warning Score 2 (NEWS2) at day 14, compared with 20·8% of participants in the placebo group (post hoc; p = 0.0223). Among participants in this trial with mild-to-moderate COVID-19 adverse events rates were numerically but not statistically significantly lower in leronlimab participants (33.9%) compared with placebo participants (50.0%). IMPLICATIONS At the time the TEMPEST trial was designed although CCR5 was known to be implicated in COVID-19 disease severity the exact pathophysiology of SARS-CoV-2 infection was poorly understood. Today it is well accepted that SARS-CoV-2 infection in asymptomatic-to-mild cases is primarily characterized by viral replication, with a heightened immune response, accompanied by diminished viral replication in moderate-to-severe disease and a peak in inflammatory responses with excessive production of pro-inflammatory cytokines in critical disease. It is therefore perhaps not surprising that no differences between treatments were observed in the primary endpoint or in pre-specified secondary endpoints among participants with mild-to-moderate COVID-19. However, the results of the exploratory post hoc analysis showing that participants in the leronlimab group had greater improvement in NEWS2 assessment compared to placebo provided a suggestion that leronlimab may be associated with a lower likelihood of people with mild-to-moderate COVID-19 progressing to more severe disease and needs to be confirmed in other appropriately designed clinical trials. CLINICALTRIALS gov number, NCT04343651 https://classic. CLINICALTRIALS gov/ct2/show/NCT04343651.
Collapse
Affiliation(s)
| | - Otto O Yang
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | | | | | | | - Jonah B Sacha
- Oregon Health & Science University, Portland, Oregon
| | - Sohita Ojha
- Oregon Health & Science University, Portland, Oregon
| | | | | | | |
Collapse
|
19
|
Quesada Muñoz L, Fernández-Fradejas J, Martinez-Barros H, Sánchez Cuervo M, Martín Rufo M, Pintor Recuenco MDR, Quereda Rodríguez-Navarro C, Álvarez-Díaz AM, Saez de la Fuente J. Real-world effectiveness and factors associated with increased mortality in non-critically ill patients with COVID-19 pneumonia receiving remdesivir. Eur J Hosp Pharm 2024; 31:532-536. [PMID: 37339865 PMCID: PMC11672499 DOI: 10.1136/ejhpharm-2023-003729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/06/2023] [Indexed: 06/22/2023] Open
Abstract
OBJECTIVES Evidence on the effectiveness of remdesivir when used in real-life clinical practice is controversial. This study aims to analyse its effectiveness and the factors associated with increased mortality in non-critically ill patients with COVID-19 pneumonia who require supplemental low-flow oxygen and received remdesivir. METHODS A retrospective cohort study was conducted at Ramón y Cajal University Hospital (Madrid, Spain) which included all patients treated with remdesivir in our institution during the second pandemic breakout in Spain, from August to November 2020. Treatment with remdesivir was limited to non-critically ill patients with COVID-19 pneumonia requiring low-flow supplemental oxygen, with a treatment duration of 5 days. RESULTS A total of 1757 patients were admitted with COVID-19 pneumonia during the study period, of which 281 non-critically ill patients were treated with remdesivir and included in the analysis. Mortality at 28 days after initiation of treatment was 17.1%. The median (IQR) time to recovery was 9 days (6-15). 104 (37.0%) patients had complications during hospitalisation, with renal failure being the most frequent (31 patients; 36.5%). After adjustment for confounding factors, high-flow oxygen therapy was associated with increased 28-day mortality (HR 2.77; 95% CI 1.39 to 5.53; p=0.004) and decreased 28-day clinical improvement (HR 0.54; 95% CI 0.35 to 0.85; p=0.008). A significant difference in survival and clinical improvement was identified between patients treated with high and low-flow oxygen. CONCLUSION The 28-day mortality rate in patients treated with remdesivir needing low-flow oxygen therapy was higher than that published in clinical trials. Age and increased oxygen therapy needed after the beginning of treatment were the main risk factors associated with mortality.
Collapse
|
20
|
Badary HA, Hashem MB, El-Kassas M. Drug-induced liver injury during the era of COVID-19 polypharmacy: a statement of account, lessons learned, and a proposed approach. EGYPTIAN LIVER JOURNAL 2024; 14:75. [DOI: 10.1186/s43066-024-00381-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 10/07/2024] [Indexed: 01/03/2025] Open
Abstract
AbstractThe coronavirus disease 2019 (COVID-19) causes a systemic illness that can result in various manifestations. In addition to severe acute respiratory syndrome, patients often exhibit complications unrelated to the respiratory system. Potential liver damage can occur in 14.8 to 53.0% of the affected patients. Liver impairment in COVID-19 can also occur because of the use of polypharmacy during disease management. It is essential to be aware of drug-induced liver injury (DILI) in patients diagnosed with COVID-19, especially when considering the off-label usage of medications in both preventative and therapeutic regimens used on a wide scale. This review aims to give pertinent information regarding drugs utilized thus far in COVID-19 patients and their potential toxicity to the liver. We also present a suggested management approach to DILI in COVID-19 patients and lessons learned from the pharmacological management of this pandemic.
Collapse
|
21
|
McCarthy MW. The first five years of SARS-CoV-2: inpatient treatment updates and future directions. Expert Opin Pharmacother 2024; 25:1873-1878. [PMID: 39305134 DOI: 10.1080/14656566.2024.2408375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION In December 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first identified in adults with pneumonia in Wuhan, China. AREAS COVERED It is now believed that several billion humans have been infected with SARS-CoV-2 and more than ten million have died from coronavirus disease 2019 (COVID-19), the disease caused by SARS-CoV-2. EXPERT OPINION The first five years of the SARS-CoV-2 pandemic have been marked by unfathomable suffering as well as remarkable scientific progress. This manuscript examines what has been learned about the treatment of inpatients with COVID-19 and explores how the therapeutic approach may evolve in the years ahead.
Collapse
|
22
|
Novitsky V, Beckwith CG, Carpenter-Azevedo K, Shin J, Hague J, Sam S, Steingrimsson J, Huard RC, Lethbridge K, Sahu S, Rapoza K, Chandran K, Bazerman L, Hipolito E, Diaz I, Carnevale D, Guang A, Gillani F, Caliendo AM, Kantor R. Limited Short-Term Evolution of SARS-CoV-2 RNA-Dependent RNA Polymerase under Remdesivir Exposure in Upper Respiratory Compartments. Viruses 2024; 16:1511. [PMID: 39459846 PMCID: PMC11512361 DOI: 10.3390/v16101511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND The extent of the SARS-CoV-2 short-term evolution under Remdesivir (RDV) exposure and whether it varies across different upper respiratory compartments are not fully understood. METHODS Patients hospitalized for COVID-19, with or without RDV therapy, were enrolled and completed up to three visits, in which they provided specimens from four respiratory compartments. Near full-length genome SARS-CoV-2 sequences were obtained from viral RNA, standard lineage and variant assignments were performed, and viral mutations in the RNA-dependent RNA polymerase (RdRp) region-the RDV target gene-were detected and compared between participants with and without RDV, across the four compartments, within participants across visits, and versus a larger sequence dataset. The statistical analysis used a generalized linear mixed-effects model. RESULTS A total of 139 sequences were obtained from 37 out of the 44 (84%) enrolled participants. The genotyping success varied across respiratory compartments, which ranged from 42% with oropharyngeal specimens to 67% with nasopharyngeal specimens and showed improvement with higher viral loads. No RdRp mutations known to be associated with RDV resistance were identified, and for 34 detected mutations at 32 amino acid positions that are not known as RDV-associated, there was no evidence of any associations with the RDV exposure, respiratory compartment, or time. At least 1 of these 34 mutations were detected in all participants, and some differed from the larger sequence dataset. CONCLUSIONS This study highlighted the SARS-CoV-2 short-term genomic stability within hosts and across upper respiratory compartments, which suggests a lack of evolution of RDV resistance over time. This contributes to our understanding of SARS-CoV-2 genomic dynamics.
Collapse
Affiliation(s)
- Vladimir Novitsky
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (V.N.); (C.G.B.); (J.H.); (S.S.); (F.G.); (A.M.C.)
| | - Curt G. Beckwith
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (V.N.); (C.G.B.); (J.H.); (S.S.); (F.G.); (A.M.C.)
| | - Kristin Carpenter-Azevedo
- State Health Laboratories, Rhode Island Department of Health, Providence, RI 02912, USA; (K.C.-A.); (R.C.H.)
| | - Jimin Shin
- School of Medicine, University of Connecticut, Farmington, CT 06030, USA;
| | - Joel Hague
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (V.N.); (C.G.B.); (J.H.); (S.S.); (F.G.); (A.M.C.)
| | - Soya Sam
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (V.N.); (C.G.B.); (J.H.); (S.S.); (F.G.); (A.M.C.)
| | - Jon Steingrimsson
- Department of Biostatistics, School of Public Health, Brown University, Providence, RI 02912, USA;
| | - Richard C. Huard
- State Health Laboratories, Rhode Island Department of Health, Providence, RI 02912, USA; (K.C.-A.); (R.C.H.)
| | - Kevin Lethbridge
- Rhode Island Hospital, Providence, RI 02912, USA; (K.L.); (E.H.); (I.D.); (D.C.)
| | - Sujata Sahu
- The Miriam Hospital, Providence, RI 02912, USA; (S.S.); (K.R.); (K.C.); (L.B.)
| | - Kim Rapoza
- The Miriam Hospital, Providence, RI 02912, USA; (S.S.); (K.R.); (K.C.); (L.B.)
| | - Karen Chandran
- The Miriam Hospital, Providence, RI 02912, USA; (S.S.); (K.R.); (K.C.); (L.B.)
| | - Lauri Bazerman
- The Miriam Hospital, Providence, RI 02912, USA; (S.S.); (K.R.); (K.C.); (L.B.)
| | - Evelyn Hipolito
- Rhode Island Hospital, Providence, RI 02912, USA; (K.L.); (E.H.); (I.D.); (D.C.)
| | - Isabella Diaz
- Rhode Island Hospital, Providence, RI 02912, USA; (K.L.); (E.H.); (I.D.); (D.C.)
| | - Daniella Carnevale
- Rhode Island Hospital, Providence, RI 02912, USA; (K.L.); (E.H.); (I.D.); (D.C.)
| | - August Guang
- Computational Biology Core, Center for Computation and Visualization, Brown University, Providence, RI 02912, USA;
| | - Fizza Gillani
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (V.N.); (C.G.B.); (J.H.); (S.S.); (F.G.); (A.M.C.)
- Rhode Island Hospital, Providence, RI 02912, USA; (K.L.); (E.H.); (I.D.); (D.C.)
| | - Angela M. Caliendo
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (V.N.); (C.G.B.); (J.H.); (S.S.); (F.G.); (A.M.C.)
| | - Rami Kantor
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; (V.N.); (C.G.B.); (J.H.); (S.S.); (F.G.); (A.M.C.)
| |
Collapse
|
23
|
Krisht AAH, Grapin K, de Beauchene RC, Bonnet B, Cassagnes L, Evrard B, Adda M, Souweine B, Dupuis C. SARS-CoV2 pneumonia patients admitted to the ICU: Analysis according to clinical and biological parameters and the extent of lung parenchymal lesions on chest CT scan, a monocentric observational study. PLoS One 2024; 19:e0308014. [PMID: 39298399 PMCID: PMC11412649 DOI: 10.1371/journal.pone.0308014] [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: 02/06/2024] [Accepted: 07/16/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND CT-scan and inflammatory and coagulation biomarkers could help in prognostication of COVID-19 in patients on ICU admission. OBJECTIVE The objectives of this study were to measure the prognostic value of the extent of lung parenchymal lesions on computed tomography (CT) and of several coagulation and inflammatory biomarkers, and to explore the characteristics of the patients depending on the extent of lung parenchymal lesions. DESIGN Retrospective monocentric observational study achieved on a dataset collected prospectively. SETTING Medical ICU of the university hospital of Clermont-Ferrand, France. PATIENTS All consecutive adult patients aged ≥18 years admitted between 20 March, 2020 and 31 August, 2021 for COVID-19 pneumonia. INTERVENTIONS Characteristics at baseline and during ICU stay, and outcomes at day 60 were recorded. The extent of lung parenchyma lesions observed on the chest CT performed on admission was established by artificial intelligence software. MEASUREMENTS Several clinical characteristics and laboratory features were collected on admission including plasma interleukin-6, HLA-DR monocytic-expression rate (mHLA-DR), and the extent of lung parenchymal lesions. Factors associated with day-60 mortality were investigated by uni- and multivariate survival analyses. RESULTS 270 patients were included. Inflammation biomarkers including the levels of neutrophils, CRP, ferritin and Il10 were the indices the most associated with the severity of the extent of the lung lesions. Patients with more extensive lung parenchymal lesions (≥ 75%) on admission had higher CRP serum levels. The extent of lung parenchymal lesions was associated with a decrease in the PaO2/FiO2 ratio(p<0.01), fewer ventilatory-free days (p = 0.03), and a higher death rate at day 60(p = 0.01). Extent of the lesion of more than 75% was independently associated with day-60 mortality (aHR = 1.72[1.06; 2.78], p = 0.03). The prediction of death at day 60 was improved when considering simultaneously biological and radiological markers obtained on ICU admission (AUC = 0.78). CONCLUSIONS The extent of lung parenchyma lesions on CT was associated with inflammation, and the combination of coagulation and inflammatory biomarkers and the extent of the lesions predicted the poorest outcomes.
Collapse
Affiliation(s)
- Abed al Hadi Krisht
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Kévin Grapin
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | | | - Benjamin Bonnet
- CHU Clermont-Ferrand, Service d’Immunologie, Clermont-Ferrand, France
- Université Clermont Auvergne, Laboratoire d’Immunologie, ECREIN, UMR1019 UNH, UFR Médecine de Clermont-Ferrand, Clermont-Ferrand, France
| | - Lucie Cassagnes
- CHU Clermont-Ferrand, Service de Radiologie, Clermont-Ferrand, France
- Université Clermont Auvergne, Unité de Nutrition Humaine, INRAe, CRNH Auvergne, Clermont Ferrand, France
| | - Bertrand Evrard
- CHU Clermont-Ferrand, Service d’Immunologie, Clermont-Ferrand, France
- Université Clermont Auvergne, Laboratoire d’Immunologie, ECREIN, UMR1019 UNH, UFR Médecine de Clermont-Ferrand, Clermont-Ferrand, France
| | - Mireille Adda
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Bertrand Souweine
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
- Université Clermont Auvergne, CNRS, LMGE, Clermont-Ferrand, France
| | - Claire Dupuis
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
- Université Clermont Auvergne, Unité de Nutrition Humaine, INRAe, CRNH Auvergne, Clermont Ferrand, France
| |
Collapse
|
24
|
Player B, Huppler AR, Pan AY, Liegl M, Havens PL, Ray K, Mitchell M, Graff K. Safety of remdesivir in the treatment of acute SARS-CoV-2 infection in pediatric patients. BMC Infect Dis 2024; 24:987. [PMID: 39289614 PMCID: PMC11406769 DOI: 10.1186/s12879-024-09833-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Transaminase and creatinine elevations have been well described in adults treated with remdesivir for COVID-19. It is hypothesized that a similar safety profile exists in children with COVID-19 treated with remdesivir, but available data are limited, especially in children < 12 months. The primary aim of this study was to determine the prevalence and timing of elevations in transaminases and creatinine in children with COVID-19 who were treated with remdesivir. METHODS This was a retrospective, observational cohort study including all pediatric patients admitted to a single, freestanding children's hospital who were positive for COVID-19 and received at least 1 dose of remdesivir between 1/1/2020 and 5/31/2022. Available baseline and peak transaminase and creatinine concentrations were evaluated. Multivariable logistic regression analysis was performed to identify risk factors for transaminase elevation. RESULTS A total of 180 patients met inclusion criteria. Creatinine elevation of any grade was noted in 16% and remained elevated only in those with underlying chronic kidney disease. Transaminase elevation of any grade was noted in 58% of patients and remained elevated in only 1%. Older age and critical respiratory disease were associated with higher risk of significant transaminase elevation, whereas non-Hispanic ethnicity was strongly associated with protection against significant transaminase elevation. CONCLUSIONS In our cohort of hospitalized children with COVID-19 who were treated with remdesivir, most patients experienced only mild transaminitis and normal creatinine concentrations. A limited number of patients experienced laboratory abnormalities which were transient, suggesting a favorable safety profile for remdesivir use in pediatrics.
Collapse
Affiliation(s)
- Brittany Player
- Department of Pediatrics, Section of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, USA.
- , PO Box 1997, Suite C450 Pediatric Infectious Diseases, Milwaukee, WI, USA.
| | - Anna R Huppler
- Department of Pediatrics, Section of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Amy Y Pan
- Department of Pediatrics, Section of Quantitative Health Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Melodee Liegl
- Department of Pediatrics, Section of Quantitative Health Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Peter L Havens
- Department of Pediatrics, Section of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Katie Ray
- Department of Enterprise Safety, Children's Wisconsin, Milwaukee, WI, 53201-1997, USA
| | - Michelle Mitchell
- Department of Pediatrics, Section of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Kelly Graff
- Department of Pediatrics, Section of Infectious Diseases, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
25
|
Bansal SB, Babras M, Rana A, Mahapatra A, Yadav DK, Sethi SK. A Prospective Study of Incidence, Risk Factors, and Outcomes of Acute Kidney Injury in Coronavirus Disease 2019. Indian J Nephrol 2024; 34:461-466. [PMID: 39372632 PMCID: PMC11450852 DOI: 10.25259/ijn_399_23] [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: 01/16/2024] [Accepted: 01/25/2024] [Indexed: 10/08/2024] Open
Abstract
Background Acute kidney injury (AKI) is common after coronavirus 2 infection (COVID-19), leading to higher morbidity and mortality. There is little prospective data from India regarding the incidence, risk factors, and outcome of AKI in COVID-19. Materials and Methods This study was conducted prospectively in adult patients between September and December 2020 in a tertiary care hospital in the national capital region of Delhi. A total of 856 patients with COVID-19 infection were enrolled in the study. Survivors were followed for 3 months after discharge. Results Out of 856 patients, 207 (24%) developed AKI. AKI was significantly higher in those with severe disease as compared to mild-moderate disease (88% vs. 12%, P = 0.04). Out of all AKI, 3.4% had stage 1, 9.2% had stage 2, and the rest 87.4% had stage 3 AKI. 183/207 (88%) patients were on mechanical ventilators, 133 (64%) required inotropic support, and 137/207 (83.6%) patients required kidney replacement therapy. Out of 207 AKI patients, 74% (153) died as compared to 4% (27) in non-AKI group (P = 0.0001). After 3 months, chronic kidney disease (CKD) developed in 10/54 (18.5%) patients. On multivariable analysis, the presence of diabetes mellitus, severe COVID-19 disease, high levels of C reactive protein, lactate dehydrogenase, D-Dimer, and use of intravenous steroids, tocilizumab and remdesivir, were found to be significant predictors of AKI. Conclusion AKI is common after COVID-19 infection and it is a significant risk factor for mortality in COVID-19. Patients with diabetes and high levels of inflammatory markers have higher mortality. CKD may develop in many patients after discharge.
Collapse
Affiliation(s)
- Shyam Bihari Bansal
- Department of Nephrology and Kidney Transplantation, Medanta Kidney and Urology Institute, Medanta-The Medicity, Gurgaon, Haryana, India
| | - Mayur Babras
- Department of Nephrology, Seth Nandlal, Dhoot Hospital, Aurangabad, Maharashtra, India
| | - Abhyudaysingh Rana
- Department of Nephrology and Kidney Transplantation, Medanta Kidney and Urology Institute, Medanta-The Medicity, Gurgaon, Haryana, India
| | - Amit Mahapatra
- Department of Nephrology and Kidney Transplantation, Medanta Kidney and Urology Institute, Medanta-The Medicity, Gurgaon, Haryana, India
| | - Dinesh Kumar Yadav
- Department of Nephrology and Kidney Transplantation, Medanta Kidney and Urology Institute, Medanta-The Medicity, Gurgaon, Haryana, India
| | - Sidharth Kumar Sethi
- Department of Nephrology and Kidney Transplantation, Medanta Kidney and Urology Institute, Medanta-The Medicity, Gurgaon, Haryana, India
| |
Collapse
|
26
|
Terzić V, Miantezila Basilua J, Billard N, de Gastines L, Belhadi D, Fougerou-Leurent C, Peiffer-Smadja N, Mercier N, Delmas C, Ferrane A, Dechanet A, Poissy J, Espérou H, Ader F, Hites M, Andrejak C, Greil R, Paiva JA, Staub T, Tacconelli E, Burdet C, Costagliola D, Mentré F, Yazdanpanah Y, Diallo A. Cardiac Adverse Events and Remdesivir in Hospitalized Patients With COVID-19: A Post Hoc Safety Analysis of the Randomized DisCoVeRy Trial. Clin Infect Dis 2024; 79:382-391. [PMID: 38552208 PMCID: PMC11327784 DOI: 10.1093/cid/ciae170] [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: 09/28/2023] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND We aimed to evaluate the cardiac adverse events (AEs) in hospitalized patients with coronavirus disease 2019 (COVID-19) who received remdesivir plus standard of care (SoC) compared with SoC alone (control), as an association was noted in some cohort studies and disproportionality analyses of safety databases. METHODS This post hoc safety analysis is based on data from the multicenter, randomized, open-label, controlled DisCoVeRy trial in hospitalized patients with COVID-19. Any first AE that occurred between randomization and day 29 in the modified intention-to-treat (mITT) population randomized to either remdesivir or control group was considered. Analysis was performed using Kaplan-Meier survival curves, and Kaplan-Meier estimates were calculated for event rates. RESULTS Cardiac AEs were reported in 46 (11.2%) of 410 and 48 (11.3%) of 423 patients in the mITT population (n = 833) enrolled in the remdesivir and control groups, respectively. The difference between both groups was not significant (hazard ratio [HR], 1.0; 95% confidence interval [CI], .7-1.5; P = .98), even when serious and nonserious cardiac AEs were evaluated separately. The majority of reports in both groups were of arrhythmic nature (remdesivir, 84.8%; control, 83.3%) and were associated with a favorable outcome. There was no significant difference between the two groups in the occurrence of cardiac AE subclasses, including arrhythmic events (HR, 1.1; 95% CI, .7-1.7; P = .68). CONCLUSIONS Remdesivir treatment was not associated with an increased risk of cardiac AEs compared with control in patients hospitalized with moderate or severe COVID-19. These results are consistent with other randomized, controlled trials and meta-analyses. Clinical Trials Registration. NCT04315948; EudraCT 2020-000936-23.
Collapse
Affiliation(s)
- Vida Terzić
- Clinical Trial Safety and Public Health, ANRS|Emerging Infectious Diseases, Paris, France
- Clinical Research Safety Department, INSERM, Paris, France
| | - Joe Miantezila Basilua
- Clinical Trial Safety and Public Health, ANRS|Emerging Infectious Diseases, Paris, France
- Clinical Research Safety Department, INSERM, Paris, France
| | - Nicolas Billard
- Department of Epidemiology, Biostatistics and Clinical Research, Hospital Bichat, APHP, Paris, France
| | - Lucie de Gastines
- Clinical Trial Safety and Public Health, ANRS|Emerging Infectious Diseases, Paris, France
- Clinical Research Safety Department, INSERM, Paris, France
| | - Drifa Belhadi
- Department of Epidemiology, Biostatistics and Clinical Research, Hospital Bichat, APHP, Paris, France
- Université Paris Cité, IAME, INSERM, Paris, France
| | - Claire Fougerou-Leurent
- Pharmacology Unit, University Hospital Rennes, CIC Inserm 1414, University Hospital Rennes, Rennes, France
| | - Nathan Peiffer-Smadja
- Infectious Diseases Department, Hôpital Bichat—Claude-Bernard, APHP, Paris, France
- Université Paris Cité, IAME, INSERM, Paris, France
| | - Noémie Mercier
- Clinical Trial Safety and Public Health, ANRS|Emerging Infectious Diseases, Paris, France
- Clinical Research Safety Department, INSERM, Paris, France
| | | | | | - Aline Dechanet
- Department of Epidemiology, Biostatistics and Clinical Research, Hospital Bichat, APHP, Paris, France
| | - Julien Poissy
- UMR 8576—UGSF—Unité de Glycobiologie Structurale et Fonctionnelle, Université de Lille, Inserm U1285, CHU Lille, Pôle de réanimation, CNRS, Lille, France
| | | | - Florence Ader
- Département des Maladies infectieuses et tropicales, Hospices Civils de Lyon, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Inserm 1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, Lyon, France
| | - Maya Hites
- Clinic of Infectious Diseases, Hôpital Universitaire de Bruxelles (HUB)-Erasme, Brussels, Belgium
| | - Claire Andrejak
- Pulmonolgy Unit, University Hospital Amiens-Picardie, UR 4294 AGIR, Université Picardie Jules Verne, Amiens, France
| | - Richard Greil
- IIIrd Medical Department, Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute-Center for clinical Cancer and Immunology Trials (SCRI-CCCIT), Cancer Cluster Salzburg, Austrian Group for Medical Tumor Therapy (AGMT), Salzburg, Austria
| | - José-Artur Paiva
- Serviço de Medicina Intensiva, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Thérèse Staub
- Infectious Diseases Department, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - Evelina Tacconelli
- Infectious Diseases, Dept. Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Charles Burdet
- Department of Epidemiology, Biostatistics and Clinical Research, Hospital Bichat, APHP, Paris, France
| | - Dominique Costagliola
- Sorbonne Université, INSERM, Institut Pierre Louis d’Épidémiologie et de Santé Publique, Paris, France
| | - France Mentré
- Department of Epidemiology, Biostatistics and Clinical Research, Hospital Bichat, APHP, Paris, France
- Université Paris Cité, IAME, INSERM, Paris, France
| | - Yazdan Yazdanpanah
- Université Paris Cité, IAME, INSERM, Paris, France
- Infectious Diseases Department, Hôpital Bichat—Claude-Bernard, APHP, Paris, France
- ANRS|Emerging Infectious Diseases, Paris, France
| | - Alpha Diallo
- Clinical Trial Safety and Public Health, ANRS|Emerging Infectious Diseases, Paris, France
- Clinical Research Safety Department, INSERM, Paris, France
| |
Collapse
|
27
|
Okoli GN, Reddy VK, Lam OL, Askin N, Rabbani R. Update on efficacy of the approved remdesivir regimen for treatment of COVID-19: a systematic review with meta-analysis and trial sequential analysis of randomized controlled trials. Curr Med Res Opin 2024; 40:1277-1287. [PMID: 38850519 DOI: 10.1080/03007995.2024.2366443] [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: 03/28/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Efficacy of remdesivir for COVID-19 remains unclear. We updated our published systematic review to better inform on the use of remdesivir for COVID-19. METHODS We searched for randomized controlled trials (RCTs) among hospitalized COVID-19 patients. Meta-analysis was conducted using an inverse variance, random-effects model, presenting relative risk (RR) or mean difference (MD) and their associated 95% confidence intervals (CIs). Statistical heterogeneity was calculated using the I2 statistic. In addition, we conducted trial sequential analysis (TSA). Outcomes with additional data were clinical progression, hospitalization days, and all-cause mortality. RESULTS We included nine RCTs (12,876 individuals). Three trials each were of a low, unclear, and a high risk of bias. Compared with no treatment/placebo, remdesivir (100 mg daily, over 10 days) significantly improved clinical progression (RR 1.06, CI 1.02-1.11), but did not significantly reduce hospitalization days (MD -0.48, CI -2.18-1.21) and all-cause mortality (RR 0.92, CI 0.84-1.01). TSA suggested that further information is not required to conclude on the efficacy of remdesivir in improving clinical progression, and that, while more information is required for hospitalization days and all-cause mortality, further RCTs to prove fewer hospitalization days may be futile, as efficacy of remdesivir for this outcome is unlikely. CONCLUSIONS Remdesivir appeared promising for COVID-19, but there is insufficient evidence of its efficacy. High quality RCTs are needed for a stronger evidence base.
Collapse
Affiliation(s)
- George N Okoli
- George and Fay Yee Centre for Healthcare Innovation, Max Rady College of Medicine, , University of Manitoba, Winnipeg, Canada
- College of Pharmacy, University of Manitoba, Winnipeg, Canada
| | - Viraj K Reddy
- George and Fay Yee Centre for Healthcare Innovation, Max Rady College of Medicine, , University of Manitoba, Winnipeg, Canada
| | - Otto Lt Lam
- George and Fay Yee Centre for Healthcare Innovation, Max Rady College of Medicine, , University of Manitoba, Winnipeg, Canada
| | - Nicole Askin
- Neil John Maclean Health Sciences Library, University of Manitoba, Winnipeg, Canada
| | - Rasheda Rabbani
- George and Fay Yee Centre for Healthcare Innovation, Max Rady College of Medicine, , University of Manitoba, Winnipeg, Canada
| |
Collapse
|
28
|
Abogunrin S, Adelakun A, Akinola T, Bashir U, Fagbohungbe B, Mueller E, Neeser K, Ogunnubi O, Parekh K. Challenges of consolidating evidence collected during a pandemic and lessons for the future. Curr Med Res Opin 2024; 40:1311-1322. [PMID: 38975733 DOI: 10.1080/03007995.2024.2377676] [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: 12/07/2023] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVE To illustrate the challenges encountered when gathering rapidly synthesized evidence in response to the coronavirus disease 2019 (COVID-19) pandemic. METHODS In this article, we describe the challenges encountered when we performed a systematic literature review (SLR) of randomized controlled trials (RCTs) on the efficacy and safety of treatments for severe COVID-19. The methods of the SLR are described in full, to show the context of our objectives. Then we use the results of the SLR to demonstrate the problems of producing synthesized evidence in this setting. RESULTS Various challenges were identified during this SLR. These were primarily a result of heterogeneity in the study methodology of eligible studies. Definitions of the patient populations and outcome measurements were highly variable and the majority of studies demonstrated a high risk of bias, preventing quantitative synthesis of the collated evidence. CONCLUSION Consolidating evidence from RCTs evaluating COVID-19 interventions was problematic. Guidance is needed for scenarios with high rapid output in primary research.
Collapse
Affiliation(s)
| | - Alex Adelakun
- Manchester University Foundation Trust, Manchester, UK
| | | | - Usman Bashir
- Community Medicine Department, Bayero University Kano, Kano, Nigeria
| | | | | | | | - Oluseun Ogunnubi
- Department of Psychiatry, College of Medicine of the University of Lagos, Lagos, Nigeria
| | | |
Collapse
|
29
|
Anand P, D’Andrea E, Feldman W, Wang SV, Liu J, Brill G, DiCesare E, Lin KJ. A Dynamic Prognostic Model for Identifying Vulnerable COVID-19 Patients at High Risk of Rapid Deterioration. Pharmacoepidemiol Drug Saf 2024; 33:e5872. [PMID: 39135513 PMCID: PMC11418916 DOI: 10.1002/pds.5872] [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: 12/12/2023] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 09/25/2024]
Abstract
PURPOSE We aimed to validate and, if performance was unsatisfactory, update the previously published prognostic model to predict clinical deterioration in patients hospitalized for COVID-19, using data following vaccine availability. METHODS Using electronic health records of patients ≥18 years, with laboratory-confirmed COVID-19, from a large care-delivery network in Massachusetts, USA, from March 2020 to November 2021, we tested the performance of the previously developed prediction model and updated the prediction model by incorporating data after availability of COVID-19 vaccines. We randomly divided data into development (70%) and validation (30%) cohorts. We built a model predicting worsening in a published severity scale in 24 h by LASSO regression and evaluated performance by c-statistic and Brier score. RESULTS Our study cohort consisted of 8185 patients (Development: 5730 patients [mean age: 62; 44% female] and Validation: 2455 patients [mean age: 62; 45% female]). The previously published model had suboptimal performance using data after November 2020 (N = 4973, c-statistic = 0.60. Brier score = 0.11). After retraining with the new data, the updated model included 38 predictors including 18 changing biomarkers. Patients hospitalized after Jun 1st, 2021 (when COVID-19 vaccines became widely available in Massachusetts) were younger and had fewer comorbidities than those hospitalized before. The c-statistic and Brier score were 0.77 and 0.13 in the development cohort, and 0.73 and 0.14 in the validation cohort. CONCLUSION The characteristics of patients hospitalized for COVID-19 differed substantially over time. We developed a new dynamic model for rapid progression with satisfactory performance in the validation set.
Collapse
Affiliation(s)
- Priyanka Anand
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Elvira D’Andrea
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - William Feldman
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Shirley V. Wang
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Jun Liu
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Gregory Brill
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Elyse DiCesare
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
| | - Kueiyu Joshua Lin
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School
| |
Collapse
|
30
|
Fujita Y, Hatazaki M, Fujimi S. Impact of Diabetes Mellitus On In-Hospital Mortality of COVID-19 Patients in Japan Since COVID-19 Became a Common Infectious Disease. Cureus 2024; 16:e66373. [PMID: 39246912 PMCID: PMC11378744 DOI: 10.7759/cureus.66373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
AIM The number of severe cases of coronavirus disease 2019 (COVID-19) has been decreasing since the emergence of the Omicron variant at the end of 2021. COVID-19 has become a common infectious disease in Japan and was downgraded to a category five infectious disease on May 8, 2023. This study aimed to compare the impact of diabetes mellitus on in-hospital mortality in COVID-19 patients since COVID-19 became a common infectious disease. PATIENTS AND METHODS We conducted a retrospective observational study using data from an advanced critical care center in Osaka, Japan. The study included 1,381 patients of COVID-19 admitted to the center between March 1, 2020, and May 7, 2023, before COVID-19 became a category five infectious disease in Japan. Individuals younger than 18 years and pregnant women were excluded. We divided the patients into two groups: pre- and post-Omicron epidemic groups. The primary endpoint of the study was the in-hospital mortality, and the prognostic impact of diabetes mellitus was compared between the groups. RESULTS The Kaplan-Meier curve showed a significantly lower rate of in-hospital mortality in the post-Omicron epidemic group than in the pre-Omicron epidemic group. The hazard ratio (HR) was 1.83 (95% CI, 1.36-2.50; p < 0.0001). Patients with diabetes mellitus had higher in-hospital mortality in both the pre- and post-Omicron epidemic groups; their HRs were 1.39 (95% CI, 1.21-1.59; p < 0.0001) and 1.45 (95% CI, 1.15-1.83; p = 0.0012), respectively. Diabetes mellitus had no significant interaction effect on the association between the post-Omicron epidemic and in-hospital mortality (p for interaction = 0.2154). CONCLUSION Diabetes mellitus may continue contributing to COVID-19 in-hospital mortality in the future, as the Omicron sub-strain may still be prevalent.
Collapse
Affiliation(s)
- Yohei Fujita
- Department of Diabetes and Endocrinology, Osaka General Medical Center, Osaka, JPN
| | - Masahiro Hatazaki
- Department of Diabetes and Endocrinology, Osaka General Medical Center, Osaka, JPN
| | - Satoshi Fujimi
- Division of Trauma and Surgical Critical Care, Osaka General Medical Center, Osaka, JPN
| |
Collapse
|
31
|
Khan U, Mubariz M, Khlidj Y, Nasir MM, Ramadan S, Saeed F, Muhammad A, Abuelazm M. Safety and Efficacy of Camostat Mesylate for Covid-19: a systematic review and Meta-analysis of Randomized controlled trials. BMC Infect Dis 2024; 24:709. [PMID: 39030491 PMCID: PMC11264738 DOI: 10.1186/s12879-024-09468-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 06/03/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Camostat mesylate, an oral serine protease inhibitor, is a powerful TMPRSS2 inhibitor and has been reported as a possible antiviral treatment against COVID-19. Therefore, we aim to assess the safety and efficacy of camostat mesylate for COVID-19 treatment. METHODS A systematic review and meta-analysis synthesizing randomized controlled trials from PubMed, Scopus, Embase, Cochrane, Web of Science, clinical trials.gov, and medrxiv until June 2023. The outcomes were pooled using Mean difference (MD) for continuous outcomes and risk ratio (RR) for dichotomous outcomes. The protocol is registered in PROSPERO with ID CRD42023439633. RESULTS Nine RCTs, including 1,623 patients, were included in this analysis. There was no difference between camostat mesylate and placebo in producing negative PCR test results at 1-7 days (RR: 0.76, 95% CI: [0.54, 1.06] P = 0.1), 8-14 days (RR: 1.02, 95% CI: [0.84, 1.23] P = 0.87), or 15-21 days (RR: 0.99, 95% CI: [0.82, 1.19] P = 0.90); clinical resolution of symptoms at 1-7 days (RR: 0.94 (95% CI: 0.58, 1.53) P = 0.81), 8-14 days (RR: 0.91, 95% CI: [0.74, 1.11] P = 0.33, ), or 15-21 days (RR: 0.77, 95% CI: [0.40, 1.51] P = 0.45); and time to symptom improvement (MD:-0.38 weeks (95% CI: [-1.42, 0.66] P = 0.47, I2 = 85%). CONCLUSION Camostat mesylate did not improve clinical outcomes in patients with COVID-19, compared to placebo.
Collapse
Affiliation(s)
- Ubaid Khan
- King Edward Medical University, Lahore, Pakistan.
| | | | - Yehya Khlidj
- Faculty of medicine, Algiers University, Alger Centre, Algeria
| | | | | | - Fatima Saeed
- King Edward Medical University, Lahore, Pakistan
| | | | | |
Collapse
|
32
|
Nasir N, Khanum I, Habib K, Wagley A, Arshad A, Majeed A. Insight into COVID-19 associated liver injury: Mechanisms, evaluation, and clinical implications. HEPATOLOGY FORUM 2024; 5:139-149. [PMID: 39006140 PMCID: PMC11237249 DOI: 10.14744/hf.2023.2023.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/25/2023] [Accepted: 11/02/2023] [Indexed: 07/16/2024]
Abstract
COVID-19 has affected millions worldwide, causing significant morbidity and mortality. While predominantly involving the respiratory tract, SARS-CoV-2 has also caused systemic illnesses involving other sites. Liver injury due to COVID-19 has been variably reported in observational studies. It has been postulated that liver damage may be due to direct damage by the SARS-CoV-2 virus or multifactorial secondary to hepatotoxic therapeutic options, as well as cytokine release syndrome and sepsis-induced multiorgan dysfunction. The approach to a COVID-19 patient with liver injury requires a thorough evaluation of the pattern of hepatocellular injury, along with the presence of underlying chronic liver disease and concurrent medications which may cause drug-induced liver injury. While studies have shown uneventful recovery in the majority of mildly affected patients, severe COVID-19 associated liver injury has been associated with higher mortality, prolonged hospitalization, and greater morbidity in survivors. Furthermore, its impact on long-term outcomes remains to be ascertained as recent studies report an association with metabolic-fatty liver disease. This present review provides insight into the subject by describing the postulated mechanism of liver injury, its impact in the presence of pre-existing liver disease, and its short- and long-term clinical implications.
Collapse
Affiliation(s)
- Nosheen Nasir
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Iffat Khanum
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Kiren Habib
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Abdullah Wagley
- Research Facilitation Office, Medical College, Aga Khan University, Karachi, Pakistan
| | - Aleena Arshad
- Section of Adult Infectious Diseases, Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Atif Majeed
- Section of Gastroenterology, Department of Medicine, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
33
|
Chang MC, Wu PF, Ho YC, Lin WY, Wu CY, Liu SY, Liu CJ, Lin YT. Clinical outcomes and safety of remdesivir in hospitalized individuals with COVID-19, with or without severe renal impairment. J Infect Public Health 2024; 17:102460. [PMID: 38820894 DOI: 10.1016/j.jiph.2024.05.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/01/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND The use of remdesivir in patients with coronavirus disease 2019 (COVID-19) and severe renal impairment has been approved; however, limited clinical data exist. Accordingly, we aimed to compare outcomes and adverse events associated with remdesivir in hospitalized patients with COVID-19, with and without severe renal impairment. METHODS Hospitalized patients with COVID-19 undergoing a 5-day remdesivir course at Taipei Veterans General Hospital from April 1 to July 31, 2022, were enrolled. Comparative analysis of outcomes and safety between patients with or without severe renal impairment (estimated glomerular filtration rate of < 30 mL/min per 1.73 m2) were conducted. Prognostic factors associated with 28-day mortality in patients with severe renal impairment were investigated using logistic regression analysis. RESULTS A total of 671 hospitalized patients, including 132 patients with severe renal impairment, who received a 5-day course of remdesivir were analyzed. The 28-day mortality was higher in patients with severe renal impairment than in patients without severe renal impairment (15.2% vs. 7.8%). The proportion of patients with acute kidney injury (AKI) and deteriorated liver function after completing remdesivir therapy was similar between the patients with and without severe renal impairment, and the recovery rate of AKI was similar in both groups. The sequential organ failure assessment score was an independent factor associated with 28-day mortality in patients with severe renal impairment. CONCLUSIONS Remdesivir was well-tolerated in hospitalized patients with COVID-19, regardless of renal function. Our findings support the recent recommendation to administer remdesivir in patients with severe renal impairment.
Collapse
Affiliation(s)
- Min-Chi Chang
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ping-Feng Wu
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chien Ho
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wen-Ying Lin
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Ying Wu
- Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Szu-Yu Liu
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Jen Liu
- Division of Transfusion Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Yi-Tsung Lin
- Division of Infectious Diseases, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Emergency and Critical Care Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
34
|
Zhang R, Wei R, Yuan Y, Li N, Hu Y, Chan KH, Hung IFN, Tse HF. Human-induced pluripotent stem cell-derived hepatocyte platform in modeling of SARS-CoV-2 infection. JGH Open 2024; 8:e13039. [PMID: 39006099 PMCID: PMC11239974 DOI: 10.1002/jgh3.13039] [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: 06/27/2023] [Revised: 12/31/2023] [Accepted: 02/08/2024] [Indexed: 07/16/2024]
Abstract
Background and Aim Currently, SARS-CoV-2 is still spreading rapidly and globally. A large proportion of patients with COVID-19 developed liver injuries. The human-induced pluripotent stem cell (iPSC)-derived hepatocytes recapitulate primary human hepatocytes and have been widely used in studies of liver diseases. Methods To explore the susceptibility of hepatocytes to SARS-CoV-2, we differentiated iPSCs to functional hepatocytes and tried infecting them with different MOI (1, 0.1, 0.01) of SARS-CoV-2. Results The iPSC-derived hepatocytes are highly susceptible to virus infection, even at 0.01 MOI. Other than the ancestral strain, iHeps also support the replication of SARS-CoV-2 variants including alpha, beta, theta, and delta. More interestingly, the ACE2 expression significantly upregulated after infection, suggesting a vicious cycle between virus infection and liver injury. Conclusions The iPSC-derived hepatocytes can support the replication of SARS-CoV-2, and this platform could be used to investigate the SARS-CoV-2 hepatotropism and hepatic pathogenic mechanisms.
Collapse
Affiliation(s)
- Ruiqi Zhang
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Rui Wei
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
- Department of Gastroenterology and Hepatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences) Southern Medical University Guangzhou China
- Center for Translational Stem Cell Biology Hong Kong SAR China
| | - Yangyang Yuan
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
- Center for Translational Stem Cell Biology Hong Kong SAR China
| | - Na Li
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Yang Hu
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Kwok-Hung Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
| | - Hung-Fat Tse
- Department of Medicine, Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong SAR China
- Center for Translational Stem Cell Biology Hong Kong SAR China
- Cardiac and Vascular Center Hong Kong University Shenzhen Hospital Shenzhen China
- Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine The University of Hong Kong Hong Kong SAR China
| |
Collapse
|
35
|
Abedipour F, Mirzaei HH, Ansari H, Ehsanzadeh N, Rashki A, Vahedi MM, Rashki A. Remdesivir-Related Cardiac Adverse Effects in COVID-19 Patients: A Case-Control Study. Drug Res (Stuttg) 2024; 74:290-295. [PMID: 38968952 DOI: 10.1055/a-2332-3253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
BACKGROUND There have been reports of serious side effects of Remdesivir, including cardiovascular complications. The present study aimed to determine the adverse cardiovascular effects of Remdesivir and the factors affecting them in COVID-19 patients. METHODS The patients were classified into two groups: those receiving Remdesivir without cardiac complications and those receiving Remdesivir with cardiovascular complications. After reviewing the patient's medical records, the relationship of some factors with the incidence of adverse cardiovascular effects was measured. RESULTS Chi-square test showed that the distribution of complications in men was significantly higher than in women (P=0.001). The independent t-test revealed that the mean age in the group with complications was significantly higher than the group without complications (P=0.013). Fisher's exact test demonstrated a significant relationship between smoking and cardiovascular complications (P=0.05). According to the Mann-Whitney test, a significant difference was found in the mean changes of Bilirubin (P=0.02) and ALKP (P=0.01) before and after treatment in the groups with and without heart complications. CONCLUSION Our findings indicated that most of the COVID-19 patients suffered from sinus bradycardia, and the distribution of complications was more pronounced in men than in women. The mean age in the group with complications was higher than the group without complications. Smoking was found to be associated with the occurrence of cardiovascular complications and the mean changes of Bilirubin and ALKP before and after treatment were significantly different in the groups with and without cardiovascular complications.
Collapse
Affiliation(s)
- Fatemah Abedipour
- Department of Infectious Disease, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
- Infectious Diseases and Tropical Medicine Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Hossein Hadavand Mirzaei
- Department of Infectious Disease, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Hossein Ansari
- Health Promotion Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Neda Ehsanzadeh
- Department of Cardiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Amin Rashki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohammad Mahdi Vahedi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Pharmacology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Asma Rashki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Pharmacology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
36
|
Bhimraj A, Morgan RL, Shumaker AH, Baden L, Cheng VCC, Edwards KM, Gallagher JC, Gandhi RT, Muller WJ, Nakamura MM, O’Horo JC, Shafer RW, Shoham S, Murad MH, Mustafa RA, Sultan S, Falck-Ytter Y. Infectious Diseases Society of America Guidelines on the Treatment and Management of Patients With COVID-19 (September 2022). Clin Infect Dis 2024; 78:e250-e349. [PMID: 36063397 PMCID: PMC9494372 DOI: 10.1093/cid/ciac724] [Citation(s) in RCA: 81] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 02/07/2023] Open
Abstract
There are many pharmacologic therapies that are being used or considered for treatment of coronavirus disease 2019 (COVID-19), with rapidly changing efficacy and safety evidence from trials. The objective was to develop evidence-based, rapid, living guidelines intended to support patients, clinicians, and other healthcare professionals in their decisions about treatment and management of patients with COVID-19. In March 2020, the Infectious Diseases Society of America (IDSA) formed a multidisciplinary guideline panel of infectious disease clinicians, pharmacists, and methodologists with varied areas of expertise to regularly review the evidence and make recommendations about the treatment and management of persons with COVID-19. The process used a living guideline approach and followed a rapid recommendation development checklist. The panel prioritized questions and outcomes. A systematic review of the peer-reviewed and grey literature was conducted at regular intervals. The Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach was used to assess the certainty of evidence and make recommendations. Based on the most recent search conducted on 31 May 2022, the IDSA guideline panel has made 32 recommendations for the treatment and management of the following groups/populations: pre- and postexposure prophylaxis, ambulatory with mild-to-moderate disease, and hospitalized with mild-to-moderate, severe but not critical, and critical disease. As these are living guidelines, the most recent recommendations can be found online at: https://idsociety.org/COVID19guidelines. At the inception of its work, the panel has expressed the overarching goal that patients be recruited into ongoing trials. Since then, many trials were conducted that provided much-needed evidence for COVID-19 therapies. There still remain many unanswered questions as the pandemic evolved, which we hope future trials can answer.
Collapse
Affiliation(s)
- Adarsh Bhimraj
- Division of Infectious Diseases, Houston Methodist Hospital, Houston, Texas
| | - Rebecca L Morgan
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Case Western Reserve University, School of Medicine, Cleveland, Ohio
| | - Amy Hirsch Shumaker
- Department of Medicine, Case Western Reserve University, School of Medicine, Cleveland, Ohio
- VA Northeast Ohio Healthcare System, Cleveland, Ohio
| | | | - Vincent Chi Chung Cheng
- Queen Mary Hospital, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kathryn M Edwards
- Division of Infectious Diseases, Department of Pediatrics, Vanderbilt University Medical Center,Nashville, Tennessee
| | - Jason C Gallagher
- Department of Pharmacy Practice, Temple University, Philadelphia, Pennsylvania
| | - Rajesh T Gandhi
- Infectious Diseases Division, Department of Medicine, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts
| | - William J Muller
- Division of Pediatric Infectious Diseases, Ann & Robert H. Lurie Children’s Hospital of Chicago and Northwestern University, Chicago, Illinois
| | - Mari M Nakamura
- Antimicrobial Stewardship Program and Division of Infectious Diseases, Boston Children’s Hospital and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - John C O’Horo
- Division of Infectious Diseases, Joint Appointment Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota
| | - Robert W Shafer
- Division of Infectious Diseases, Department of Medicine, Stanford University, Palo Alto, California
| | - Shmuel Shoham
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - M Hassan Murad
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic, Rochester, Minnesota
| | - Reem A Mustafa
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Shahnaz Sultan
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis VA Healthcare System, Minneapolis, Minnesota
| | - Yngve Falck-Ytter
- Department of Medicine, Case Western Reserve University, School of Medicine, Cleveland, Ohio
- VA Northeast Ohio Healthcare System, Cleveland, Ohio
| |
Collapse
|
37
|
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
|
38
|
Kinsella PM, Moso MA, Morrissey CO, Dendle C, Guy S, Bond K, Sasadeusz J, Slavin MA. Antiviral therapies for the management of persistent coronavirus disease 2019 in immunocompromised hosts: A narrative review. Transpl Infect Dis 2024; 26:e14301. [PMID: 38809102 DOI: 10.1111/tid.14301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/11/2024] [Accepted: 05/08/2024] [Indexed: 05/30/2024]
Abstract
Antiviral agents with activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have played a critical role in disease management; however, little is known regarding the efficacy of these medications in the treatment of SARS-CoV-2 infection in immunocompromised patients, particularly in the management of persistent SARS-CoV-2 positivity. This narrative review discusses the management of persistent coronavirus disease 2019 in immunocompromised hosts, with a focus on antiviral therapies. We identified 84 cases from the literature describing a variety of approaches, including prolonged antiviral therapy (n = 11), combination antivirals (n = 13), and mixed therapy with antiviral and antibody treatments (n = 60). A high proportion had an underlying haematologic malignancy (n = 67, 80%), and were in receipt of anti-CD20 agents (n = 51, 60%). Success was reported in 70 cases (83%) which varied according to the therapy type. Combination therapies with antivirals may be an effective approach for individuals with persistent SARS-CoV-2 positivity, particularly those that incorporate treatments aimed at increasing neutralizing antibody levels. Any novel approaches taken to this difficult management dilemma should be mindful of the emergence of antiviral resistance.
Collapse
Affiliation(s)
- Paul M Kinsella
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Infectious Diseases, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Michael A Moso
- Department of Infectious Diseases, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | | | - Claire Dendle
- Monash Infectious Diseases, Monash Health, Melbourne, Australia
- School of Clinical Sciences, Monash University, Melbourne, Australia
| | - Stephen Guy
- Department of Infectious Diseases, Eastern Health, Melbourne, Australia
- Eastern Health Clinical School, Monash University, Melbourne, Australia
| | - Katherine Bond
- Department of Microbiology, Royal Melbourne Hospital, Melbourne, Australia
- Victorian Infectious Diseases Reference Laboratory (VIDRL) at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Joseph Sasadeusz
- Department of Infectious Diseases, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Monica A Slavin
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Doherty Institute of Infection and Immunity, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
39
|
Salvadori N, Fridman M, Chiang M, Chen L, Wang C, Lee E, Fonseca V, Fusco DN, Jourdain G, Drouin AC. Real-world evidence of survival benefit of remdesivir: study of 419 propensity score-matched patients hospitalized over the alpha and delta waves of COVID-19 in New Orleans, LA. Front Med (Lausanne) 2024; 11:1390164. [PMID: 38818394 PMCID: PMC11137210 DOI: 10.3389/fmed.2024.1390164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024] Open
Abstract
Background The direct acting antiviral remdesivir (RDV) has shown promising results in randomized clinical trials. This study is a unique report of real clinical practice RDV administration for COVID-19 from alpha through delta variant circulation in New Orleans, Louisiana (NOLA). Patients in NOLA have among US worst pre-COVID health outcomes, and the region was an early epicenter for severe COVID. Methods Data were directly extracted from electronic medical records through REACHnet. Of 9,106 adults with COVID, 1,928 were admitted to inpatient care within 7 days of diagnosis. The propensity score is based upon 22 selected covariates, related to both RDV assignment and outcome of interest. RDV and non-RDV patients were matched 1:1 with replacement, by location and calendar period of admission. Primary and secondary endpoints were, death from any cause and inpatient discharge, within 28 and 14 days after inpatient admission. Results Of 448 patients treated with RDV, 419 (94%) were successfully matched to a non-RDV patient. 145 (35%) patients received RDV for < 5 days, 235 (56%) for 5 days, and 39 (9%) for > 5 days. 96% of those on RDV received it within 2 days of admission. RDV was more frequently prescribed in patients with pneumonia (standardized difference: 0.75), respiratory failure, hypoxemia, or dependence on supplemental oxygen (0.69), and obesity (0.35) within 5 days prior to RDV initiation or corresponding day in non-RDV patients (index day). RDV patients were numerically more likely to be on steroids within 5 days prior to index day (86 vs. 82%) and within 7 days after inpatient admission (96 vs. 87%). RDV was significantly associated with lower risk of death within 14 days after admission (hazard ratio [HR]: 0.37, 95% CI: 0.19 to 0.69, p = 0.002) but not within 28 days (HR: 0.62, 95% CI: 0.36 to 1.07, p = 0.08). Discharge within 14 days of admission was significantly more likely for RDV patients (p < 0.001) and numerically more likely within 28 days after admission (p = 0.06). Conclusion Overall, our findings support recommendation of RDV administration for COVID-19 in a highly comorbid, highly impoverished population representative of both Black and White subjects in the US Gulf South.
Collapse
Affiliation(s)
- Nicolas Salvadori
- Department of Statistics, Faculty of Science, Chiang Mai University, Chiang Mai, Thailand
- Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | | | - Mel Chiang
- Gilead Sciences, Inc., Foster City, CA, United States
| | - Linda Chen
- Gilead Sciences, Inc., Foster City, CA, United States
| | - ChenYu Wang
- Gilead Sciences, Inc., Foster City, CA, United States
| | - EunYoung Lee
- Gilead Sciences, Inc., Foster City, CA, United States
| | - Vivian Fonseca
- Endocrinology Section, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Dahlene N. Fusco
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, United States
- University Medical Center, New Orleans, LA, United States
| | - Gonzague Jourdain
- Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Arnaud C. Drouin
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States
- University Medical Center, New Orleans, LA, United States
| |
Collapse
|
40
|
Li J, de Melo Jorge DM, Wang W, Sun S, Frum T, Hang YA, Liu Y, Zhou X, Xiao J, Wang X, Spence JR, Wobus CE, Zhu HJ. Differential Bioactivation Profiles of Different GS-441524 Prodrugs in Cell and Mouse Models: ProTide Prodrugs with High Cell Permeability and Susceptibility to Cathepsin A Are More Efficient in Delivering Antiviral Active Metabolites to the Lung. J Med Chem 2024; 67:7470-7486. [PMID: 38690769 PMCID: PMC11246197 DOI: 10.1021/acs.jmedchem.4c00234] [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] [Indexed: 05/03/2024]
Abstract
We assessed factors that determine the tissue-specific bioactivation of ProTide prodrugs by comparing the disposition and activation of remdesivir (RDV), its methylpropyl and isopropyl ester analogues (MeRDV and IsoRDV, respectively), the oral prodrug GS-621763, and the parent nucleotide GS-441524 (Nuc). RDV and MeRDV yielded more active metabolite remdesivir-triphosphate (RDV-TP) than IsoRDV, GS-621763, and Nuc in human lung cell models due to superior cell permeability and higher susceptivity to cathepsin A. Intravenous administration to mice showed that RDV and MeRDV delivered significantly more RDV-TP to the lung than other compounds. Nevertheless, all four ester prodrugs exhibited very low oral bioavailability (<2%), with Nuc being the predominant metabolite in blood. In conclusion, ProTides prodrugs, such as RDV and MeRDV, are more efficient in delivering active metabolites to the lung than Nuc, driven by high cell permeability and susceptivity to cathepsin A. Optimizing ProTides' ester structures is an effective strategy for enhancing prodrug activation in the lung.
Collapse
Affiliation(s)
- Jiapeng Li
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109, USA
| | - Daniel Macedo de Melo Jorge
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Weiwen Wang
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109, USA
| | - Shuxin Sun
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109, USA
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109, USA
| | - Tristan Frum
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Yu-An Hang
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109, USA
| | - Yueting Liu
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109, USA
| | - Xingwu Zhou
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109, USA
| | - Jingcheng Xiao
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109, USA
| | - Xinwen Wang
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University College of Pharmacy, Rootstown, Ohio 44272, USA
| | - Jason R. Spence
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan 48109, USA
| | - Christiane E. Wobus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Hao-Jie Zhu
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
41
|
Sari H, Putri HH, Paksi PW, Hidayat G, Amelia SR, Sundari CDD, Rachmawati H, Ivansyah AL, Muttaqien F, Iskandar F. Theoretical Investigation of the Green-Synthesized Carbon-Based Nanomaterial Potential as Inhibitors of ACE2 for Blocking SARS-CoV-2 Binding. ACS OMEGA 2024; 9:16701-16715. [PMID: 38617634 PMCID: PMC11007854 DOI: 10.1021/acsomega.4c00759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/01/2024] [Accepted: 03/12/2024] [Indexed: 04/16/2024]
Abstract
Since the emergence of SARS-CoV-2 in 2020, the world has faced a global pandemic, emphasizing the urgent need for effective treatments to combat COVID-19. This study explores the use of green-synthesized carbon-based nanomaterials as potential inhibitors of ACE2, a critical receptor for SARS-CoV-2 entry into host cells. Specifically, the study examines four carbon-based nanomaterials, namely, CD1, CD2, CD3, and CD4 in amino, graphitic, pyridinic, and pyrrolic forms, respectively, synthesized from curcumin, to investigate their binding affinity with ACE2. Molecular docking studies revealed that CD3 (pyridinic form) exhibited the highest binding affinity with ACE2, surpassing that of the control compound, curcumin. Notably, CD3 formed hydrophobic interactions and hydrogen bonds with key ACE2 residues, suggesting its potential to block the binding of SARS-CoV-2 to human cells. Moreover, molecular dynamics simulations demonstrated the stability of these ligand-ACE2 complexes, further supporting the promise of CD3 as an inhibitor. Quantum chemical analyses, including frontier molecular orbitals, natural bond orbital analysis, and the quantum theory of atoms in molecules, unveiled valuable insights into the reactivity and interaction strengths of these ligands. CD3 exhibited desirable chemical properties, signifying its suitability for therapeutic development. The study's findings suggest that green-synthesized carbon-based nanomaterials, particularly CD3, have the potential to serve as effective inhibitors of ACE2, offering a promising avenue for the development of treatments against COVID-19. Further experimental validation is warranted to advance these findings and establish new therapies for the ongoing global pandemic.
Collapse
Affiliation(s)
- Harsiwi
Candra Sari
- Master
Program in Computational Science, Faculty of Mathematics and Natural
Sciences, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
| | - Haliza Hasnia Putri
- Master
Program in Computational Science, Faculty of Mathematics and Natural
Sciences, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
| | - Pinantun Wiguna
Kusuma Paksi
- Master
Program in Computational Science, Faculty of Mathematics and Natural
Sciences, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
| | - Gabriel Hidayat
- Master
Program in Computational Science, Faculty of Mathematics and Natural
Sciences, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
| | - Silmi Rahma Amelia
- Master
Program in Computational Science, Faculty of Mathematics and Natural
Sciences, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
| | - Citra Deliana Dewi Sundari
- Department
of Chemistry, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
- Chemistry
Education, Universitas Islam Negeri Sunan
Gunung Djati Bandung, Jl. A. H. Nasution No. 105, Bandung, West Java 40614, Indonesia
| | - Heni Rachmawati
- School
of Pharmacy, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
- Research
Center for Nanosciences and Nanotechnology, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
| | - Atthar Luqman Ivansyah
- Master
Program in Computational Science, Faculty of Mathematics and Natural
Sciences, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
- Department
of Physics, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
| | - Fahdzi Muttaqien
- Master
Program in Computational Science, Faculty of Mathematics and Natural
Sciences, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
- Department
of Physics, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
| | - Ferry Iskandar
- Research
Center for Nanosciences and Nanotechnology, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
- Department
of Physics, Institut Teknologi Bandung, Jalan Ganesha 10, Bandung, West Java 40132, Indonesia
- Collaboration
Research Center for Advanced Energy Materials, National Research and Innovation Agency - Institut Teknologi Bandung, Jl. Ganesha 10∇, Bandung 40132, Indonesia
| |
Collapse
|
42
|
Selman CJ, Lee KJ, Ferguson KN, Whitehead CL, Manley BJ, Mahar RK. Statistical analyses of ordinal outcomes in randomised controlled trials: a scoping review. Trials 2024; 25:241. [PMID: 38582924 PMCID: PMC10998402 DOI: 10.1186/s13063-024-08072-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 03/22/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Randomised controlled trials (RCTs) aim to estimate the causal effect of one or more interventions relative to a control. One type of outcome that can be of interest in an RCT is an ordinal outcome, which is useful to answer clinical questions regarding complex and evolving patient states. The target parameter of interest for an ordinal outcome depends on the research question and the assumptions the analyst is willing to make. This review aimed to provide an overview of how ordinal outcomes have been used and analysed in RCTs. METHODS The review included RCTs with an ordinal primary or secondary outcome published between 2017 and 2022 in four highly ranked medical journals (the British Medical Journal, New England Journal of Medicine, The Lancet, and the Journal of the American Medical Association) identified through PubMed. Details regarding the study setting, design, the target parameter, and statistical methods used to analyse the ordinal outcome were extracted. RESULTS The search identified 309 studies, of which 144 were eligible for inclusion. The most used target parameter was an odds ratio, reported in 78 (54%) studies. The ordinal outcome was dichotomised for analysis in 47 ( 33 % ) studies, and the most common statistical model used to analyse the ordinal outcome on the full ordinal scale was the proportional odds model (64 [ 44 % ] studies). Notably, 86 (60%) studies did not explicitly check or describe the robustness of the assumptions for the statistical method(s) used. CONCLUSIONS The results of this review indicate that in RCTs that use an ordinal outcome, there is variation in the target parameter and the analytical approaches used, with many dichotomising the ordinal outcome. Few studies provided assurance regarding the appropriateness of the assumptions and methods used to analyse the ordinal outcome. More guidance is needed to improve the transparent reporting of the analysis of ordinal outcomes in future trials.
Collapse
Affiliation(s)
- Chris J Selman
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia.
- Department of Paediatrics, University of Melbourne, Parkville, VIC, 3052, Australia.
| | - Katherine J Lee
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Kristin N Ferguson
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Clare L Whitehead
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, 3052, Australia
- Department of Maternal Fetal Medicine, The Royal Women's Hospital, Parkville, VIC, 3052, Australia
| | - Brett J Manley
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, 3052, Australia
- Newborn Research, The Royal Women's Hospital, Parkville, VIC, 3052, Australia
- Clinical Sciences, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Robert K Mahar
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC, 3052, Australia
| |
Collapse
|
43
|
Seyedalipour F, Alipour S, Mehdinezhad H, Akrami R, Shirafkan H. Incidence of Elevated Liver Enzyme Levels in Patients Receiving Remdesivir and Its Effective Factors. Middle East J Dig Dis 2024; 16:109-113. [PMID: 39131106 PMCID: PMC11316192 DOI: 10.34172/mejdd.2024.377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 03/02/2024] [Indexed: 08/13/2024] Open
Abstract
Background Emergency use of remdesivir was approved for COVID-19 in some countries. Based on the promising results of remdesivir, the most common side effects were nausea, worsening respiratory failure, increased alanine aminotransferase levels, and constipation. The aim of this study was to determine the incidence of elevated liver enzymes in patients with COVID-19 receiving remdesivir. Methods In this retrospective study, information was collected from patients' files. The study population included patients with moderate to severe COVID-19 who were admitted to Rouhani Babol Hospital. For daily patient selection, the list of patients was extracted from the system, and based on the census, the patient file was selected. Data were analyzed using Stata 16. Results 620 patients suffering from moderate to severe COVID-19 were included in this study, 43% of whom were men. Of these patients, 120 were selected as the control group who did not receive remdesivir. The increase in liver enzymes in patients receiving remdesivir compared with the control, for alanine transaminase (ALT) and aspartate transaminase (AST), respectively, was 6.20 and 3.64 times, but it was not statistically significant for alkaline phosphatase (ALP). Also, the increase in bilirubin levels in patients receiving remdesivir was not statistically significant. Conclusion The recipients of remdesivir had high liver enzymes, which is one of the possible side effects of this drug. The intensity of the enzymes was mild and moderate, and they were not dangerous to the health of any of the consumers. Deaths in patients with COVID-19 were not due to drug-induced liver complications but to other factors such as disease-related complications.
Collapse
Affiliation(s)
- Fatere Seyedalipour
- Clinical Research Development Unit of Ayatollah Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Shabnam Alipour
- Faculty of Pharmacy, Ayatollah Amoli branch, Islamic Azad University, Amol, Iran
| | - Hamed Mehdinezhad
- Department of Internal Medicine, Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Rahim Akrami
- School of Health, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hoda Shirafkan
- Social Determinants of Health Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
44
|
Kamtalwar S, Mirgh S, More A, Sharma P, Patkar N, Rajpal S, Chatterjee G, Shetty N, Gokarn A. Recurrent Cytokine Storm in SARS-CoV-2 Infected Patients with Hematolymphoid Malignancy: A New Perspective. South Asian J Cancer 2024; 13:157-162. [PMID: 38919664 PMCID: PMC11196155 DOI: 10.1055/s-0043-1761443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024] Open
Affiliation(s)
- Sujeet Kamtalwar
- Department of General Medicine, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai and Homi Bhabha National Institute (HBNI), Maharashtra, India
| | - Sumeet Mirgh
- Department of Medical Oncology, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai and Homi Bhabha National institute (HBNI), Maharashtra, India
| | - Ashwini More
- Department of General Medicine, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai and Homi Bhabha National Institute (HBNI), Maharashtra, India
| | - Palak Sharma
- Department of Medical Administration, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai and Homi Bhabha National institute (HBNI), Maharashtra, India
| | - Nikhil Patkar
- Department of Hematopathology Laboratory, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai and Homi Bhabha National institute (HBNI), Maharashtra, India
| | - Sweta Rajpal
- Department of Hematopathology Laboratory, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai and Homi Bhabha National institute (HBNI), Maharashtra, India
| | - Gaurav Chatterjee
- Department of Hematopathology Laboratory, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai and Homi Bhabha National institute (HBNI), Maharashtra, India
| | - Nitin Shetty
- Department of Radiodiagnosis, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai and Homi Bhabha National institute (HBNI), Maharashtra, India
| | - Anant Gokarn
- Department of Medical Oncology, Advanced Centre for Treatment, Research & Education in Cancer (ACTREC), Navi Mumbai and Homi Bhabha National institute (HBNI), Maharashtra, India
| |
Collapse
|
45
|
Hedskog C, Spinner CD, Protzer U, Hoffmann D, Ko C, Gottlieb RL, Askar M, Roestenberg M, de Vries JJC, Carbo EC, Martin R, Li J, Han D, Rodriguez L, Parvangada A, Perry JK, Ferrer R, Antón A, Andrés C, Casares V, Günthard HF, Huber M, McComsey GA, Sadri N, Aberg JA, van Bakel H, Porter DP. No Remdesivir Resistance Observed in the Phase 3 Severe and Moderate COVID-19 SIMPLE Trials. Viruses 2024; 16:546. [PMID: 38675889 PMCID: PMC11053423 DOI: 10.3390/v16040546] [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: 03/04/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Remdesivir (RDV) is a broad-spectrum nucleotide analog prodrug approved for the treatment of COVID-19 in hospitalized and non-hospitalized patients with clinical benefit demonstrated in multiple Phase 3 trials. Here we present SARS-CoV-2 resistance analyses from the Phase 3 SIMPLE clinical studies evaluating RDV in hospitalized participants with severe or moderate COVID-19 disease. The severe and moderate studies enrolled participants with radiologic evidence of pneumonia and a room-air oxygen saturation of ≤94% or >94%, respectively. Virology sample collection was optional in the study protocols. Sequencing and related viral load data were obtained retrospectively from participants at a subset of study sites with local sequencing capabilities (10 of 183 sites) at timepoints with detectable viral load. Among participants with both baseline and post-baseline sequencing data treated with RDV, emergent Nsp12 substitutions were observed in 4 of 19 (21%) participants in the severe study and none of the 2 participants in the moderate study. The following 5 substitutions emerged: T76I, A526V, A554V, E665K, and C697F. The substitutions T76I, A526V, A554V, and C697F had an EC50 fold change of ≤1.5 relative to the wildtype reference using a SARS-CoV-2 subgenomic replicon system, indicating no significant change in the susceptibility to RDV. The phenotyping of E665K could not be determined due to a lack of replication. These data reveal no evidence of relevant resistance emergence and further confirm the established efficacy profile of RDV with a high resistance barrier in COVID-19 patients.
Collapse
Affiliation(s)
- Charlotte Hedskog
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (R.M.); (J.L.); (D.H.); (L.R.); (A.P.); (J.K.P.); (D.P.P.)
| | - Christoph D. Spinner
- TUM School of Medicine and Health, Department of Clinical Medicine—Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, 81675 Munich, Germany;
| | - Ulrike Protzer
- German Center for Infection Research (DZIF), Munich Partner Site, 81675 Munich, Germany; (U.P.); (D.H.)
- Institute of Virology, Technical University of Munich School of Medicine, 81675 Munich, Germany;
- Institute of Virology, Helmholtz Munich, 85764 Munich, Germany
| | - Dieter Hoffmann
- German Center for Infection Research (DZIF), Munich Partner Site, 81675 Munich, Germany; (U.P.); (D.H.)
- Institute of Virology, Technical University of Munich School of Medicine, 81675 Munich, Germany;
| | - Chunkyu Ko
- Institute of Virology, Technical University of Munich School of Medicine, 81675 Munich, Germany;
- Institute of Virology, Helmholtz Munich, 85764 Munich, Germany
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Robert L. Gottlieb
- Center for Advanced Heart and Lung Disease, Department of Internal Medicine, Baylor University Medical Center, Dallas, TX 75246, USA; (R.L.G.); (M.A.)
- Baylor Scott & White Research Institute, Dallas, TX 75246, USA
- Department of Internal Medicine, Texas A&M Health Science Center, Dallas, TX 75246, USA
- Department of Internal Medicine, Burnett School of Medicine at TCU, Fort Worth, TX 76109, USA
| | - Medhat Askar
- Center for Advanced Heart and Lung Disease, Department of Internal Medicine, Baylor University Medical Center, Dallas, TX 75246, USA; (R.L.G.); (M.A.)
- QU Health and Department of Immunology, College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar
| | - Meta Roestenberg
- Leiden University Medical Center for Infectious Diseases (LUCID), 2333 ZA Leiden, The Netherlands; (M.R.); (J.J.C.d.V.); (E.C.C.)
| | - Jutte J. C. de Vries
- Leiden University Medical Center for Infectious Diseases (LUCID), 2333 ZA Leiden, The Netherlands; (M.R.); (J.J.C.d.V.); (E.C.C.)
| | - Ellen C. Carbo
- Leiden University Medical Center for Infectious Diseases (LUCID), 2333 ZA Leiden, The Netherlands; (M.R.); (J.J.C.d.V.); (E.C.C.)
| | - Ross Martin
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (R.M.); (J.L.); (D.H.); (L.R.); (A.P.); (J.K.P.); (D.P.P.)
| | - Jiani Li
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (R.M.); (J.L.); (D.H.); (L.R.); (A.P.); (J.K.P.); (D.P.P.)
| | - Dong Han
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (R.M.); (J.L.); (D.H.); (L.R.); (A.P.); (J.K.P.); (D.P.P.)
| | - Lauren Rodriguez
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (R.M.); (J.L.); (D.H.); (L.R.); (A.P.); (J.K.P.); (D.P.P.)
| | - Aiyappa Parvangada
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (R.M.); (J.L.); (D.H.); (L.R.); (A.P.); (J.K.P.); (D.P.P.)
| | - Jason K. Perry
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (R.M.); (J.L.); (D.H.); (L.R.); (A.P.); (J.K.P.); (D.P.P.)
| | - Ricard Ferrer
- Vall d’Hebron Hospital Universitari, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Medicine Department, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (R.F.); (A.A.); (C.A.); (V.C.)
| | - Andrés Antón
- Vall d’Hebron Hospital Universitari, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Medicine Department, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (R.F.); (A.A.); (C.A.); (V.C.)
| | - Cristina Andrés
- Vall d’Hebron Hospital Universitari, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Medicine Department, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (R.F.); (A.A.); (C.A.); (V.C.)
| | - Vanessa Casares
- Vall d’Hebron Hospital Universitari, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Medicine Department, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (R.F.); (A.A.); (C.A.); (V.C.)
| | - Huldrych F. Günthard
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8057 Zurich, Switzerland;
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Michael Huber
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Grace A. McComsey
- Department of Medicine, University Hospitals of Cleveland and Case Western Reserve University, Cleveland, OH 44106, USA; (G.A.M.); (N.S.)
| | - Navid Sadri
- Department of Medicine, University Hospitals of Cleveland and Case Western Reserve University, Cleveland, OH 44106, USA; (G.A.M.); (N.S.)
| | - Judith A. Aberg
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Danielle P. Porter
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (R.M.); (J.L.); (D.H.); (L.R.); (A.P.); (J.K.P.); (D.P.P.)
| |
Collapse
|
46
|
Ramón A, Bas A, Herrero S, Blasco P, Suárez M, Mateo J. Personalized Assessment of Mortality Risk and Hospital Stay Duration in Hospitalized Patients with COVID-19 Treated with Remdesivir: A Machine Learning Approach. J Clin Med 2024; 13:1837. [PMID: 38610602 PMCID: PMC11013017 DOI: 10.3390/jcm13071837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Background: Despite advancements in vaccination, early treatments, and understanding of SARS-CoV-2, its impact remains significant worldwide. Many patients require intensive care due to severe COVID-19. Remdesivir, a key treatment option among viral RNA polymerase inhibitors, lacks comprehensive studies on factors associated with its effectiveness. Methods: We conducted a retrospective study in 2022, analyzing data from 252 hospitalized COVID-19 patients treated with remdesivir. Six machine learning algorithms were compared to predict factors influencing remdesivir's clinical benefits regarding mortality and hospital stay. Results: The extreme gradient boost (XGB) method showed the highest accuracy for both mortality (95.45%) and hospital stay (94.24%). Factors associated with worse outcomes in terms of mortality included limitations in life support, ventilatory support needs, lymphopenia, low albumin and hemoglobin levels, flu and/or coinfection, and cough. For hospital stay, factors included vaccine doses, lung density, pulmonary radiological status, comorbidities, oxygen therapy, troponin, lactate dehydrogenase levels, and asthenia. Conclusions: These findings underscore XGB's effectiveness in accurately categorizing COVID-19 patients undergoing remdesivir treatment.
Collapse
Affiliation(s)
- Antonio Ramón
- Department of Pharmacy, University General Hospital, 46014 Valencia, Spain; (A.R.); (A.B.); (S.H.); (P.B.)
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
| | - Andrés Bas
- Department of Pharmacy, University General Hospital, 46014 Valencia, Spain; (A.R.); (A.B.); (S.H.); (P.B.)
| | - Santiago Herrero
- Department of Pharmacy, University General Hospital, 46014 Valencia, Spain; (A.R.); (A.B.); (S.H.); (P.B.)
| | - Pilar Blasco
- Department of Pharmacy, University General Hospital, 46014 Valencia, Spain; (A.R.); (A.B.); (S.H.); (P.B.)
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
| | - Miguel Suárez
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
- Department of Gastroenterology, Virgen de la Luz Hospital, 16002 Cuenca, Spain
| | - Jorge Mateo
- Medical Analysis Expert Group, Institute of Technology, University of Castilla-La Mancha, 16002 Cuenca, Spain
- Medical Analysis Expert Group, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), 45071 Toledo, Spain
| |
Collapse
|
47
|
Willis ZI, Oliveira CR, Abzug MJ, Anosike BI, Ardura MI, Bio LL, Boguniewicz J, Chiotos K, Downes K, Grapentine SP, Hersh AL, Heston SM, Hijano DR, Huskins WC, James SH, Jones S, Lockowitz CR, Lloyd EC, MacBrayne C, Maron GM, Hayes McDonough M, Miller CM, Morton TH, Olivero RM, Orscheln RC, Schwenk HT, Singh P, Soma VL, Sue PK, Vora SB, Nakamura MM, Wolf J. Guidance for prevention and management of COVID-19 in children and adolescents: A consensus statement from the Pediatric Infectious Diseases Society Pediatric COVID-19 Therapies Taskforce. J Pediatric Infect Dis Soc 2024; 13:159-185. [PMID: 38339996 PMCID: PMC11494238 DOI: 10.1093/jpids/piad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 12/27/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND Since November 2019, the SARS-CoV-2 pandemic has created challenges for preventing and managing COVID-19 in children and adolescents. Most research to develop new therapeutic interventions or to repurpose existing ones has been undertaken in adults, and although most cases of infection in pediatric populations are mild, there have been many cases of critical and fatal infection. Understanding the risk factors for severe illness and the evidence for safety, efficacy, and effectiveness of therapies for COVID-19 in children is necessary to optimize therapy. METHODS A panel of experts in pediatric infectious diseases, pediatric infectious diseases pharmacology, and pediatric intensive care medicine from 21 geographically diverse North American institutions was re-convened. Through a series of teleconferences and web-based surveys and a systematic review with meta-analysis of data for risk factors, a guidance statement comprising a series of recommendations for risk stratification, treatment, and prevention of COVID-19 was developed and refined based on expert consensus. RESULTS There are identifiable clinical characteristics that enable risk stratification for patients at risk for severe COVID-19. These risk factors can be used to guide the treatment of hospitalized and non-hospitalized children and adolescents with COVID-19 and to guide preventative therapy where options remain available.
Collapse
Affiliation(s)
- Zachary I Willis
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Carlos R Oliveira
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Mark J Abzug
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, USA
| | - Brenda I Anosike
- Department of Pediatrics, The Children’s Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Monica I Ardura
- Department of Pediatrics, ID Host Defense Program, Nationwide Children’s Hospital & The Ohio State University, Columbus, OH, USA
| | - Laura L Bio
- Department of Pharmacy, Lucile Packard Children’s Hospital, Stanford, CA, USA
| | - Juri Boguniewicz
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, USA
| | - Kathleen Chiotos
- Departments of Anesthesiology, Critical Care Medicine, and Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Divisions of Critical Care Medicine and Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kevin Downes
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Steven P Grapentine
- Department of Pharmacy, University of California San Francisco Benioff Children’s Hospital, San Francisco, CA, USA
| | - Adam L Hersh
- Department of Pediatrics, Division of Infectious Diseases, University of Utah, Salt Lake City, UT, USA
| | - Sarah M Heston
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Diego R Hijano
- Department of Infectious Diseases, St. Jude Children’s Research Hospital and Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - W Charles Huskins
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Scott H James
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sarah Jones
- Department of Pharmacy, Boston Children’s Hospital, Boston, MA, USA
| | | | - Elizabeth C Lloyd
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | | - Gabriela M Maron
- Department of Infectious Diseases, St. Jude Children’s Research Hospital and Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Molly Hayes McDonough
- Center for Healthcare Quality & Analytics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Christine M Miller
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Theodore H Morton
- Department of Pharmacy, St Jude’s Children’s Research Hospital, Memphis, Tennessee, USA
| | - Rosemary M Olivero
- Department of Pediatrics and Human Development, Michigan State College of Human Medicine and Helen DeVos Children’s Hospital of Corewell Health, Grand Rapids, MI, USA
| | | | - Hayden T Schwenk
- Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA
| | - Prachi Singh
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Vijaya L Soma
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - Paul K Sue
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Surabhi B Vora
- Department of Pediatrics, University of Washington School of Medicine, and Division of Infectious Diseases, Seattle Children’s Hospital, Seattle, WA, USA
| | - Mari M Nakamura
- Antimicrobial Stewardship Program and Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
| | - Joshua Wolf
- Department of Infectious Diseases, St. Jude Children’s Research Hospital and Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
48
|
Peters J, Force L, Ng LJ, Li H, Aoki K, Taguchi N, Tanikawa T, Ishizaki A. The safety and effectiveness of remdesivir in a postmarketing surveillance study in Japan. Respir Investig 2024; 62:192-199. [PMID: 38185020 DOI: 10.1016/j.resinv.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/07/2023] [Accepted: 12/28/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND To evaluate the occurrence of adverse drug reactions (ADRs) and to assess mortality and health status in participants receiving remdesivir in real-world settings in Japan. METHODS This postmarketing surveillance study used an all-case surveillance method for enrollment. Participants with SARS-CoV-2 infection administered remdesivir from July 2020 to November 2021 in Japan were eligible for inclusion. The observation period was from remdesivir treatment initiation to 4 weeks after the end of treatment or treatment discontinuation. Clinical status and outcomes were analyzed by Kaplan-Meier plots and compared across subgroups at baseline, Day 14, Day 28, and the final observation point. RESULTS The analysis included 2128 participants (mean age, 67 years; 71.4 % male; 84.1 % with current comorbidities). ADRs and serious adverse drug reactions (SADRs) were reported among 10.4 % and 1.2 % participants, respectively. Overall, 191/2127 participants died (mortality rate [95 % confidence interval], 11.10 [9.66-12.75] per 100 person-months), 1511/2127 showed clinical improvement (117.8 [112.0-123.9] per 100 person-months), 1392/2127 recovered (103.9 [98.6-110.0] per 100 person-months), and 216/324 were extubated (107.0 [93.6-122.3] per 100 person-months). CONCLUSIONS The incidence of ADRs and SADRs was low, and no new safety concerns were identified. Observed mortality and clinical improvement results were consistent with prior studies, confirming remdesivir's benefits in real-world settings in Japan.
Collapse
Affiliation(s)
- Jami Peters
- Gilead Sciences, Inc., 333 Lakeside Dr, Foster City, CA, USA.
| | - Lindsey Force
- Gilead Sciences, Inc., 333 Lakeside Dr, Foster City, CA, USA
| | - Leslie J Ng
- Gilead Sciences, Inc., 333 Lakeside Dr, Foster City, CA, USA
| | - Hu Li
- Gilead Sciences, Inc., 333 Lakeside Dr, Foster City, CA, USA
| | - Kouji Aoki
- Gilead Sciences, K.K., 16/F GRANTOKYO SOUTHTOWER, 1-9-2, Marunouchi, Chiyoda-ku, Tokyo, 100-6616, Japan
| | - Nao Taguchi
- Gilead Sciences, K.K., 16/F GRANTOKYO SOUTHTOWER, 1-9-2, Marunouchi, Chiyoda-ku, Tokyo, 100-6616, Japan
| | - Tetsuya Tanikawa
- Gilead Sciences, K.K., 16/F GRANTOKYO SOUTHTOWER, 1-9-2, Marunouchi, Chiyoda-ku, Tokyo, 100-6616, Japan
| | - Akinobu Ishizaki
- Gilead Sciences, K.K., 16/F GRANTOKYO SOUTHTOWER, 1-9-2, Marunouchi, Chiyoda-ku, Tokyo, 100-6616, Japan
| |
Collapse
|
49
|
Ahmed A, Munoz FM, Muller WJ, Agwu A, Kimberlin DW, Galli L, Deville JG, Sue PK, Mendez-Echevarria A, Humeniuk R, Guo S, Rodriguez L, Han D, Hedskog C, Maxwell H, Palaparthy R, Kersey K, Rojo P. Remdesivir for COVID-19 in Hospitalized Children: A Phase 2/3 Study. Pediatrics 2024; 153:e2023063775. [PMID: 38332740 DOI: 10.1542/peds.2023-063775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 02/10/2024] Open
Abstract
OBJECTIVES Remdesivir decreases the risk of SARS-CoV-2 infection progressing to severe disease in adults. This study evaluated remdesivir safety and pharmacokinetics in infants and children. METHODS This was a phase 2/3, open-label trial in children aged 28 days to 17 years hospitalized for polymerase chain reaction-confirmed SARS-CoV-2 infection. Participants received for ≤10 days once-daily intravenous remdesivir doses defined using physiologically based pharmacokinetic modeling (for ≥40 kg, 200 mg day 1, then 100 mg/day; for age ≥28 days and ≥3 to <40 kg, 5 mg/kg day 1, then 2.5 mg/kg/day). Sparse pharmacokinetic samples were analyzed using population-pharmacokinetic approaches for remdesivir and metabolites GS-704277 and GS-441524. RESULTS Among 53 participants, at enrollment the median (Q1, Q3) number of days of COVID-19 symptoms was 5 (3, 7) and hospitalization was 1 (1, 3). Underlying conditions included obesity in 19 (37%), asthma in 11 (21%), and cardiac disorders in 11 (21%). Median duration of remdesivir treatment was 5 days (range, 1-10). Remdesivir treatment had no new apparent safety trends. Two participants discontinued treatment because of adverse events including elevated transaminases; both had elevated transaminases at baseline. Three deaths occurred during treatment (and 1 after). When compared with phase 3 adult data, estimated mean pediatric parameters (area under the concentration-time curve over 1 dosing interval, AUCτ, Cmax, and Cτ) were largely overlapping but modestly increased (remdesivir, 33%-129%; GS-704277, 37%-124%; GS-441524, 0%-60%). Recovery occurred for 62% of participants on day 10 and 83% at last assessment. CONCLUSIONS In infants and children with COVID-19, the doses of remdesivir evaluated provided drug exposure similar to adult dosing. In this study with a small sample size, no new safety concerns were observed.
Collapse
Affiliation(s)
- Amina Ahmed
- Department of Pediatrics, Levine Children's Hospital at Atrium Health, Charlotte, North Carolina
- Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Flor M Munoz
- Departments of Pediatrics and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
- Texas Children's Hospital, Houston, Texas
| | - William J Muller
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Allison Agwu
- Division of Infectious Diseases, Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Luisa Galli
- Department of Health Sciences, University of Florence; Pediatric Infectious Diseases Unit, Meyer Children's University Hospital, IRCCS, Florence, Italy
| | - Jaime G Deville
- Division of Infectious Diseases, Department of Pediatrics, University of California, Los Angeles, California
| | - Paul K Sue
- Division of Infectious Diseases, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ana Mendez-Echevarria
- Servicio de Pediatría, Enfermedades Infecciosas y Tropicales, Hospital Universitario La Paz, Madrid, Spain
- Centro de Investigación en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Susan Guo
- Gilead Sciences, Inc., Foster City, California
| | | | - Dong Han
- Gilead Sciences, Inc., Foster City, California
| | | | | | | | | | - Pablo Rojo
- Hospital Universitario12 de Octubre, Madrid, Spain
- Instituto de Investigación 12 de Octubre, Madrid, Spain
- Universidad Complutense, Madrid, Spain
| |
Collapse
|
50
|
Romani L, Roversi M, Bernardi S, Venturini E, Garazzino S, Donà D, Krzysztofiak A, Montagnani C, Funiciello E, Calò Carducci FI, Marabotto C, Castagnola E, Salvini F, Lancella L, Lo Vecchio A, Galli L, Castelli Gattinara G. Use of Remdesivir in children with COVID-19: report of an Italian multicenter study. Ital J Pediatr 2024; 50:32. [PMID: 38413992 PMCID: PMC10900665 DOI: 10.1186/s13052-024-01606-z] [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: 09/28/2023] [Accepted: 02/11/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND COVID-19 is generally milder in children than in adults, however severe infection has been described in some patients. Few data are available on use of Remdesivir (RDV) in children, as most clinical trials focused on adult patients. We report a multicenter study conducted in 10 Italian Hospitals to investigate the safety of RDV in children affected by COVID-19. METHODS We collected the clinical data of children with COVID-19 treated with RDV between March 2020 and February 2022 in 10 Italian hospitals. Clinical data were compared according to a duration of RDV therapy more or less than 5 days. Linear regression model was used to determine the association of significant variables from the bivariate analysis to the duration of RDV therapy. RESULTS A total of 50 patients were included, with a median age of 12.8 years. Many patients had at least one comorbidity (78%), mostly obesity. Symptoms were fever (88%), cough (74%) and dyspnea (68%). Most patients were diagnosed with pneumonia of either viral and/or bacterial etiology. Blood test showed leukopenia in 66% and increased C-reactive protein (CRP) levels in 63% of cases. Thirty-six patients received RDV for 5 days, nine patients up to 10 days. Most children who received RDV longer were admitted to the PICU (67%). Treatment with RDV was well tolerated with rare side effects: bradycardia was recorded in 6% of cases, solved in less than 24 h after discontinuation. A mild elevation of transaminases was observed in 26% of cases, however for the 8%, it was still detected before the RDV administration. Therefore, in these cases, we could not establish if it was caused by COVID-19, RDV o both. Patients who received RDV for more than 5 days waited longer for its administration after pneumonia diagnosis. The presence of comorbidities and the duration of O2 administration significantly correlated with the duration of RDV therapy at the linear regression analysis. CONCLUSION Our experience indicates that RDV against SARS-CoV-2 is safe and well-tolerated in pediatric populations at high risk of developing severe COVID-19. Our data suggest that delaying RDV therapy after diagnosis of pneumonia may be associated with a longer duration of antiviral therapy, especially in patients with comorbidities.
Collapse
Affiliation(s)
- Lorenza Romani
- Infectious Diseases Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Marco Roversi
- PhD Course "Immunology, Molecular Medicine and Applied Biotechnology", University of Rome Tor Vergata, Rome, Italy
| | - Stefania Bernardi
- Infectious Diseases Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Silvia Garazzino
- Pediatric Infectious Diseases Unit, Regina Margherita Children's Hospital, University of Turin, Turin, Italy
| | - Daniele Donà
- Division of Pediatric Infectious Diseases, Department for Woman and Child Health, University of Padua, Padua, Italy
| | | | - Carlotta Montagnani
- Infectious Diseases Unit, Meyer Children's University Hospital, Florence, Italy
| | - Elisa Funiciello
- Pediatric Infectious Diseases Unit, Regina Margherita Children's Hospital, University of Turin, Turin, Italy
| | | | - Caterina Marabotto
- Department of Pediatrics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Elio Castagnola
- Hematology and Oncology, Department of Pediatrics, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Filippo Salvini
- Pediatrics Division, Azienda Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Laura Lancella
- Infectious Diseases Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Andrea Lo Vecchio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Luisa Galli
- Infectious Diseases Unit, Meyer Children's University Hospital, Florence, Italy
| | | |
Collapse
|