1
|
Grikscheit K, Berger A, Rabenau H, Kohmer N, Appel KS, Scherer M, Bals R, Blaschke S, Hamprecht A, Hopff SM, Krefting D, Meybohm P, Nürnberger C, Heuschmann P, Pley C, Nunes de Miranda SM, Dahl E, Jensen B, Illig T, Anton G, Vehreschild JJ, Ciesek S. Occurrence and clinical correlates of SARS-CoV-2 viremia in two German patient cohorts. Emerg Microbes Infect 2025; 14:2459137. [PMID: 39868965 PMCID: PMC11812106 DOI: 10.1080/22221751.2025.2459137] [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: 10/04/2024] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
Viremia defined as detectable SARS-CoV-2 RNA in the blood is a potential marker of disease severity and prognosis in COVID-19 patients. Here, we determined the frequency of viremia in serum of two independent COVID-19 patient cohorts within the German National Pandemic Cohort Network (German: Nationales Pandemie Kohorten Netzwerk, NAPKON) with diagnostic RT-PCR against SARS-CoV-2. A cross-sectional cohort with 1122 COVID-19 patients (German: Sektorenuebergreifende Platform, SUEP) and 299 patients recruited in a high-resolution platform with patients at high risk to develop severe courses (German: Hochaufloesende Plattform, HAP) were tested for viremia. Our study also involved a comprehensive analysis and association of serological, diagnostic, and clinical parameters of the NAPKON medical dataset. Prevalence of viremia at the recruitment visit was 12.8% (SUEP) and 13% (HAP), respectively. Serological analysis revealed that viremic patients had lower levels of SARS-CoV-2 specific antibodies as well as lower neutralizing antibodies compared to aviremic patients. Viremia was associated with severity (<0.0001 SUEP; 0.002 HAP) and mortality of COVID-19 (both cohorts <0.0001) compared to aviremic patients. While rare, viremia was also detected in patients with mild disease (0.7%). In patients of the SUEP cohort with acute kidney disease (p = 0.0099) and hematooncological conditions (p = 0.0091), viremia was detected more frequently. Compared to the aviremic group, treatment with immunomodulating drugs as well as elevated levels of inflammatory markers in the blood was more frequent in the viremic group. In conclusion, our analysis revealed that detectable viremia correlates with hyperinflammatory conditions and higher risk for severe COVID-19 disease.
Collapse
Affiliation(s)
- Katharina Grikscheit
- Institute for Medical Virology, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Annemarie Berger
- Institute for Medical Virology, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Holger Rabenau
- Institute for Medical Virology, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Niko Kohmer
- Institute for Medical Virology, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Katharina S. Appel
- Faculty of Medicine, Institute for Digital Medicine and Clinical Data Science, Goethe University Frankfurt, Frankfurt am Main, Germany
- Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, University of Cologne, Cologne, Germany
| | - Margarete Scherer
- Faculty of Medicine, Institute for Digital Medicine and Clinical Data Science, Goethe University Frankfurt, Frankfurt am Main, Germany
- Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, University of Cologne, Cologne, Germany
| | - Robert Bals
- Department of Internal Medicine V - Pulmonology, Infectious Diseases, Intensive Care Medicine, Saarland University, Homburg, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University Campus, Saarbrücken, Germany
| | - Sabine Blaschke
- Emergency Department, University Medical Center Göttingen, Göttingen, Germany
| | - Axel Hamprecht
- Institute of Medical Microbiology and Virology, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Sina M. Hopff
- Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, Cologne, Germany
| | - Dagmar Krefting
- Department of Medical Informatics, University Medical Center Göttingen, Göttingen, Germany
| | - Patrick Meybohm
- Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Carolin Nürnberger
- Institute for Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
- Institute for medical Data Sciences, University Hospital Würzburg, Würzburg, Germany
| | - Peter Heuschmann
- Institute for Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
- Institute for medical Data Sciences, University Hospital Würzburg, Würzburg, Germany
- Clinical Trial Center, University Hospital Würzburg, Würzburg, Germany
| | - Caitlin Pley
- Department of Infectious Diseases and Respiratory Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Susana M. Nunes de Miranda
- Faculty of Medicine and University Hospital Cologne, Department I for Internal Medicine, University of Cologne, Cologne, Germany
| | - Edgar Dahl
- RWTH cBMB at the Institute of Pathology, Medical Faculty of RWTH Aachen University, Aachen, Germany
| | - Björn Jensen
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Düsseldorf University Hospital, Heinrich Heine University, Düsseldorf, Germany
| | - Thomas Illig
- Hannover Unified Biobank, Hannover Medical School, Hannover, Germany
| | - Gabriele Anton
- Medical School OWL, Bielefeld University, Bielefeld, Germany
| | - Jörg Janne Vehreschild
- Faculty of Medicine, Institute for Digital Medicine and Clinical Data Science, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Sandra Ciesek
- Institute for Medical Virology, Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany
- German Center for Infection Research (DZIF), Partner Site Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology, Branch Translational Medicine and Pharmacology, Frankfurt am Main, Germany
| |
Collapse
|
2
|
Aruna AS, Remesh Babu KR, Deepthi K. Autoencoder-based drug-virus association prediction with reliable negative sample selection: A case study with COVID-19. Biophys Chem 2025; 322:107434. [PMID: 40096790 DOI: 10.1016/j.bpc.2025.107434] [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: 01/07/2025] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/19/2025]
Abstract
Emergence of viruses cause unprecedented challenges and thus leading to wide-ranging consequences today. The world has faced massive disruptions like COVID-19 and continues to suffer in terms of public health and world economy. Fighting with this emergence of viruses and its reemergence plays a critical role in the health care industry. Identification of novel virus-drug associations is a vital step in drug discovery. Prediction and prioritization of novel virus-drug associations through computational approaches is an alternative and best choice considering the cost and risk of biological experiments. This study proposes a method, KR-AEVDA that relies on k-nearest neighbor based reliable negative sample selection and autoencoder based feature extraction to explore promising virus-drug associations for further experimental validation. The method analyzes complex relationships among drugs and viruses by investigating similarity and association data between drugs and viruses. It generates feature vectors from the similarity data, and reliable negative samples are extracted through an effective distance-based algorithm from the unlabeled samples in the dataset. Then high level features are extracted via an autoencoder and is fed to an ensemble classifier for inferring novel associations. Experimental results on three different datasets showed that KR-AEVDA reliably attained better performance than other state-of-the-art methods. Molecular docking is carried out between the top-predicted drugs and the crystal structure of the SARS-CoV-2's main protease to further validate the predictions. Case studies for SARS-CoV-2 illustrate the effectiveness of KR-AEVDA in identifying potential virus-drug associations.
Collapse
Affiliation(s)
- A S Aruna
- Dept. of Information Technology, Government Engineering College Palakkad, Palakkad-678633, APJ Abdul Kalam Technological University, Kerala, India; Department of Computer Science, College of Engineering Vadakara, Kozhikode 673105, Kerala, India.
| | - K R Remesh Babu
- Dept. of Information Technology, Government Engineering College Palakkad, Palakkad-678633, APJ Abdul Kalam Technological University, Kerala, India.
| | - K Deepthi
- Department of Computer Science, Central University of Kerala (Govt. of India), Kasaragod 671320, Kerala, India.
| |
Collapse
|
3
|
van Niekerk I, Panieri M, Müller T, Mapahla L, Dzanibe S, Day C, Stein DJ, Peter J. Acute serum protein biomarker profile and prevalence of persistent (>6 months) neuropsychiatric symptoms in a cohort of SARS-CoV-2 PCR positive patients in Cape Town, South Africa. Brain Behav Immun Health 2025; 46:100990. [PMID: 40386506 PMCID: PMC12084414 DOI: 10.1016/j.bbih.2025.100990] [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: 06/11/2024] [Revised: 03/03/2025] [Accepted: 04/03/2025] [Indexed: 05/20/2025] Open
Abstract
Background SARS-CoV-2 is a neurotrophic and pro-inflammatory virus, with several acute and more persistent neuropsychiatric sequelae reported. There are limited data from African cohorts and few acute illness biomarkers of persistent neuropsychiatric symptoms. Objectives To examine the association of neuropsychiatric outcomes with clinical illness severity, systemic inflammation, cardiovascular and renin-angiotensin-system (RAS) biomarkers. Second, to determine the prevalence of neuropsychiatric symptoms in a cohort of South African SARS-CoV-2 PCR positive patients at least six months following infection/hospitalization. Methodology SARS-CoV-2 PCR positive patients were recruited prospectively from Cape Town, South Africa, including hospitalized patients from ancestral, beta and delta-dominant COVID-19 waves (pre-vaccine rollout); and asymptomatic/mild SARS-CoV-2 positive patients. The 96-protein O-link inflammation and cardiovascular panels, RAS fingerprinting, and antibody responses were measured in serum samples collected at peak severity and recovery (>3 months post-infection). Telephonic interviews were conducted at least six months post infection/hospitalization. Validated measures employed were: WHO Self-Report Questionnaire (SRQ-20), Generalized Anxiety Disorder Scale (GAD-7), Chalder Fatigue Scale (CFS-11) and Telephonic Montreal Cognitive Assessment (T-MoCA). Results Ninety-seven participants completed telephonic interviews. The median (IQR) age was 48 (37-59) years, and 54 % were female. There were no significant associations between neuropsychiatric outcomes and illness severity, systemic inflammation, cardiovascular and/or renin-angiotensin-system (RAS) biomarkers from either peak illness or recovery samples. More than half of this SA COVID-19 cohort had one or more persistent neuropsychiatric symptoms >6 months post vaccine-naïve infection. On the T-MoCA, 44 % of participants showed evidence of cognitive and/or memory impairments. Conclusion The high prevalence of persistent neuropsychiatric symptoms in this African cohort supports ongoing attention to long COVID. Acute and early serum protein biomarkers were not associated with persistent neuropsychiatric outcomes post-COVD-19.
Collapse
Affiliation(s)
| | - Monica Panieri
- Faculty of Health Sciences, University of Cape Town, South Africa
| | - Talitha Müller
- Division of Allergology and Clinical Immunology, Department of Medicine, University of Cape Town, South Africa
| | - Lovemore Mapahla
- The Modelling and Simulation Hub, Africa, Department of Statistical Science, University of Cape Town, South Africa
- Division of Epidemiology and Biostatistics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Sonwabile Dzanibe
- Division of Epidemiology and Biostatistics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- Allergy and Immunology Unit, University of Cape Town Lung Institute, South Africa
- Division of Immunology, Department of Pathology, University of Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| | - Cascia Day
- Division of Allergology and Clinical Immunology, Department of Medicine, University of Cape Town, South Africa
- Allergy and Immunology Unit, University of Cape Town Lung Institute, South Africa
| | - Dan J. Stein
- Department of Psychiatry, University of Cape Town, South Africa
- SAMRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry & Neuroscience Institute, University of Cape Town, South Africa
| | - Jonny Peter
- Division of Allergology and Clinical Immunology, Department of Medicine, University of Cape Town, South Africa
- Allergy and Immunology Unit, University of Cape Town Lung Institute, South Africa
- Division of Immunology, Department of Pathology, University of Cape Town, South Africa
| |
Collapse
|
4
|
Antolí A, Gómez-Vázquez JL, Sierra-Fortuny A, Bermudez-Carre C, Framil M, Creus-Bachiller E, Viana-Errasti J, Rofes P, Rocamora-Blanch G, Hidalgo-Peña L, García-Serrano L, Rigo-Bonnin R, Feliubadaló L, Del Valle J, Calatayud L, Morandeira F, Lázaro C, Solanich X. Autoantibodies neutralizing type I interferons remain a significant risk factor for critical COVID-19 pneumonia in vaccinated patients. Clin Immunol 2025; 276:110491. [PMID: 40185298 DOI: 10.1016/j.clim.2025.110491] [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: 02/26/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Neutralizing autoantibodies against type I interferons were strongly linked to severe COVID-19 in unvaccinated patients; however, this has yet to be evaluated in vaccinated individuals. OBJECTIVE To analyze how these autoantibodies influences disease variability in vaccine breakthrough COVID-19 pneumonia patients. METHODS Clinical and laboratory data; autoantibodies blocking interferon-α2 and -ω; and humoral response to SARS-CoV-2 vaccine were collected from all vaccinated COVID-19 pneumonia patients admitted from April 2021 to December 2022 at Bellvitge University Hospital, Spain. RESULTS 458 patients developed COVID-19 pneumonia despite an appropriate antibody response to SARS-CoV-2 vaccination. Autoantibodies neutralizing interferons were significantly more prevalent in patients with critical pneumonia compared to those with milder forms (8.8 % vs. 3.6 %; p = 0.037). Having these autoantibodies was an independent predictor for critical COVID-19 pneumonia (OR 2.88 [95 %CI 1.18-6.98]). CONCLUSION Vaccination considerably reduces the severity of COVID-19; however, patients with type I interferon autoantibodies remain at increased risk of severe disease.
Collapse
Affiliation(s)
- Arnau Antolí
- Internal Medicine Department, Hospital Universitari de Bellvitge. L'Hospitalet de Llobregat, Barcelona, Spain; Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - José Luis Gómez-Vázquez
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Angels Sierra-Fortuny
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Carla Bermudez-Carre
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Mario Framil
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain; Immunology Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Edgar Creus-Bachiller
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Cancer Network (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Julen Viana-Errasti
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Paula Rofes
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Cancer Network (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Gemma Rocamora-Blanch
- Internal Medicine Department, Hospital Universitari de Bellvitge. L'Hospitalet de Llobregat, Barcelona, Spain; Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain
| | - Lara Hidalgo-Peña
- Internal Medicine Department, Hospital Universitari de Bellvitge. L'Hospitalet de Llobregat, Barcelona, Spain; Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Lydia García-Serrano
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Immunology Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Raúl Rigo-Bonnin
- Clinical Laboratory Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Lidia Feliubadaló
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Cancer Network (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Jesús Del Valle
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Cancer Network (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Laura Calatayud
- Microbiology Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Respiratory Diseases Network (CIBERES), Carlos III Health Institute, Madrid, Spain
| | - Francisco Morandeira
- Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Immunology Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Conxi Lázaro
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain; Hereditary Cancer Program, Catalan Institute of Oncology, ICO, Hospitalet de Llobregat, Barcelona, Spain; Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain; Biomedical Research Centre in Cancer Network (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Xavier Solanich
- Internal Medicine Department, Hospital Universitari de Bellvitge. L'Hospitalet de Llobregat, Barcelona, Spain; Adult Primary Immunodeficiency Unit (UFIPA), Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain; Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain; Faculty of Medicine and Health Sciences, Universitat de Barcelona, Barcelona, Spain; Biomedical Research Centre in Obesity and Nutrition Physiopathology Network (CIBEROBN), Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
5
|
Alsoubani M, Chow J. Navigating Coronavirus Disease 2019 in Immunocompromised Populations: Evolving Risk Factors, Treatment, and Outcomes. Infect Dis Clin North Am 2025; 39:309-329. [PMID: 40055107 DOI: 10.1016/j.idc.2025.02.005] [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: 05/10/2025]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has disproportionately impacted immunocompromised hosts, leading to higher morbidity and mortality. The clinical outcomes have varied based on the degree of immunosuppression, treatment availability, severe acute respiratory syndrome coronavirus 2 variants, and vaccination status. This review discusses the evolving epidemiology, clinical presentation, treatment, and prevention strategies for COVID-19 in immunocompromised populations, including patients living with human immunodeficiency virus, solid organ transplant, and hematopoietic stem cell transplant recipients.
Collapse
Affiliation(s)
- Majd Alsoubani
- Division of Geographic Medicine and Infectious Diseases, Department of Medicine, Tufts Medical Center, Boston, MA, USA; The Stuart B. Levy Center for the Integrated Management of Antimicrobial Resistance, Tufts University School of Medicine, Boston, MA, USA.
| | - Jennifer Chow
- Division of Geographic Medicine and Infectious Diseases, Department of Medicine, Tufts Medical Center, Boston, MA, USA. https://twitter.com/JennKChow
| |
Collapse
|
6
|
Shumaker AH, Bhimraj A. Pharmacologic Treatment and Management of Coronavirus Disease 2019. Infect Dis Clin North Am 2025; 39:275-291. [PMID: 40089443 DOI: 10.1016/j.idc.2025.02.004] [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: 03/17/2025]
Abstract
Widespread vaccination, hybrid immunity, and reduced pathogenicity with circulating Omicron variants have decreased the rate of severe coronavirus disease 2019 (COVID-19) outcomes in the general population. Certain patients with COVID-19 remain at high risk for severe outcomes. Clinicians must individualize treatments based on expected benefits and relative harms for patients with mild-to-moderate COVID-19. Guideline-directed therapy for severe and critical COVID-19 has remained static over the last couple of years. Data on immunomodulatory agents have improved our understanding of the management of severe and critical COVID-19, yet uncertainty remains on the role and timing of these agents in the Omicron era.
Collapse
Affiliation(s)
- Amy Hirsch Shumaker
- Department of Pharmacy, VA Northeast Ohio Healthcare System, OH, USA; School of Medicine, Case Western Reserve University, OH, USA.
| | - Adarsh Bhimraj
- Division of Infectious Diseases, Houston Methodist Hospital, 6560 Fannin Street Scurlock tower, Suite 1512, Houston, TX 77030, USA
| |
Collapse
|
7
|
Aruna AS, Babu KRR, Deepthi K. A deep drug prediction framework for viral infectious diseases using an optimizer-based ensemble of convolutional neural network: COVID-19 as a case study. Mol Divers 2025; 29:2473-2487. [PMID: 39379663 DOI: 10.1007/s11030-024-11003-7] [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: 07/28/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024]
Abstract
The SARS-CoV-2 outbreak highlights the persistent vulnerability of humanity to epidemics and emerging microbial threats, emphasizing the lack of time to develop disease-specific treatments. Therefore, it appears beneficial to utilize existing resources and therapies. Computational drug repositioning is an effective strategy that redirects authorized drugs to new therapeutic purposes. This strategy holds significant promise for newly emerging diseases, as drug discovery is a lengthy and expensive process. Through this study, we present an ensemble method based on the convolutional neural network integrated with genetic algorithm and deep forest classifier for virus-drug association prediction (CGDVDA). We generated feature vectors by combining drug chemical structure and virus genomic sequence-based similarities, and extracted prominent deep features by applying the convolutional neural network. The convoluted features are optimized using the genetic algorithm and classified using the ensemble deep forest classifier to predict novel virus-drug associations. The proposed method predicts drugs for COVID-19 and other viral diseases in the dataset. The model could achieve ROC-AUC scores of 0.9159 on fivefold cross-validation. We compared the performance of the model with state-of-the-art approaches and classifiers. The experimental results and case studies illustrate the efficacy of CGDVDA in predicting drugs against viral infectious diseases.
Collapse
Affiliation(s)
- A S Aruna
- Dept. of Information Technology, Government Engineering College Palakkad, APJ Abdul Kalam Technological University, Palakkad, Kerala, 678633, India.
- Department of Computer Science, College of Engineering Vadakara, Kozhikode, Kerala, 673105, India.
| | - K R Remesh Babu
- Dept. of Information Technology, Government Engineering College Palakkad, APJ Abdul Kalam Technological University, Palakkad, Kerala, 678633, India
| | - K Deepthi
- Department of Computer Science, Central University of Kerala (Govt. of India), Kasaragod, Kerala, 671320, India
| |
Collapse
|
8
|
Kriz SA, Heldt CL. Current experimental, statistical, and mechanistic approaches to optimizing biomolecule separations in aqueous two-phase systems. J Chromatogr A 2025; 1749:465881. [PMID: 40127604 DOI: 10.1016/j.chroma.2025.465881] [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: 12/30/2024] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 03/26/2025]
Abstract
Aqueous two-phase systems (ATPS) have been used to purify a range of biomolecules, including small molecules, monoclonal antibodies, viruses, and whole cells. They are known for selective separations, creating a stabilizing, low-shear environment, and high yields. Recently, as biomanufacturing attempts to adopt continuous processing, attention has shifted to ATPS for its ability to operate fully continuously while incurring lower costs than many chromatographic methods. But despite 60 years of exploration and development, the complex network of interlinked driving forces controlling these separations has prevented robust development of process understanding, and most ATPS separations are still optimized using slow and costly manual screening methods. As a result, industry has been unwilling to adopt ATPS. Fortunately, a growing body of literature is developing statistical and mechanistic models of ATPS to predict liquid-liquid equilibria and separations with reduced experimental burden. This review surveys the application of these models to ATPS, comparing their progress and potential to promote rapid development of bioseparations in the near and long term. The discussion evaluates the adaptability of statistical tools, like response surface methodology and artificial neural networks, and contrasts it with the process understanding generated through application of semi-empirical thermodynamic models. Strategies are explored to automate optimization of separations for new biomolecules using these models to create artificial data. By understanding the landscape of models applied to ATPS, this review will start a discussion about bringing this technology closer to commercialization and enabling continuous processing on a broader scale.
Collapse
Affiliation(s)
- Seth A Kriz
- Department of Chemical Engineering, Michigan Technological University, Houghton MI 49931, USA
| | - Caryn L Heldt
- Department of Chemical Engineering, Michigan Technological University, Houghton MI 49931, USA.
| |
Collapse
|
9
|
Sharma T, Mondal T, Saralamma VVG, Hassan MI, Kim CJ, Churqui MP, Nyström K, Thombare K, Baig MH, Dong JJ. Exploring the exportin-1 inhibitors for COVID-19 and anticancer treatment. J Biomol Struct Dyn 2025:1-12. [PMID: 40395159 DOI: 10.1080/07391102.2025.2503981] [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: 01/28/2024] [Accepted: 06/27/2024] [Indexed: 05/22/2025]
Abstract
Nuclear export protein 1, also known as XPO1, plays a crucial role in cellular homeostasis and assists in the nucleocytoplasmic transfer of ribonucleic acids (RNAs) and proteins. In addition, this nuclear export receptor is essential for the export of a variety of cargo molecules, such as proteins implicated in the immune response, tumor suppression, and cell cycle regulation. XPO1 has emerged as a promising target to disrupt the life cycles of multiple viruses and treat cancers. In our current work, we used a computational approach consisting of pharmacophore-assisted virtual screening complemented by molecular docking, molecular dynamics, and solvation-based free-energy studies to identify new inhibitors of the XPO1 protein. The identified compounds displayed highly stable RMSD plots, hydrogen bonding interactions, and relatively good binding affinities in both docking and free energy studies. These molecules were validated in vitro against SARS-CoV-2 and cancer cell lines. The study identified novel inhibitors of the XPO1 protein with both antiviral and anticancer activities.
Collapse
Affiliation(s)
- Tanuj Sharma
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tanmoy Mondal
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Chemistry, Gothenburg, Sweden
| | | | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | | | - Marianela Patzi Churqui
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina Nyström
- Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ketan Thombare
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Chemistry, Gothenburg, Sweden
| | - Mohammad Hassan Baig
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-June Dong
- Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Negi V, Kuhn RJ. A BSL-2 chimeric system designed to screen SARS-CoV-2 E protein ion channel inhibitors. J Virol 2025; 99:e0225224. [PMID: 40304492 DOI: 10.1128/jvi.02252-24] [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: 12/18/2024] [Accepted: 03/12/2025] [Indexed: 05/02/2025] Open
Abstract
A major hindrance to the identification of new drug targets and the large-scale testing of new or existing compound libraries against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is that research on the virus is restricted to biosafety level 3 (BSL-3) laboratories. In such cases, BSL-2 surrogate systems or chimeric and attenuated versions of the virus are developed for safer, faster, and cheaper examination of the stages of the virus life cycle and specific drug targets. In this study, we describe a BSL-2 chimeric viral system utilizing a Sindbis virus background as a tool to study one such target, the SARS-CoV-2 Envelope (E) protein channel activity. This protein is fully conserved between SARS-CoV and SARS-CoV-2 variants of concern (VOCs), except for a threonine to isoleucine mutation in the Omicron variant, making the E ion channel domain an attractive antiviral target for combination therapy. Using a BSL-2-chimeric system, we have been able to show similar inhibition profiles using channel inhibitors as previously reported for E-channel inhibition in authentic SARS-CoV-2. This system has the potential to allow faster initial screening of E-channel inhibitors and can be useful in developing broad-spectrum antivirals against viral channel proteins.IMPORTANCEDespite its importance in viral infections, no antivirals exist against the ion channel activity of the SARS-CoV-2 Envelope (E) protein. The E protein is highly conserved among SARS-CoV-2 variants, making it an attractive target for antiviral therapies. Research on SARS-CoV-2 is restricted to BSL-3 laboratories, creating a bottleneck for screening potential antiviral compounds. This study presents a BSL-2 chimeric system using a Sindbis virus background to study the ion channel activity of the E protein. This novel BSL-2 system bypasses this limitation, offering a safer and faster approach for the initial screening of ion channel inhibitors. By replicating the channel inhibition profiles of authentic SARS-CoV-2 in a more accessible system, this research paves the way for the development of broad-spectrum antivirals against viral channel proteins, potentially expediting the discovery of life-saving treatments for COVID-19 and other viral diseases.
Collapse
Affiliation(s)
- Vashi Negi
- Department of Biological Sicences, Purdue University, West Lafayette, Indiana, USA
| | - Richard J Kuhn
- Department of Biological Sicences, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
11
|
Isha S, Raavi L, Jonna S, Nataraja H, Craver EC, Jenkins A, Hanson AJ, Balasubramanian P, Balavenkataraman A, Tekin A, Bansal V, Reddy S, Caples SM, Khan SA, Jain NK, LaNou AT, Kashyap R, Cartin-Ceba R, Milian RD, Venegas CP, Shapiro AB, Bhattacharyya A, Chaudhary S, Kiley SP, Quinones QJ, Patel NM, Guru PK, Franco PM, Roy A, Sanghavi DK. Role of Procalcitonin as a Prognostic Biomarker in Hospitalized COVID-19 Patients: A Comparative Analysis. Biomark Insights 2025; 20:11772719241296624. [PMID: 40386243 PMCID: PMC12084704 DOI: 10.1177/11772719241296624] [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: 07/11/2024] [Accepted: 10/09/2024] [Indexed: 05/20/2025] Open
Abstract
Background Procalcitonin (PCT) is recognized as an inflammatory biomarker, often elevated in COVID-19 pneumonia alongside other biomarkers. Understanding its association with severe outcomes and comparing its predictive ability with other biomarkers is crucial for clinical management. Objectives This retrospective multicenter observational study aimed to investigate the association between PCT levels and adverse outcomes in hospitalized COVID-19 patients. Additionally, it sought to compare the predictive performance of various biomarkers. Design The study analyzed data from the Society of Critical Care Medicine (SCCM) Viral Infection and Respiratory Illness Universal Study (VIRUS) registry, comprising COVID-19 patients hospitalized across multiple Mayo Clinic sites between March 2020 and June 2022. Methods A total of 7851 adult COVID-19 patients were included. Patients were categorized into 6 groups based on the worst WHO ordinal scale. Multivariate models were constructed using peak biomarker levels within 72 hours of admission, adjusted for confounders. Results Elevated PCT levels were independently associated with increased odds of adverse outcomes, including ICU admission (adjusted odds ratio [aOR] 1.32, 95%CI 1.27-1.38), IMV requirement (aOR 1.35, 95%CI: 1.28-1.42), and in-hospital mortality (aOR 1.30, 95%CI: 1.22-1.37). A 3.48-fold increase in IMV requirement and 3.55 times increase in in-hospital mortality were noted with peak PCT ⩾ 0.25 ng/ml. Similar associations were observed with other biomarkers like NLR (AUC 0.730), CRP, IL-6, LDH (AUC 0.800), and D-dimer (AUC 0.719). Models incorporating NLR, LDH, D-dimer, and PCT demonstrated the highest predictive accuracy, with a combined model exhibiting an area under the curve (AUC) of 0.826 (95%CI 0.803-0.849). Conclusions Higher PCT levels were significantly linked to worse outcomes in COVID-19 patients, emphasizing its potential as a prognostic marker. Biomarker-based predictive models, particularly those including PCT, showed promising utility for risk assessment and clinical decision-making. Further prospective studies are warranted to validate these findings on a larger scale.
Collapse
Affiliation(s)
- Shahin Isha
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Lekhya Raavi
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Sadhana Jonna
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Hrishikesh Nataraja
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Emily C Craver
- Department of Quantitative Health Sciences, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Anna Jenkins
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Abby J Hanson
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | | | | | - Aysun Tekin
- Department of Critical Care Medicine, Mayo Clinic Rochester, MN, USA
| | - Vikas Bansal
- Department of Critical Care Medicine, Mayo Clinic Rochester, MN, USA
| | - Swetha Reddy
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Sean M Caples
- Division of Pulmonary and Critical Care, Mayo Clinic Rochester, MN, USA
| | - Syed Anjum Khan
- Department of Critical Care Medicine, Mayo Clinic Health System in Mankato, Mankato, MN, USA
| | - Nitesh K Jain
- Department of Critical Care Medicine, Mayo Clinic Health System in Mankato, Mankato, MN, USA
| | - Abigail T LaNou
- Emergency Medicine and Critical Care, Mayo Clinic Health System, Eau Claire, WI, USA
| | - Rahul Kashyap
- Department of Anesthesia and Critical Care Medicine, Mayo Clinic Rochester, MN, USA
| | | | - Ricardo Diaz Milian
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Carla P Venegas
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Anna B Shapiro
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | | | - Sanjay Chaudhary
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Sean P Kiley
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Quintin J Quinones
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Neal M Patel
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Pramod K Guru
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Pablo Moreno Franco
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| | - Archana Roy
- Division of Hospital Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Devang K Sanghavi
- Department of Critical Care Medicine, Mayo Clinic in Florida, Jacksonville, FL, USA
| |
Collapse
|
12
|
Sweeney DA, Kalil AC. Guidelines without borders: the case for JAK inhibitors as the first-line immunomodulator COVID-19 treatment. THE LANCET. RESPIRATORY MEDICINE 2025:S2213-2600(25)00081-5. [PMID: 40378862 DOI: 10.1016/s2213-2600(25)00081-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 02/27/2025] [Indexed: 05/19/2025]
Affiliation(s)
- Daniel A Sweeney
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, CA, USA
| | - Andre C Kalil
- Division of Infectious Diseases, Department of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
13
|
Alves MCS, da Silva RCC, de Leitão-Júnior SSP, de Balbino VQ. Therapeutic Approaches for COVID-19: A Review of Antiviral Treatments, Immunotherapies, and Emerging Interventions. Adv Ther 2025:10.1007/s12325-025-03218-3. [PMID: 40338485 DOI: 10.1007/s12325-025-03218-3] [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: 03/19/2025] [Accepted: 04/22/2025] [Indexed: 05/09/2025]
Abstract
The coronavirus disease 2019 (COVID-19) global health crisis, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has presented unprecedented challenges to global healthcare systems, leading to rapid advances in treatment development. This review comprehensively examines the current therapeutic approaches for managing COVID-19, including direct-acting antivirals, immunomodulators, anticoagulants, and adjuvant therapies, as well as emerging and experimental approaches. Direct-acting antivirals target various stages of the viral life cycle, offering specific intervention points, while immunomodulators aim to modulate the host's immune response, reducing disease severity. Anticoagulant therapies address the coagulopathy frequently observed in severe cases, and adjuvant treatments provide supportive care to improve overall outcomes. We also explore the challenges and limitations of implementing these treatments, such as drug resistance, variable patient responses, and access to therapies, especially in resource-limited settings. The review also discusses future perspectives, including the potential of next-generation vaccines, personalized medicine, and global collaboration in shaping future COVID-19 treatment paradigms. Continuous innovation, combined with an integrated and adaptable approach, will be crucial to effectively managing COVID-19 and mitigating the impact of future pandemics.
Collapse
Affiliation(s)
- Maria C S Alves
- Laboratory of Bioinformatics and Evolutionary Biology, Center for Biosciences, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, 50670-423, Brazil.
| | - Ruana C C da Silva
- Laboratory of Health Sciences Research, Federal University of Grande Dourados, Dourados, Mato Grosso do Sul, 79825-070, Brazil
| | - Sérgio S P de Leitão-Júnior
- Laboratory of Bioinformatics and Evolutionary Biology, Center for Biosciences, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, 50670-423, Brazil
- Serra Talhada Academic Unit, Federal Rural University of Pernambuco, Serra Talhada, Pernambuco, 56909-535, Brazil
| | - Valdir Q de Balbino
- Laboratory of Bioinformatics and Evolutionary Biology, Center for Biosciences, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, 50670-423, Brazil.
| |
Collapse
|
14
|
Mayerhöfer T, Joannidis M, Klein S, Franke A, Margarita S, Ronzoni L, Pertler E, Wagner S, Sahanic S, Tancevski I, Haschka D, Hochhold C, Treml B, Valenti L, Tilg H, Schaefer B, Zoller H. The common genetic variant rs1278960 determining expression of Interferon-lambda predicts inflammatory response in critically ill COVID-19 patients. Sci Rep 2025; 15:15802. [PMID: 40328868 PMCID: PMC12056047 DOI: 10.1038/s41598-025-91628-2] [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: 09/25/2024] [Accepted: 02/21/2025] [Indexed: 05/08/2025] Open
Abstract
The single nucleotide polymorphism rs12979860 is associated with the production of IFNλ4, a type III interferon, which offers protection from viral infection via its proinflammatory properties. We investigated if a genetically determined increase in IFNλ4 affects disease progression in SARS-CoV-2. This prospective, single-center study involved critically ill SARS-CoV-2 patients admitted to the intensive care unit. We performed genotyping for rs12979860 and analyzed daily laboratory data. Genotype frequencies were compared with an external validation cohort. Critically ill individuals with COVID-19 (n = 184; 29.3% women) were included. Median age was 63 years. The TT genotype was present in 11%, CT in 48% and CC in 41%. At baseline, CRP, ferritin, transferrin and neopterin did not differ significantly between groups. Longitudinal analysis revealed significant genotype-dependent differences in CRP, ferritin and neopterin with the highest peak in TT patients after 10-15 days. A higher need for renal replacement therapy (31.6% vs. 11.7%, p = 0.044) and mechanical ventilation (22 days vs. 15 days, p = 0.018) was observed in the TT group. The SNP rs12979860 near IFNL4 is associated with distinct inflammatory trajectories in critically ill COVID-19 patients. Genetic determinants of the immune response influence the severity of inflammation and clinical outcomes in severe COVID-19.
Collapse
Affiliation(s)
- Timo Mayerhöfer
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Michael Joannidis
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Sebastian Klein
- Division of Intensive Care and Emergency Medicine, Department of Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
- Karl Landsteiner University of Health Sciences, Krems, Austria
- Department of Internal Medicine 2, University Hospital St. Pölten, St. Pölten, Austria
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Sara Margarita
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luisa Ronzoni
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Elke Pertler
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
- Christian Doppler Laboratory for Iron and Phosphate Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sonja Wagner
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
- Christian Doppler Laboratory for Iron and Phosphate Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sabina Sahanic
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - David Haschka
- Department of Internal Medicine II, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Hochhold
- Department of Anesthesiology and Intensive Care Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Benedikt Treml
- Department of Anesthesiology and Intensive Care Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Luca Valenti
- Precision Medicine Lab, Biological Resource Center - Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Herbert Tilg
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Benedikt Schaefer
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Heinz Zoller
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
- Christian Doppler Laboratory for Iron and Phosphate Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
15
|
Bittle E, Arnold S, Hijano DR, Landman BM, Morton T, Hines M. Safety Data for Baricitinib Use in Children With Severe SARS-CoV-2 Infection. Hosp Pediatr 2025; 15:e203-e208. [PMID: 40204287 DOI: 10.1542/hpeds.2024-008245] [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: 11/15/2024] [Accepted: 02/04/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND In November 2020, the Food and Drug Administration issued an Emergency Use Authorization for baricitinib, a Janus Kinase protein inhibitor, for hospitalized children and adults with COVID-19 requiring supplemental oxygen, but safety data for this pediatric indication is lacking. METHODS This retrospective case series describes patients aged younger than 21 years treated with baricitinib for severe COVID-19 between 2021 and 2022. Patient characteristics, treatments, adverse events, reasons for early discontinuation of baricitinib, durations of oxygen supplementation and hospitalization, and complications were recorded. RESULTS There were 37 patients who received baricitinib. Median age was 16 years (range 1-20). All had a comorbidity (59% obesity, 5% malignancy), and 76% were cared for in the intensive care unit. All received remdesivir (median 5 days; range: 2-11), and 34 (92%) received corticosteroids (median 6 days; range: 1-10). Median duration of baricitinib was 6 days (range 3-14). Baricitinib was discontinued early for clinical improvement (2), and adverse events (7; 6 elevated liver enzymes [only 1 meeting discontinuation criteria), 1 thrombocytosis]). One patient had deep vein thrombosis without pulmonary embolism, 5 patients had concurrent infections (4 bacterial, 1 fungal, 2 herpes simplex virus reactivation). All adverse events were resolved. There were no deaths. Median hospitalization was 7 days (range 2-108) and mechanical ventilation was 4.5 days (range 1-86 days). Two patients (5%) were discharged with supplemental oxygen and one with a tracheostomy. CONCLUSIONS Baricitinib appears safe in children hospitalized for severe COVID-19. Most early baricitinib discontinuation for abnormal laboratory studies was secondary to provider caution.
Collapse
Affiliation(s)
- Elspeth Bittle
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee
- Department of Pediatrics, Division of Infectious Diseases, Le Bonheur Children's Hospital, University of TN Health Science Center, Memphis, Tennessee
| | - Sandra Arnold
- Department of Pediatrics, Division of Infectious Diseases, Le Bonheur Children's Hospital, University of TN Health Science Center, Memphis, Tennessee
| | - Diego R Hijano
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee
- Department of Pediatrics, Division of Infectious Diseases, Le Bonheur Children's Hospital, University of TN Health Science Center, Memphis, Tennessee
| | | | - Ted Morton
- Department of Pharmacy and Pharmaceutical Services, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Melissa Hines
- Department of Pediatric Medicine, Division of Critical Care, St. Jude Children's Research Hospital, Memphis, Tennessee
- Department of Pediatrics, Division of Critical Care, Le Bonheur Children's Hospital, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
16
|
Monteiro AHA, Freitas KM, Montuori-Andrade ACM, de Lima EBS, Carvalho AFS, Cardoso C, Lara ES, Oliveira LC, Zaidan I, da Santos FRS, Resende F, Souza-Costa LP, Queiroz-Junior CM, Chaves IDM, Nóbrega NRC, Rabelo MBO, Rocha MP, Campana PRV, Pádua RM, Ferreira RS, Barreto LV, Kronenberger T, Maltarollo VG, de Godoy MO, Oliva G, Guido RVC, Teixeira MM, Costa VV, Sousa LP, Braga FC. Ouratein D, a Biflavanone From Ouratea spectabilis, Alleviates Betacoronavirus Infection by Mitigating Inflammation, Lung Damage and Viral Replication. Phytother Res 2025; 39:2180-2196. [PMID: 40099709 DOI: 10.1002/ptr.8462] [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: 08/19/2024] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 03/20/2025]
Abstract
Severe coronavirus outbreaks, including SARS, MERS, and COVID-19, have underscored the urgent need for effective antiviral therapies. This study evaluated the antiviral activity of biflavanones isolated from Ouratea spectabilis-specifically ouratein (Our-) A, B, C, and D-against murine hepatitis virus (MHV-3) and human SARS-CoV-2. Cells infected with MHV-3 or SARS-CoV-2 were treated with ourateins, and viral replication was assessed using plaque assays. Mice infected with MHV-3 were treated with Our-D either orally or intraperitoneally. Key assessments included leukocyte counts, cytokine and chemokine levels, histological analysis, and survival rates. The mechanism of action was explored through in silico and in vitro studies focused on the binding and inhibition of the main protease (Mpro). Our-D significantly inhibited the replication of both viruses, with a selective index of 2.5 for MHV-3 and 14.9 for SARS-CoV-2. In vivo, Our-D reduced leukocyte infiltration in the lungs, decreased CCL2 levels, increased IL-10, and lowered plasma IL-6 and CXCL1 levels. Additionally, Our-D mitigated lung damage, partially restored betacoronavirus-induced lymphopenia, and reduced viral loads in the lungs, heart, and spleen, ultimately improving survival in mice. In silico studies revealed that Our-A and Our-C had strong binding affinity for Mpro, and both significantly inhibited Mpro activity in vitro, unlike Our-D. Our-D protected mice from coronavirus infection by modulating the inflammatory response and reducing viral loads, with minimal effect on Mpro inhibition, suggesting alternative mechanisms for its antiviral activity.
Collapse
Affiliation(s)
- Adelson Héric A Monteiro
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kátia M Freitas
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Clara M Montuori-Andrade
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Erick Bryan Sousa de Lima
- Department of Clinical Analysis and Toxicology, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antônio Felipe S Carvalho
- Department of Clinical Analysis and Toxicology, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila Cardoso
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Edvaldo S Lara
- Department of Clinical Analysis and Toxicology, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo Camilo Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabella Zaidan
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Felipe Rocha Silva da Santos
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Filipe Resende
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiz Pedro Souza-Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ian de Meira Chaves
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Natália R C Nóbrega
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Beatriz O Rabelo
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marina P Rocha
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Priscilla R V Campana
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo M Pádua
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rafaela S Ferreira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiza V Barreto
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thales Kronenberger
- Partner-Site Tübingen, German Center for Infection Research (DZIF), Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vinícius G Maltarollo
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Glaucius Oliva
- Institute of Physics, Universidade de São Paulo, São Carlos, Brazil
| | - Rafael V C Guido
- Institute of Physics, Universidade de São Paulo, São Carlos, Brazil
| | - Mauro M Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian V Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Department of Clinical Analysis and Toxicology, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernão C Braga
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
17
|
Yoshida M, Taguchi N, Piao Y, Gupta R, Berry M, Peters J, Abdelghany M, Chiang M, Wang CY, Yotsuyanagi H. Treatment pattern and clinical outcomes of remdesivir in hospitalized COVID-19 patients with severe chronic kidney disease: a database analysis of acute care hospitals in Japan. Clin Exp Nephrol 2025; 29:624-637. [PMID: 39739156 PMCID: PMC12049339 DOI: 10.1007/s10157-024-02609-0] [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: 06/10/2024] [Accepted: 12/02/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND There is limited evidence on clinical outcomes and treatment pattern in Japanese patients with severe chronic kidney disease (CKD), hospitalized for coronavirus disease-2019 (COVID-19). We aimed to describe patient characteristics, treatment pattern, and clinical outcomes in Japanese patients with severe CKD, hospitalized for COVID-19 who received remdesivir (RDV). METHODS We used the anonymized claims database from Medical Data Vision Co., Ltd., Japan. The analysis included patients aged ≥ 18 years with severe CKD, hospitalized for moderate to severe COVID-19, and administered ≥ 1 dose of RDV between October 2021 and September 2023. All-cause inpatient mortality, disease progression, and recovery up to 56 days from hospitalization were evaluated. RESULTS Data of 847 patients were analyzed (mean age 73.0 ± 14.1 years). Median (Q1-Q3) time to RDV initiation was 1.0 day (1.0-2.0) from hospitalization and treatment duration was 5.0 days (3.0-5.0). At RDV initiation, 44.27% patients required non-invasive positive pressure ventilation/high or low flow oxygen; 4.25% required invasive mechanical ventilation/extracorporeal membrane oxygenation/intensive care unit hospitalization. Proportion of patients with all-cause mortality was 11.45% (stage 4, 14.89%; stage 5, 10.47%) by 28 days and 12.28% (stage 4, 16.49%; stage 5, 11.08%) by 56 days. At 28 days, 12.28% had disease progression and 72.14% recovered. CONCLUSION Most patients with severe CKD received RDV immediately after hospitalization. The majority of patients recovered by 28 days. The study provided insights into RDV treatment in inpatient settings, which could contribute to the discussion on standard of care in this population in Japan.
Collapse
Affiliation(s)
- Manami Yoshida
- Gilead Sciences, K.K., 16/F GRAN TOKYO SOUTH TOWER, 1-9-2, Marunouchi, Chiyoda-ku, Tokyo, 100-6616, Japan.
| | - Nao Taguchi
- Gilead Sciences, K.K., 16/F GRAN TOKYO SOUTH TOWER, 1-9-2, Marunouchi, Chiyoda-ku, Tokyo, 100-6616, Japan
| | - Yi Piao
- Gilead Sciences, K.K., 16/F GRAN TOKYO SOUTH TOWER, 1-9-2, Marunouchi, Chiyoda-ku, Tokyo, 100-6616, Japan
| | - Rikisha Gupta
- Gilead Sciences, Inc, 333 Lakeside Dr, Foster City, CA, USA
| | - Mark Berry
- Gilead Sciences, Inc, 333 Lakeside Dr, Foster City, CA, USA
| | - Jami Peters
- Gilead Sciences, Inc, 333 Lakeside Dr, Foster City, CA, USA
| | | | - Mel Chiang
- Gilead Sciences, Inc, 333 Lakeside Dr, Foster City, CA, USA
| | - Chen-Yu Wang
- Gilead Sciences, Inc, 333 Lakeside Dr, Foster City, CA, USA
| | | |
Collapse
|
18
|
Moreni G, Calitz C, Koen G, van Eijk H, Johannesson N, De Ruijter J, Benschop KSM, Cremer J, Pajkrt D, Sridhar A, Peters EJ, Wolthers KC. Toward Personalized Medicine: The Effect of Treatment of Chronic Enterovirus Diarrhea in an Immunocompromised Patient and the Correlation With In Vitro Models. Open Forum Infect Dis 2025; 12:ofaf212. [PMID: 40302715 PMCID: PMC12039486 DOI: 10.1093/ofid/ofaf212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/08/2025] [Indexed: 05/02/2025] Open
Abstract
Enteroviruses (EV) usually cause acute, mild, self-limiting disease. Chronic infections with EVs are rare, and typically occur in patients with immunodeficiency, posing a high risk of severe outcomes. We report a rare case of chronic diarrhea caused by coxsackievirus A1 (CVA1) (from EV-C species) infection in a patient with a common variable immunodeficiency, who was on treatment with pooled intravenous immunoglobulin (IVIG) from the Netherlands. To explore treatment options, we assessed the presence of neutralizing antibodies (nAbs) against CVA1 in pooled IVIG from South Africa, where EV-Cs are prevalent, and tested the antiviral efficacy of US Food and Drug Administration-approved drugs like fluoxetine, itraconazole, ribavirin, and remdesivir (RDV) against CVA1 in vitro. Both Dutch and South African IVIG showed low nAb titers against CVA1. The patient, treated with Dutch IVIG, also received a combination of amantadine and fluoxetine, which were discontinued due to side effects. Among the drugs tested, only RDV significantly inhibited CVA1 replication in rhabdomyosarcoma (RD) cells. This in vitro efficacy was not reflected by a favorable clinical response after treatment of the patient with RDV. In concordance with unfavorable antiviral response in the patient, preliminary tests on a co-culture model containing isogenic human intestinal cells and intestinal fibroblasts showed no significant reduction in CVA1 RNA copies after RDV administration. In conclusion, our results showed that repurposing of drugs that have shown in vitro efficacy does not translate well to the patients, and this is also reflected in a more physiologically relevant model of the human intestine.
Collapse
Affiliation(s)
- Giulia Moreni
- Department of Medical Microbiology, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Department of Pediatric Infectious Diseases, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Carlemi Calitz
- Department of Medical Microbiology, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Department of Pediatric Infectious Diseases, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Gerrit Koen
- Department of Medical Microbiology, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Hetty van Eijk
- Department of Medical Microbiology, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Nina Johannesson
- Department of Medical Microbiology, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Department of Pediatric Infectious Diseases, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Jamy De Ruijter
- Department of Anesthesiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Jeroen Cremer
- National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Dasja Pajkrt
- Department of Pediatric Infectious Diseases, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Emma Children’s Hospital, Amsterdam, The Netherlands
| | - Adithya Sridhar
- Department of Medical Microbiology, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
- Department of Pediatric Infectious Diseases, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Emma Children’s Hospital, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Amsterdam, The Netherlands
| | - Edgar J Peters
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Infectious Diseases, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Katja C Wolthers
- Department of Medical Microbiology, OrganoVIR Labs, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Qian B, Luo R, Shen B, Fan L, Zhang J, Zhang S, Sun Y, Deng X, Pang X, Zhong W, Gao Y. EIDD-2801 resists to infection and co-infection of SARS-CoV-2 and influenza virus. Virol J 2025; 22:126. [PMID: 40296172 PMCID: PMC12039283 DOI: 10.1186/s12985-025-02755-1] [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: 02/19/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has exerted a catastrophic impact on public health. Meanwhile, the seasonal influenza outbreak overlaps with the current pandemic wave. There is still an urgent need to develop effective therapeutic agents for the treatment of co-infection of multiple respiratory viruses. This study aimed to investigate antiviral effects of EIDD-2801, an orally bioavailable ribonucleoside analog, and its potent therapeutic effects in co-infection of multiple respiratory viruses. METHODS BALB/c mice and hamsters were infected with IFV or SARS-CoV-2, then were dosed orally with EIDD-2801 to measure the antiviral effects of EIDD-2801. Viral replication and mRNA transcription were evaluated by quantitative polymerase chain reaction (qPCR) and protein expression by Western Blot. Influenza viral titer was assessed using EID50 assay. RESULTS EIDD-2801 was found to be significantly effective against influenza A virus and influenza B virus. The antiviral activity against SARS-CoV-2 and further co-infection with influenza virus was also distinct. EIDD-2801 had potent antiviral effects against multiple respiratory viruses both in vitro and in vivo. CONCLUSION This study demonstrated that the small-molecule compound EIDD-2801, an orally available broad-spectrum antiviral agent, significantly inhibited the infection of influenza virus and SARS-CoV-2 and effectively protected animals from lethal influenza virus co-infection.
Collapse
Affiliation(s)
- Bingshuo Qian
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Rongbo Luo
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Beilei Shen
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Lingjun Fan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Junkui Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Shijun Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Yan Sun
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Xiuwen Deng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Xiaobin Pang
- School of Pharmacy, Henan University, Kaifeng, 475004, China.
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Yuwei Gao
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China.
| |
Collapse
|
20
|
Zhang J, Wang D, Kwok C, Xu L, Famulok M. Aptamer-engaged nanotherapeutics against SARS-CoV-2. DISCOVER NANO 2025; 20:71. [PMID: 40289185 PMCID: PMC12034613 DOI: 10.1186/s11671-025-04245-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/26/2025] [Indexed: 04/30/2025]
Abstract
The COVID-19 pandemic, caused by the virus SARS-CoV-2 infection, has underscored the critical importance of rapid and accurate therapeutics. The neutralization of SARS-CoV-2 is paramount in controlling the spread and impact of COVID-19. In this context, the integration of aptamers and aptamer-related nanotherapeutics presents a valuable and scientifically significant approach. Despite the potential, current reviews in this area are often not comprehensive and specific enough to encapsulate the full scope of therapeutic principles, strategies, advancements, and challenges. This review aims to fill that gap by providing an in-depth examination of the role of aptamers and their related molecular medicine in COVID-19 therapeutics. We first introduce the unique properties, selection, and recognition mechanism of aptamers to bind with high affinity to various targets. Next, we delve into the therapeutic potential of aptamers, focusing on their ability to inhibit viral entry and replication, as well as modulate the host immune response. The integration of aptamers with nucleic acid nanomedicine is explored. Finally, we address the challenges and future perspectives of aptamer and nucleic acid nanomedicine in COVID-19 therapeutics, including issues of stability, delivery, and manufacturing scalability. We conclude by underscoring the importance of continued research and development in this field to meet the ongoing challenges posed by COVID-19 and potential future pandemics. Our review will be a valuable resource for researchers and clinicians interested in the latest developments at the intersection of molecular biology, nanotechnology, and infectious disease management.
Collapse
Affiliation(s)
- Jing Zhang
- Life Science and Chemistry College, Hunan University of Technology, Zhuzhou, 412007, China
| | - Dan Wang
- Life & Medical Sciences Institute (LIMES), Pharmaceutical Institute, Universität Bonn, 53121, Bonn, Germany.
| | - Chiu Kwok
- Life & Medical Sciences Institute (LIMES), Pharmaceutical Institute, Universität Bonn, 53121, Bonn, Germany
| | - Liujun Xu
- Department of Respiratory and Critical Care, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, 324000, China.
| | - Michalina Famulok
- Life & Medical Sciences Institute (LIMES), Pharmaceutical Institute, Universität Bonn, 53121, Bonn, Germany
| |
Collapse
|
21
|
Mallinson PAC, Joshi M, Mathpathi M, Perkins A, Clayton T, Shah AS, Mathur R, Birk N, Dhillon A, Lieber J, Beg SS, Hopkins L, Khan A, Allaham S, Kam VT, Sutaria S, R G, Rajagopala S, Bhamra A, Pillai GKG, Khunti K, Nesari T, Kinra S. Ashwagandha ( Withania somnifera (L.) Dunal) for promoting recovery in long covid: protocol for a randomised placebo-controlled clinical trial (APRIL Trial). BMJ Open 2025; 15:e094526. [PMID: 40280611 PMCID: PMC12035422 DOI: 10.1136/bmjopen-2024-094526] [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: 10/02/2024] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Long covid describes a syndrome of persistent symptoms following COVID-19 and is responsible for substantial healthcare and economic burden. Currently, no effective treatments have been established. Ashwagandha (Withania somnifera (L.) Dunal) is a medicinal herb traditionally used in India for its immune-strengthening and anti-inflammatory properties. Withanolides, a family of steroid-derived molecules unique to Ashwagandha, have been shown to modulate inflammatory pathways in animal models, and several small randomised trials in humans support its effectiveness for reducing symptoms that are also associated with long covid. Therefore, this study aims to assess whether Ashwagandha is effective and safe for improving functional status and reducing symptom burden in adults living with long covid. METHODS A randomised double-blind placebo-controlled trial will be performed at participating general practice (GP) surgeries and long covid clinics across the UK. Individuals diagnosed with long covid will be screened for eligibility and then randomised 1:1 to take 1000 mg daily of Ashwagandha root extract tablets (standardised to <0.9% withanolides) or matching placebo tablets for 3 months (target, n = 2500). Monthly online surveys will be performed to collect patient-reported outcomes, and monthly safety monitoring, including liver function tests, will be conducted by clinical site teams. The primary outcome of the Post-COVID Functional Status Scale score at 3 months will be assessed by baseline-adjusted ordinal logistic regression, according to a pre-published statistical analysis plan. The secondary outcomes included validated quality of life and long covid symptom scales, work status and productivity and adverse events. The trial has been approved as a Clinical Trial of an Investigational Medicinal Produce by the Medicines and Healthcare Regulatory Authority and by the NHS Research Ethics Committee and Health Research Authority. DISCUSSION Treatments for long covid are urgently needed. This trial will robustly evaluate the safety and efficacy of a candidate treatment with a promising efficacy and safety profile. If found to be effective, the findings will likely influence treatment guidelines and improve health outcomes in those living with long covid. TRIAL REGISTRATION NUMBER This trial was pre-registered on 15/08/2022: ISRCTN12368131.
Collapse
Affiliation(s)
| | - Manisha Joshi
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Mahesh Mathpathi
- Department of Global Health and Development, London School of Hygiene & Tropical Medicine, London, UK
| | - Alexander Perkins
- Centre for Global Chronic Conditions, Faculty of Epidemiology and Population Health, Department of Non-communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Tim Clayton
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Anoop Sv Shah
- Centre for Global Chronic Conditions, Faculty of Epidemiology and Population Health, Department of Non-communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Rohini Mathur
- Centre for Primary Care and Public Health, Queen Mary University of London Wolfson Institute of Population Health, London, UK
| | - Nick Birk
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Arandeep Dhillon
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Judith Lieber
- Department of Medical Statistics, London School of Hygiene & Tropical Medicine, London, UK
| | - Sidra S Beg
- The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Lily Hopkins
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Archie Khan
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
| | - Shereen Allaham
- Department of Epidemiology and Public Health, UCL, London, UK
- Aceso Global Health Consultants Ltd, London, UK
| | - Vanessa Tw Kam
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Shailen Sutaria
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
- Global Public Health Unit, Queen Mary University of London, London, UK
| | - Galib R
- All India Institute of Ayurveda, New Delhi, New Delhi, India
| | - S Rajagopala
- All India Institute of Ayurveda, New Delhi, New Delhi, India
| | - Amarjeet Bhamra
- All-Party Parliamentary Group on Indian Traditional Sciences, London, UK
| | | | - Kamlesh Khunti
- Diabetes Research Centre, University of Leicester, Leicester, UK
| | - Tanuja Nesari
- All India Institute of Ayurveda, New Delhi, New Delhi, India
| | - Sanjay Kinra
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
22
|
Kufel WD, Zagoria ZJ, Seabury RW, Zeineddine N, Thomas SJ, Spinler SA, Steele JM. Impact of Remdesivir on Heart Rate and Bradycardia Incidence Among Hospitalized Adults With COVID-19. Ann Pharmacother 2025:10600280251327154. [PMID: 40269618 DOI: 10.1177/10600280251327154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Bradycardia is not currently described as an adverse effect in prescribing information for remdesivir but has been reported postapproval. Therefore, effects on heart rate (HR) and bradycardia incidence after remdesivir initiation may be underrecognized by clinicians. OBJECTIVE To evaluate HR and bradycardia incidence after remdesivir initiation among patients with COVID-19. METHODS This was a single-center, retrospective cohort study between May 1, 2020 and December 1, 2021. Hospitalized patients eligible for inclusion were ≥18 years and received > 1 dose of remdesivir. Patients were excluded if they were pregnant, incarcerated, or received new medications associated with bradycardia. The primary outcome was to evaluate differences in median HR among patients preremdesivir (up to 24 hours prior to remdesivir) and postremdesivir (first dose through the treatment duration). Secondary outcomes included bradycardia episodes postremdesivir, nadir HR postremdesivir, and interventions for bradycardia management. Variables to assess postremdesivir bradycardia were considered in multivariate logistic regression if they had a P < 0.1 on univariate analysis. RESULTS Among 514 patients, 328 were included. Most were male (53.4%), had severe COVID-19 (59.8%), and median (interquartile range [IQR]) age was 62 (23.7) years. Median (IQR) remdesivir duration was 4.9 (1.5) days. Median (IQR) HR was significantly lower postremdesivir than preremdesivir (74 (15) vs 87 (19), P < 0.001). There were significantly more bradycardia episodes postremdesivir than before (48.8% [160/328] vs (2.4% [8/328]), P < 0.001). Among 48.8% (160/328) of patients with bradycardia postremdesivir, median (IQR) nadir HR was 53 (6.8). Remdesivir was discontinued early in 1 patient (0.6%). In multivariate logistic regression, remdesivir duration (odds ratio [OR] = 1.26, 95% confidence interval [CI] = 1.04 to 1.54, P = 0.019) and median preremdesivir HR (OR = 0.96, 95% CI = 0.94 to 0.97, P < 0.001) were identified as significant predictors for bradycardia. CONCLUSION AND RELEVANCE Remdesivir was associated with a significantly lower HR and higher incidence of bradycardia among hospitalized patients with COVID-19. These data may help improve recognition and management of these remdesivir-associated effects during COVID-19 treatment.
Collapse
Affiliation(s)
- Wesley D Kufel
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
- Upstate University Hospital, State University of New York Upstate Medical University, Syracuse, NY, USA
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Zoey J Zagoria
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
- The Brooklyn Hospital Center, Brooklyn, NY, USA
| | - Robert W Seabury
- Upstate University Hospital, State University of New York Upstate Medical University, Syracuse, NY, USA
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Nabil Zeineddine
- Upstate University Hospital, State University of New York Upstate Medical University, Syracuse, NY, USA
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Stephen J Thomas
- Upstate University Hospital, State University of New York Upstate Medical University, Syracuse, NY, USA
- State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Sarah A Spinler
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
| | - Jeffrey M Steele
- Upstate University Hospital, State University of New York Upstate Medical University, Syracuse, NY, USA
- State University of New York Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
23
|
Saito-Tarashima N, Koma T, Hinotani N, Yoshida K, Ogasa M, Murai A, Inoue S, Kondo T, Doi N, Tsuneyama K, Nomaguchi M, Minakawa N. 3-Deazaguanosine inhibits SARS-CoV-2 viral replication and reduces the risk of COVID-19 pneumonia in hamster. iScience 2025; 28:112140. [PMID: 40171487 PMCID: PMC11960675 DOI: 10.1016/j.isci.2025.112140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/12/2024] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
The COVID-19 pandemic highlighted the serious threat that coronaviruses have on public health. Because coronavirus continuously undergoes cross-species transmission, additional therapeutic agents and targets are urgently needed. Here, we show that a 3-deazapurine ribonucleoside, 3-Deazaguanosine (C3Guo, 2), has potent antiviral activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Unexpectedly, C3Guo (2) does not act as an inhibitor of RNA-dependent RNA polymerase (RdRp), which is the therapeutic target of two key nucleoside/nucleotide inhibitors approved for the treatment of COVID-19 (Remdesivir and Molnupiravir); instead, it seems to function by targeting the capping machinery of viral RNA. In hamsters infected with SARS-CoV-2, administration of 2 markedly reduced infectious viral titers, and prevented the development of COVID-19 pneumonia better than Molnupiravir. The potency of 2 against SARS-CoV-2 underscores its potential as an effective therapeutic agent for COVID-19 and future zoonotic coronavirus infections and raises the possibility of antiviral nucleoside analogs with alternative therapeutic targets to RdRp.
Collapse
Affiliation(s)
- Noriko Saito-Tarashima
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Takaaki Koma
- Department of Microbiology, Graduate School of Medicine, Tokushima University, 3-18-15 Kuramoto, Tokushima, Tokushima 770-8503, Japan
- Department of PostLED Photonics Research, Institute of PostLED Photonics, Tokushima University, 2-1 Minamijosanjima, Tokushima, Tokushima 770-8506, Japan
| | - Naoto Hinotani
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Keigo Yoshida
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Moka Ogasa
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Akiho Murai
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Syuya Inoue
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| | - Tomoyuki Kondo
- Department of Microbiology, Graduate School of Medicine, Tokushima University, 3-18-15 Kuramoto, Tokushima, Tokushima 770-8503, Japan
| | - Naoya Doi
- Department of Microbiology, Graduate School of Medicine, Tokushima University, 3-18-15 Kuramoto, Tokushima, Tokushima 770-8503, Japan
| | - Koichi Tsuneyama
- Department of PostLED Photonics Research, Institute of PostLED Photonics, Tokushima University, 2-1 Minamijosanjima, Tokushima, Tokushima 770-8506, Japan
- Department of Pathology and Laboratory Medicine, Graduate School of Medicine, Tokushima University, 3-18-15 Kuramoto, Tokushima, Tokushima 770-8503, Japan
| | - Masako Nomaguchi
- Department of Microbiology, Graduate School of Medicine, Tokushima University, 3-18-15 Kuramoto, Tokushima, Tokushima 770-8503, Japan
- Department of PostLED Photonics Research, Institute of PostLED Photonics, Tokushima University, 2-1 Minamijosanjima, Tokushima, Tokushima 770-8506, Japan
| | - Noriaki Minakawa
- Graduate School of Pharmaceutical Science, Tokushima University, 1-78-1 Shomachi, Tokushima, Tokushima 770-8505, Japan
| |
Collapse
|
24
|
Taha MS, Akram A, Abdelbary GA. Unlocking the potential of remdesivir: innovative approaches to drug delivery. Drug Deliv Transl Res 2025:10.1007/s13346-025-01843-7. [PMID: 40244526 DOI: 10.1007/s13346-025-01843-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2025] [Indexed: 04/18/2025]
Abstract
Given the recurrent waves of COVID-19 and the emergence of new viral infections, optimizing the potential of remdesivir as an antiviral agent is critical. While several reviews have explored the efficacy of remdesivir, few have comprehensively addressed its challenges, such as the necessity for intravenous infusion, suboptimal lung accumulation, and safety concerns related to its formulation. This review critically examines these challenges while proposing innovative solutions and effective combinations with other antiviral agents and repurposed drugs. By highlighting the role of complex generics, we aim to enhance therapeutic efficacy in ways not previously discussed in existing literature. Furthermore, we address the development of novel drug delivery systems which specifically aim to improve remdesivir's pharmacological profile. By analyzing recent findings, we assess both the successes and limitations of current approaches, providing insights into ongoing challenges and strategies for further optimization. This review uniquely focuses on targeted drug delivery systems and innovative formulations, thereby maximizing remdesivir's therapeutic benefits and broadening its application in combating emerging viral threats. In doing so, we fill a critical gap in literature, offering a comprehensive overview that informs future research and clinical strategies.
Collapse
Affiliation(s)
- Maie S Taha
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Alaa Akram
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ghada A Abdelbary
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
25
|
Katayama M, Shikano M. Accelerated therapeutic development during COVID-19: insights, regulatory strategies, and recommendations for future pandemic preparedness. Front Med (Lausanne) 2025; 12:1482035. [PMID: 40303376 PMCID: PMC12037572 DOI: 10.3389/fmed.2025.1482035] [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: 08/17/2024] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Background The clinical development of therapeutics for COVID-19 proceeded at an extraordinary pace. Given the lack of studies evaluating this experience systematically, we analyzed the clinical development methods for COVID-19 therapeutics to determine strategies for shortening the clinical development period in preparation for future pandemics. Methods We confirmed the US-FDA review documents for fourteen products that underwent Emergency Use Authorization (EUA) in the US during the COVID-19 pandemic to examine the time required for clinical development and regulatory review and the submitted data for EUA. Results Six of the fourteen products with clinical study data for other indications were evaluated in fewer studies than new molecular entities. The application data for each product included the stipulated content, and placebo-controlled comparative studies were included for all products. Clinical development measures were adopted, including adaptive protocol design, nonsequential phase development, and clinical dose adaptation based on non-clinical study results. Conclusion Products with clinical study data for other indications are advantageous for early approval. However, early approval of new molecular entities is also important because they may not be sufficiently effective against new infectious diseases. It would be effective to approve a product promptly for a limited target population at first and then gradually expand it as data becomes more abundant. To prepare for future pandemics, we recommend establishing a framework for identifying candidates from existing products, managing and disseminating information in emergencies at various levels, and clarifying the conditions for applying regulatory flexibility to encourage pharmaceutical companies to make early decisions regarding clinical development.
Collapse
Affiliation(s)
- Miyuki Katayama
- Graduate School of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan
| | - Mayumi Shikano
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
26
|
Miyashita N, Horita N, Nakamori Y, Ogata M, Fukuda N, Yamura A, Ito T. Effectiveness of ensitrelvir for cough caused by COVID-19 Omicron variant in patients with asthma. Microbiol Spectr 2025; 13:e0340724. [PMID: 40231685 PMCID: PMC12054164 DOI: 10.1128/spectrum.03407-24] [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: 12/27/2024] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
In the post-acute coronavirus disease 2019 (COVID-19) period, patients with asthma had a significantly higher risk of cough than patients without a history of asthma. In particular, cases with persistent cough were increased during the Omicron variant epidemic. In this study, we evaluated the efficacy of ensitrelvir for the treatment of cough associated with COVID-19 Omicron variants in patients with asthma. This follows the Strengthening the Reporting of Observational Studies in Epidemiology statement. A total of 223 patients were registered in this study: 121 patients chose ensitrelvir, and 102 patients chose symptomatic treatment. Cough severity, frequency, and cough-specific quality of life were evaluated using the Japanese version of the Leicester Cough Questionnaire (J-LCQ). J-LCQ documented at baseline on days 4, 7, and 14 for all patients showed a steady improvement over time in both groups. In the mixed model for repeated measures model, which accounts for repeated measurements, the change in J-LCQ score from baseline was 2.1 points higher in the ensitrelvir group (P <0.001). Additionally, patients who were using triple inhaled therapy at baseline showed a 2.3-point higher change in J-LCQ score from baseline (P <0.001). Multiple regression analysis was performed at days 4, 7, and 14, with the change in J-LCQ score from baseline as the dependent variable. Ensitrelvir was associated with scores that were 3.1 points higher on day 4, 3.5 points higher on day 7, and 2.0 points higher on day 14 compared with symptomatic treatment (P <0.001 for all). In conclusion, our results demonstrated that early administration of ensitrelvir may be effective as a treatment for cough due to the COVID-19 Omicron variant. IMPORTANCE We evaluated the efficacy of ensitrelvir for the treatment of cough due to coronavirus disease 2019 (COVID-19) Omicron variant in patients with asthma. A total of 223 patients were registered in this study: 121 patients chose ensitrelvir, and 102 patients chose symptomatic treatment. Cough severity, frequency, and cough-specific quality of life were evaluated using the Japanese version of the Leicester Cough Questionnaire (J-LCQ). Multiple regression analysis was performed at days 4, 7, and 14, with the change in J-LCQ score from baseline as the dependent variable. Ensitrelvir was associated with scores that were 3.1 points higher on day 4, 3.5 points higher on day 7, and 2.0 points higher on day 14 compared with symptomatic treatment (P <0.001 for all). Our results demonstrated that early administration of ensitrelvir may be effective as a treatment for cough due to the COVID-19 Omicron variant.
Collapse
Affiliation(s)
- Naoyuki Miyashita
- First Department of Internal Medicine, Division of Respiratory Medicine, Infectious Disease and Allergology, Kansai Medical University, Hirakata, Japan
| | - Nobuyuki Horita
- Chemotherapy Center, Yokohama City University Hospital, Yokohama, Japan
| | - Yasushi Nakamori
- Department of Emergency Medicine, Kansai Medical University Medical Center, Hirakata, Japan
| | - Makoto Ogata
- First Department of Internal Medicine, Division of Respiratory Medicine, Infectious Disease and Allergology, Kansai Medical University, Hirakata, Japan
| | - Naoki Fukuda
- First Department of Internal Medicine, Division of Respiratory Medicine, Infectious Disease and Allergology, Kansai Medical University, Hirakata, Japan
| | - Akihisa Yamura
- First Department of Internal Medicine, Division of Respiratory Medicine, Infectious Disease and Allergology, Kansai Medical University, Hirakata, Japan
| | - Tomoki Ito
- First Department of Internal Medicine, Division of Respiratory Medicine, Infectious Disease and Allergology, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
27
|
Müller T, Dzanibe S, Day C, Mpangase PT, Chimbetete T, Pedretti S, Schwager S, Gray CM, Sturrock E, Peter J. Integrated renin angiotensin system dysregulation and immune profiles predict COVID-19 disease severity in a South African cohort. Sci Rep 2025; 15:12799. [PMID: 40229302 PMCID: PMC11997227 DOI: 10.1038/s41598-025-96161-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/26/2025] [Indexed: 04/16/2025] Open
Abstract
Renin-angiotensin system (RAS) dysregulation is an important component of the complex pathophysiology of SARS-CoV-2 and other coronavirus infections. Thus, angiotensin-converting enzyme 2 (ACE2), the entry receptor and key to the alternative RAS, was proposed as a severity/prognostic biomarker for risk-stratification. However, experimental RAS data from diverse cohorts are limited, particularly analyses integrating RAS with immune biomarkers. Participants (n = 172) in Cape Town were sampled longitudinally (including a recovery timepoint [> 3-month]), across WHO asymptomatic to critical severity. Using fluorometric assays and LC-MS/MS RAS Fingerprinting®, results show serum ACE1 activity significantly decreases with increasing COVID-19 severity (P < 0.01) and mortality (P < 0.05), while increased ACE2 activity is associated with worse severity (P < 0.01). Neither enzyme activity correlates with viral load proxy or nasal ACE mRNA levels. ACE1 and ACE2 activities were the most effective severity biomarkers compared to 96 established immune markers obtained via proximity extension assay, as demonstrated by principal component analysis. A multivariate variable selection model using random forest classification identified biomarkers discriminating COVID-19 severity (AUC = 0.82), the strongest being HGF, EN-RAGE, cathepsin L. Adding ACE1 activity and anti-SARS-CoV-2 antibody titres improved differentiation between ambulatory and hospitalised participants. Notably, RAS dysregulation has unique severity associations in coronavirus infections with implications for treatment and pathophysiological mechanisms.
Collapse
Affiliation(s)
- Talitha Müller
- Division of Allergology and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sonwabile Dzanibe
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Cascia Day
- Division of Allergology and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Allergy and Immunology Unit, University of Cape Town Lung Institute, Cape Town, South Africa
| | - Phelelani Thokozani Mpangase
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Tafadzwa Chimbetete
- Division of Allergology and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sarah Pedretti
- Allergy and Immunology Unit, University of Cape Town Lung Institute, Cape Town, South Africa
| | - Sylva Schwager
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Clive M Gray
- Division of Molecular Biology and Human Genetics, Stellenbosch University, Stellenbosch, South Africa
| | - Edward Sturrock
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jonny Peter
- Division of Allergology and Clinical Immunology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
- Allergy and Immunology Unit, University of Cape Town Lung Institute, Cape Town, South Africa.
| |
Collapse
|
28
|
Arribas López JR, Ruiz Seco MP, Fanjul F, Díaz Pollán B, González Ruano Pérez P, Ferre Beltrán A, De Miguel Buckley R, Portillo Horcajada L, De Álvaro Pérez C, Barroso Santos Carvalho PJ, Riera Jaume M. Remdesivir associated with reduced mortality in hospitalized COVID-19 patients: treatment effectiveness using real-world data and natural language processing. BMC Infect Dis 2025; 25:513. [PMID: 40217145 PMCID: PMC11992806 DOI: 10.1186/s12879-025-10817-6] [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: 01/15/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Remdesivir (RDV) was the first antiviral approved for mild-to-moderate COVID-19 and for those patients at risk for progression to severe disease after clinical trials supported its association with improved outcomes. Real-world evidence (RWE) generated by artificial intelligence techniques could potentially expedite the validation of new treatments in future health crises. We aimed to use natural language processing (NLP) and machine learning (ML) to assess the impact of RDV on COVID19-associated outcomes including time to discharge and in-hospital mortality. METHODS Using EHRead®, an NLP technology including SNOMED-CT terminology that extracts unstructured clinical information from electronic health records (EHR), we retrospectively examined hospitalized COVID-19 patients with moderate-to-severe pneumonia in three Spanish hospitals between January 2021 and March 2022. Among RDV eligible patients, treated (RDV+) vs untreated (RDV‒) patients were compared after propensity score matching (PSM; 1:3.3 ratio) based on age, sex, Charlson comorbidity index, COVID-19 vaccination status, other COVID-19 treatment, hospital, and variant period. Cox proportional hazards models and Kaplan-Meier plots were used to assess statistical differences between groups. RESULTS Among 7,651,773 EHRs from 84,408 patients, 6,756 patients were detected with moderate-to-severe COVID-19 pneumonia during the study period. The study population was defined with 4,882 (72.3%) RDV eligible patients. The median age was 72 years and 57.3% were male. A total of 812 (16.6%) patients were classified as RDV+ and were matched to 2,703 RDV‒ patients (from a total of 4,070 RDV‒). After PSM, all covariates had an absolute mean standardized difference of less than 10%. The hazard ratio for in-hospital mortality at 28 days was 0.73 (95% confidence interval, CI, 0.56 to 0.96, p = 0.022) with RDV‒ as the reference group. Risk difference and risk ratio at 28 days was 2.7% and 0.76, respectively, both favoring the RDV+ group. No differences were found in length of hospital stay since RDV eligibility between groups. CONCLUSIONS Using NLP and ML we were able to generate RWE on the effectiveness of RDV in COVID-19 patients, confirming the potential of using this methodology to measure the effectiveness of treatments in pandemics. Our results show that using RDV in hospitalized patients with moderate-to-severe pneumonia is associated with significantly reduced inpatient mortality. Adherence to clinical guideline recommendations has prognostic implications and emerging technologies in identifying eligible patients for treatment and avoiding missed opportunities during public health crises are needed.
Collapse
Affiliation(s)
- José Ramón Arribas López
- Infectious Diseases Unit, Internal Medicine Service, Hospital Universitario La Paz, Hospital La Paz Institute for Health Research (IdiPAZ), Ciber Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain.
| | | | - Francisco Fanjul
- Infectious Diseases Unit, Internal Medicine Service, Hospital Universitario Son Espases, Fundació Institut de Investigació Sanitaria de Les Illes Balears (IdISBa), Palma de Mallorca, Spain
| | - Beatriz Díaz Pollán
- Infectious Diseases Unit, Internal Medicine Service, Hospital Universitario La Paz, Hospital La Paz Institute for Health Research (IdiPAZ), Ciber Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | | | - Adrián Ferre Beltrán
- Infectious Diseases Unit, Internal Medicine Service, Hospital Universitario Son Espases, Fundació Institut de Investigació Sanitaria de Les Illes Balears (IdISBa), Palma de Mallorca, Spain
| | - Rosa De Miguel Buckley
- Infectious Diseases Unit, Internal Medicine Service, Hospital Universitario La Paz, Hospital La Paz Institute for Health Research (IdiPAZ), Ciber Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | | | | | | | - Melchor Riera Jaume
- Infectious Diseases Unit, Internal Medicine Service, Hospital Universitario Son Espases, Fundació Institut de Investigació Sanitaria de Les Illes Balears (IdISBa), Palma de Mallorca, Spain
| |
Collapse
|
29
|
Lemieux G, Pérez-Vargas J, Désilets A, Hassanzadeh M, Thompson CAH, Gravel-Trudeau A, Joushomme A, Ennis S, Villanueva I, Marouseau É, Fraser BJ, Champagne W, Lepage M, Niikura M, Arrowsmith CH, Jean F, Leduc R, Boudreault PL. From N-0385 to N-0920: Unveiling a Host-Directed Protease Inhibitor with Picomolar Antiviral Efficacy against Prevalent SARS-CoV-2 Variants. J Med Chem 2025; 68:7119-7136. [PMID: 40163818 PMCID: PMC11998928 DOI: 10.1021/acs.jmedchem.4c02468] [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: 10/11/2024] [Revised: 01/30/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025]
Abstract
The worldwide spread of new SARS-CoV-2 variants emphasizes the need to diversify existing therapeutic strategies. TMPRSS2, a host protease crucial for SARS-CoV-2 entry, has garnered significant research attention as a potential target for therapeutic intervention. Here, we optimized N-0385, a previously reported TMPRSS2 ketobenzothiazole-based peptidomimetic inhibitor, by screening 135 derivatives for target affinity and antiviral potency. Among the top candidates, N-0695 exhibited low nanomolar Ki values against three TTSPs associated with respiratory virus entry: TMPRSS2, matriptase, and TMPRSS13. Notably, N-0920 demonstrated exceptional potency in reducing SARS-CoV-2 variants EG.5.1 and JN.1 entry in Calu-3 cells, representing the first in cellulo picomolar inhibitor with EC50 values of 300 and 90 pM, respectively. Additionally, molecular modeling provided insights into the binding interactions between the compounds and their targets. This study underscores the effectiveness of our screening approach in refining an existing peptidomimetic scaffold to enhance selectivity and antiviral activity.
Collapse
Affiliation(s)
- Gabriel Lemieux
- Department
of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke,
Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke J1H5N4, Quebec, Canada
| | - Jimena Pérez-Vargas
- Department
of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Antoine Désilets
- Department
of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke,
Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke J1H5N4, Quebec, Canada
| | - Malihe Hassanzadeh
- Department
of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke,
Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke J1H5N4, Quebec, Canada
| | - Connor A. H. Thompson
- Department
of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Alice Gravel-Trudeau
- Department
of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke,
Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke J1H5N4, Quebec, Canada
| | - Alexandre Joushomme
- Department
of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke,
Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke J1H5N4, Quebec, Canada
| | - Siobhan Ennis
- Faculty
of Health Sciences, Simon Fraser University, Burnaby J1H5N4, British Columbia, Canada
| | - Ivan Villanueva
- Department
of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Étienne Marouseau
- Department
of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke,
Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke J1H5N4, Quebec, Canada
| | - Bryan J. Fraser
- Department
of Medical Biophysics, University of Toronto, Toronto M5S 1A1, Ontario, Canada
- Structural
Genomics Consortium, University of Toronto, Toronto M5S 1A1, Ontario, Canada
| | - William Champagne
- Department
of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke,
Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke J1H5N4, Quebec, Canada
| | - Matthieu Lepage
- Department
of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke,
Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke J1H5N4, Quebec, Canada
| | - Masahiro Niikura
- Faculty
of Health Sciences, Simon Fraser University, Burnaby J1H5N4, British Columbia, Canada
| | - Cheryl H. Arrowsmith
- Department
of Medical Biophysics, University of Toronto, Toronto M5S 1A1, Ontario, Canada
- Structural
Genomics Consortium, University of Toronto, Toronto M5S 1A1, Ontario, Canada
- Princess
Margaret Cancer Centre, Toronto M5S 1A1, Ontario, Canada
| | - François Jean
- Department
of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Richard Leduc
- Department
of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke,
Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke J1H5N4, Quebec, Canada
| | - Pierre-Luc Boudreault
- Department
of Pharmacology-Physiology, Institut de Pharmacologie de Sherbrooke,
Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke J1H5N4, Quebec, Canada
| |
Collapse
|
30
|
Lee AR, Min HK, Lee SY, Jeon SB, Lee CR, Kim TH, Park JH, La Cho M. Remdesivir alleviates joint damage in collagen-induced arthritis and inhibits inflammatory cell death of RA synovial fibroblasts. Immunol Lett 2025; 275:107009. [PMID: 40189155 DOI: 10.1016/j.imlet.2025.107009] [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: 09/10/2024] [Revised: 03/18/2025] [Accepted: 03/31/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND The antiviral agent, remdesivir, is adenosine analogue which is currently also used as anti-coronavirus disease 2019. Remdesivir also had anti-inflammatory effect which reduced pro-inflammatory cytokine production, and inhibition of the cyclic GMP-AMP synthase-STING pathway. METHODS We evaluated the antiarthritic effects of remdesivir in a mouse model of High-fat diet (HFD) collagen-induced arthritis (CIA) and in fibroblast-like synoviocytes from patients with RA. Type II collagen was administered to DBA/1J mice to induce CIA. Vehicle or remdesivir was injected subcutaneously three times a week. During 7 weeks of treatment, the arthritis score and incidence were evaluated twice a week. Flow cytometry and confocal imaging were used to evaluate CD4 + T cells in the spleen. FLSs from patients with RA were stimulated in vitro with remdesivir and tumor necrosis factor (TNF)-α, and western blotting was used to measure the expression of STING and necroptosis-related markers. RESULTS Remdesivir administration suppressed the incidence and progression of arthritis in mice with CIA. Histological analysis revealed lower inflammation and cartilage damage scores in remdesivir-treated than in vehicle groups. Interleukin (IL)-17 + CD4 + T-cell differentiation was inhibited in the remdesivir-treated group. Furthermore, IL-17/-6/-1β, monocyte chemoattractant protein -1, and TNF-α expression was reduced in the remdesivir group. In vitro, remdesivir suppressed the expression of STING, nuclear factor-κB, RIPK3, and phosphorylated MLKL in RA-FLSs under TNF-α stimulation. CONCLUSIONS The antiviral agent remdesivir suppressed arthritis by regulating Th cell differentiation, pro-inflammatory cytokine expression, the STING pathway, and necroptosis.
Collapse
Affiliation(s)
- A Ram Lee
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Hong Ki Min
- Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, Seoul 05030, Republic of Korea
| | - Seon-Yeong Lee
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Su Been Jeon
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Chae Rim Lee
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Tae Ho Kim
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin Hyung Park
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Mi- La Cho
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea; Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea.
| |
Collapse
|
31
|
Fekrvand S, Saleki K, Abolhassani H, Almasi-Hashiani A, Hakimelahi A, Zargarzadeh N, Yekaninejad MS, Rezaei N. COVID-19 infection in inborn errors of immunity and their phenocopies: a systematic review and meta-analysis. Infect Dis (Lond) 2025:1-35. [PMID: 40178994 DOI: 10.1080/23744235.2025.2483339] [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: 10/13/2024] [Revised: 02/09/2025] [Accepted: 02/23/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Inborn errors of immunity (IEI) are congenital disorders of the immune system. Due to impaired immune system, they are at a higher risk to develop a more severe COVID-19 course compared to general population. OBJECTIVES Herein, we aimed to systematically review various aspects of IEI patients infected with SARS-CoV-2. Moreover, we performed a meta-analysis to determine the frequency of COVID-19 in patients with different IEI. METHODS Embase, Web of Science, PubMed, and Scopus were searched introducing terms related to IEI and COVID-19. RESULTS 3646 IEI cases with a history of COVID-19 infection were enrolled. The majority of patients had critical infections (1013 cases, 27.8%). The highest frequency of critical and severe cases was observed in phenocopies of IEI (95.2%), defects in intrinsic and innate immunity (69.4%) and immune dysregulation (23.9%). 446 cases (12.2%) succumbed to the disease and the highest mortality was observed in IEI phenocopies (34.6%). COVID-19 frequency in immunodeficient patients was 11.9% (95% CI: 8.3 to 15.5%) with innate immunodeficiency having the highest COVID-19 frequency [34.1% (12.1 to 56.0%)]. COVID-19 case fatality rate among IEI patients was estimated as 5.4% (95% CI: 3.5-8.3%, n = 8 studies, I2 = 17.5%). CONCLUSION IEI with underlying defects in specific branches of the immune system responding to RNA virus infection experience a higher frequency and mortality of COVID-19 infection. Increasing awareness about these entities and underlying genetic defects, adherence to prophylactic strategies and allocating more clinical attention to these patients could lead to a decrease in COVID-19 frequency and mortality in these patients.
Collapse
Affiliation(s)
- Saba Fekrvand
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Kiarash Saleki
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Division of Clinical Immunology, Department of Biosciences and Nutrition, KarolinskaInstitutet, Karolinska University Hospital, Stockholm, Sweden
| | - Amir Almasi-Hashiani
- Department of Epidemiology, School of Health, Arak University of Medical Sciences, Arak, Iran
| | - Ali Hakimelahi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nikan Zargarzadeh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mir Saeed Yekaninejad
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
32
|
Mozaffari E, Chandak A, Amin AN, Gottlieb RL, Kalil AC, Sarda V, Berry M, Brown G, Okulicz JF, Chima-Melton C. Racial and Ethnic Disparities in COVID-19 Treatments in the United States. J Racial Ethn Health Disparities 2025; 12:1052-1062. [PMID: 38409487 PMCID: PMC11914345 DOI: 10.1007/s40615-024-01942-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/28/2024]
Abstract
INTRODUCTION Racial and ethnic disparities in patient outcomes following COVID-19 exist, in part, due to factors involving healthcare delivery. The aim of the study was to characterize disparities in the administration of evidence-based COVID-19 treatments among patients hospitalized for COVID-19. METHODS Using a large, US hospital database, initiation of COVID-19 treatments was compared among patients hospitalized for COVID-19 between May 2020 and April 2022 according to patient race and ethnicity. Multivariate logistic regression models were used to examine the effect of race and ethnicity on the likelihood of receiving COVID-19 treatments, stratified by baseline supplemental oxygen requirement. RESULTS The identified population comprised 317,918 White, 76,715 Black, 9297 Asian, and 50,821 patients of other or unknown race. There were 329,940 non-Hispanic, 74,199 Hispanic, and 50,622 patients of unknown ethnicity. White patients were more likely to receive COVID-19 treatments, and specifically corticosteroids, compared to Black, Asian, and other patients (COVID-19 treatment: 87% vs. 81% vs. 85% vs. 84%, corticosteroids: 85% vs. 79% vs. 82% vs. 82%). After covariate adjustment, White patients were significantly more likely to receive COVID-19 treatments than Black patients across all levels of supplemental oxygen requirement. No clear trend in COVID-19 treatments according to ethnicity (Hispanic vs. non-Hispanic) was observed. CONCLUSION There were important racial disparities in inpatient COVID-19 treatment initiation, including the undertreatment of Black patients and overtreatment of White patients. Our new findings reveal the actual magnitude of this issue in routine clinical practice to clinicians, policymakers, and guideline developers. This is crucial to ensuring equitable and appropriate access to evidence-based therapies.
Collapse
Affiliation(s)
| | | | | | - Robert L Gottlieb
- Baylor University Medical Center, Dallas, TX, USA
- Baylor Scott & White Heart and Vascular Hospital, Dallas, TX, USA
- Baylor Scott & White The Heart Hospital, Plano, TX, USA
- Baylor Scott & White Research Institute, Dallas, TX, USA
| | - Andre C Kalil
- University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | | - Chidinma Chima-Melton
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, UCLA Health System, David Geffen School of Medicine at University of California, Los Angeles, CA, USA.
| |
Collapse
|
33
|
Maglione MA, Klausner JD, Wirnkar PK, Fallarme I, Lak R, Sysawang K, Fu N, Yagyu S, Motala A, Tolentino D, Hempel S. A Rapid Systematic Review of U.S. Food and Drug Administration-Authorized COVID-19 Treatments. Open Forum Infect Dis 2025; 12:ofaf097. [PMID: 40225829 PMCID: PMC11986950 DOI: 10.1093/ofid/ofaf097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 02/14/2025] [Indexed: 04/15/2025] Open
Abstract
Background The coronavirus disease 2019 (COVID-19) pandemic era saw numerous treatments authorized for emergency use by the United States (US) Food and Drug Administration (FDA). The purpose of the review was to determine if convalescent plasma, antivirals, or monoclonal antibodies are associated with serious adverse events (SAEs) and, if so, which specific populations are at risk. Methods PubMed, ClinicalTrials.gov, and the FDA submission database were searched through December 2023, and the Infectious Diseases Society of America guidelines, international COVID Network Meta-analysis database, and systematic reviews were reference mined to identify controlled studies with at least 1 US site. Reviewers abstracted study characteristics, number of patients experiencing each type of SAE, and methods of adverse event collection and reporting. Results Fifty-four studies met inclusion criteria, including 31 randomized controlled trials. We found insufficient evidence of association of any SAE with antivirals and spike protein receptor-binding antibodies. In patients hospitalized with COVID-19, the monoclonal antibody tocilizumab, an interleukin 6 inhibitor, may be associated with elevated risk of neutropenia (moderate certainty) and infection (limited certainty). Convalescent plasma may be associated with thrombotic events (limited certainty) as well as bleeding events and infection in patients with hematologic cancers (moderate certainty). Inclusion of studies without a US site could potentially change the findings. Conclusions Severe COVID-19 infection may have serious consequences, especially in hospitalized patients with comorbidities. These consequences may be confused with toxicities of the interventions. Based on our analysis, approved treatments for COVID-19 should be prescribed as clinically indicated, although continued vigilance is warranted to identify rare and potentially significant toxicities that may arise in clinical practice. Clinical Trials Registration PROSPERO (CRD42023467821).
Collapse
Affiliation(s)
- Margaret A Maglione
- Southern California Evidence Review Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Jeffrey D Klausner
- Southern California Evidence Review Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Patricia K Wirnkar
- Southern California Evidence Review Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Ivan Fallarme
- Southern California Evidence Review Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Rozhin Lak
- Southern California Evidence Review Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Kimny Sysawang
- Southern California Evidence Review Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Ning Fu
- Southern California Evidence Review Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Sachi Yagyu
- Southern California Evidence Review Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Aneesa Motala
- Southern California Evidence Review Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Danica Tolentino
- Southern California Evidence Review Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Susanne Hempel
- Southern California Evidence Review Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| |
Collapse
|
34
|
Iriyama C, Ichikawa T, Tamura T, Takahata M, Ishio T, Ibata M, Kawai R, Iwata M, Suzuki M, Adachi H, Nao N, Suzuki H, Kawai A, Kamiyama A, Suzuki T, Hirata Y, Iida S, Katano H, Ishii Y, Tsuji T, Oda Y, Tanaka S, Okazaki N, Katayama Y, Nakagawa S, Tsukamoto T, Doi Y, Fukuhara T, Murata T, Tomita A. Clinical and molecular landscape of prolonged SARS-CoV-2 infection with resistance to remdesivir in immunocompromised patients. PNAS NEXUS 2025; 4:pgaf085. [PMID: 40160532 PMCID: PMC11950820 DOI: 10.1093/pnasnexus/pgaf085] [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: 06/17/2024] [Accepted: 02/15/2025] [Indexed: 04/02/2025]
Abstract
Patients with hematologic diseases have experienced coronavirus disease 2019 (COVID-19) with a prolonged, progressive course. Here, we present clinical, pathological, and virological analyses of three cases of prolonged COVID-19 among patients undergoing treatment for B-cell lymphoma. These patients had all been treated with anti-CD20 antibody and bendamustine. Despite various antiviral treatments, high severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) levels persisted for >4 weeks, and two of them succumbed to COVID-19. The autopsy showed bronchopneumonia, interstitial pneumonia, alveolar hemorrhage, and fibrosis. Overlapping cytomegalovirus, fungal and/or bacterial infections were also confirmed. Sequencing of SARS-CoV-2 showed accumulation of mutations and changes in variant allele frequencies over time. NSP12 mutations V792I and M794I appeared independently in two cases as COVID-19 progressed. In vitro drug susceptibility analysis and an animal experiment using recombinant SARS-CoV-2 demonstrated that each mutation, V792 and M794I, was independently responsible for remdesivir resistance and attenuated pathogenicity. E340A, E340D, and F342INS mutations in the spike protein were found in one case, which may account for the sotrovimab resistance. Analysis of autopsy specimens indicated heterogeneous distribution of these mutations. In summary, we demonstrated temporal and spatial diversity in SARS-CoV-2 that evolved resistance to various antiviral agents in malignant lymphoma patients under immunodeficient conditions caused by certain types of immunochemotherapies. Strategies may be necessary to prevent the acquisition of drug resistance and improve outcomes, such as the selection of appropriate treatment strategies for lymphoma considering patients' immune status and the institution of early intensive antiviral therapy.
Collapse
Affiliation(s)
- Chisako Iriyama
- Department of Hematology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan
| | - Takaya Ichikawa
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo 060-8638, Japan
- Department of Hematology, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
- Department of Hematology, Sapporo City General Hospital, Sapporo 060-8604, Japan
| | - Tomokazu Tamura
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo 060-8638, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo 001-0021, Japan
- One Health Research Center, Hokkaido University, Sapporo 060-0818, Japan
| | - Mutsumi Takahata
- Department of Hematology, Sapporo-Kosei General Hospital, Sapporo 060-0033, Japan
| | - Takashi Ishio
- Department of Hematology, Sapporo-Kosei General Hospital, Sapporo 060-0033, Japan
| | - Makoto Ibata
- Department of Hematology, Sapporo-Kosei General Hospital, Sapporo 060-0033, Japan
| | - Ryuji Kawai
- Department of Emergency and General Internal Medicine, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Mitsunaga Iwata
- Department of Emergency and General Internal Medicine, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Masahiro Suzuki
- Department of Microbiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Hirokazu Adachi
- Department of Microbiology and Medical Zoology, Aichi Prefectural Institute of Public Health, Nagoya 462-8576, Japan
| | - Naganori Nao
- One Health Research Center, Hokkaido University, Sapporo 060-0818, Japan
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| | | | - Akito Kawai
- Department of Microbiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Akifumi Kamiyama
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo 060-8638, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Yuichiro Hirata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Shun Iida
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Harutaka Katano
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Yasushi Ishii
- Department of Pathology, Sapporo City General Hospital, Sapporo 060-8604, Japan
| | - Takahiro Tsuji
- Department of Pathology, Sapporo City General Hospital, Sapporo 060-8604, Japan
| | - Yoshitaka Oda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo 001-0021, Japan
| | - Nanase Okazaki
- Department of Pathology, Sapporo City General Hospital, Sapporo 060-8604, Japan
| | - Yuko Katayama
- Department of Pathology, Sapporo City General Hospital, Sapporo 060-8604, Japan
| | - Shimpei Nakagawa
- Department of Pathology, Sapporo City General Hospital, Sapporo 060-8604, Japan
| | - Tetsuya Tsukamoto
- Department of Diagnostic Pathology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Yohei Doi
- Department of Microbiology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
- Center for Infectious Disease Research, Fujita Health University, Toyoake 470-1192, Japan
- Department of Infectious Diseases, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Takasuke Fukuhara
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo 060-8638, Japan
- Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo 001-0021, Japan
- One Health Research Center, Hokkaido University, Sapporo 060-0818, Japan
- Laboratory of Virus Control, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan
- AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo 100-0004, Japan
| | - Takayuki Murata
- Center for Infectious Disease Research, Fujita Health University, Toyoake 470-1192, Japan
- Department of Virology, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Akihiro Tomita
- Department of Hematology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake 470-1192, Japan
| |
Collapse
|
35
|
Kwok W, Shea Y, Ho J, Lam D, Tam T, Tam A, Ip M, Hung I. Implication of Admission Eosinophil Count and Prognosis of Coronavirus Disease 2019 (COVID-19) in Elderly Patients With COPD: A Territory-Wide Cohort Study. THE CLINICAL RESPIRATORY JOURNAL 2025; 19:e70070. [PMID: 40143637 PMCID: PMC11947431 DOI: 10.1111/crj.70070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/15/2024] [Accepted: 02/25/2025] [Indexed: 03/28/2025]
Abstract
OBJECTIVES This study aims to investigate the association between elderly patients with COPD with different blood eosinophil on admission and those without COPD and the prognosis of COVID-19. METHOD A territory-wide retrospective study was conducted to investigate the association between elderly COPD patients with different blood eosinophil on admission and the prognosis of COVID-19. Elderly patients admitted to public hospitals and community treatment facility in Hong Kong for COVID-19 from January 23, 2020, to September 31, 2021, were included in the study. Severe diseases were defined as those who develop respiratory complications, systemic complications and death. RESULTS Among the 1925 patients included, 133 had COPD. Forty had admission blood eosinophil count ≥ 150 cells/μL, and 93 had blood eosinophil count < 150 cells/μL. Patients with COPD and admission blood eosinophil count ≥ 150 cells/μL, but not those with admission blood eosinophil count < 150 cells/μL, had severe COVID-19 with the development of respiratory and systemic complications. They were more likely to develop respiratory failure (OR = 5.235, 95% CI = 2.088-13.122, p < 0.001) and require invasive mechanical ventilation (OR = 2.433, 95% CI = 1.022-5.791, p = 0.045) and intensive care unit admission (OR = 2.214, 95% CI = 1.004-4.881, p = 0.049). DISCUSSION Our study suggested that the blood eosinophil count on admission could have significant prognostic implications among elderly patients with COPD. Patients with COPD and admission blood eosinophil count ≥ 150 cells/μL, but not those with admission blood eosinophil count < 150 cells/μL, have significantly increased risks of developing respiratory and systemic complications from COVID-19, when compared with non-COPD patients.
Collapse
Affiliation(s)
- Wang Chun Kwok
- Department of MedicineThe University of Hong Kong, Queen Mary HospitalPokfulamHong KongChina
| | - Yat Fung Shea
- Department of MedicineThe University of Hong Kong, Queen Mary HospitalPokfulamHong KongChina
| | - James Chung Man Ho
- Department of MedicineThe University of Hong Kong, Queen Mary HospitalPokfulamHong KongChina
| | - David Chi Leung Lam
- Department of MedicineThe University of Hong Kong, Queen Mary HospitalPokfulamHong KongChina
| | - Terence Chi Chun Tam
- Department of MedicineThe University of Hong Kong, Queen Mary HospitalPokfulamHong KongChina
| | - Anthony Raymond Tam
- Department of MedicineThe University of Hong Kong, Queen Mary HospitalPokfulamHong KongChina
| | - Mary Sau Man Ip
- Department of MedicineThe University of Hong Kong, Queen Mary HospitalPokfulamHong KongChina
| | - Ivan Fan Ngai Hung
- Department of MedicineThe University of Hong Kong, Queen Mary HospitalPokfulamHong KongChina
| |
Collapse
|
36
|
Inoue A, Ichikawa T, Wada D, Maruyama S, Shimazu H, Kashihara M, Okuda K, Saito F, Fukuhara T, Nakamori Y. M49L and other drug resistance mutations emerging in individuals after administration of ensitrelvir in Japanese clinical settings. Antiviral Res 2025; 236:106118. [PMID: 39970959 DOI: 10.1016/j.antiviral.2025.106118] [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: 09/19/2024] [Revised: 10/29/2024] [Accepted: 02/15/2025] [Indexed: 02/21/2025]
Abstract
Recent in-vitro and in-vivo studies and analysis of genomic information registered in GISAID have raised concerns about drug resistance mutations such as M49L after treatment with the 3C-like protease inhibitor ensitrelvir. The aim of this study was to identify resistance gene mutations to 3C-like protease inhibitors, including M49L mutations, in Japanese clinical settings. Genomic analysis of samples from our hospital admissions showed M49L mutations associated with ensitrelvir treatment in three cases and M49L mutation unrelated to ensitrelvir treatment in three cases. In a study of cases with persistent infection or rebound in viral load after 5 days of ensitrelvir treatment, 10 of 16 patients had M49L mutations and 5 had M49I mutations. Resistance gene mutations following treatment with ensitrelvir were shown to emerge even within individual patients who were not immunocompromised. In a study of persistent SARS-CoV-2 infection in severely immunocompromised patients, various drug resistance mutations emerged, with the M49L mutation especially showing a tendency to be a majority mutation. The current status of drug resistance mutations occurring in individuals following administration of ensitrelvir in Japanese clinical settings was clinically investigated for the first time. Considering that the barrier to resistance to ensitrelvir is lower than that to other antiviral drugs and that M49L is a unique mutation that occurs quickly, tends to become a majority mutation, and is maintained thereafter through its ability to replicate, the spread of strains that have acquired ensitrelvir resistance should be closely monitored.
Collapse
Affiliation(s)
- Akira Inoue
- Genome Analysis Center, Kansai Medical University General Medical Center, Osaka, Japan.
| | - Takaya Ichikawa
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan.
| | - Daiki Wada
- Department of Emergency and Critical Care Medicine, Kansai Medical University General Medical Center, Osaka, Japan.
| | - Shuhei Maruyama
- Department of Emergency and Critical Care Medicine, Kansai Medical University General Medical Center, Osaka, Japan.
| | - Haruka Shimazu
- Department of Emergency and Critical Care Medicine, Kansai Medical University General Medical Center, Osaka, Japan.
| | - Masami Kashihara
- Genome Analysis Center, Kansai Medical University General Medical Center, Osaka, Japan.
| | - Kazuyuki Okuda
- Genome Analysis Center, Kansai Medical University General Medical Center, Osaka, Japan.
| | - Fukuki Saito
- Department of Emergency and Critical Care Medicine, Kansai Medical University General Medical Center, Osaka, Japan.
| | - Takasuke Fukuhara
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Japan.
| | - Yasushi Nakamori
- Department of Emergency and Critical Care Medicine, Kansai Medical University General Medical Center, Osaka, Japan.
| |
Collapse
|
37
|
Wurm J, Ritz N, Zimmermann P. Coronavirus disease 2019 (COVID-19) in children: Evolving epidemiology, immunology, symptoms, diagnostics, treatment, post-COVID-19 conditions, prevention strategies, and future directions. J Allergy Clin Immunol 2025; 155:1071-1081. [PMID: 39551439 DOI: 10.1016/j.jaci.2024.11.012] [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: 08/29/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 11/19/2024]
Abstract
The epidemiology of coronavirus disease 2019 (COVID-19) in children has evolved throughout the pandemic, with initially low infection rates rising significantly as a result of the emergence of the more transmissible Omicron variant. Adolescents, children from ethnic minorities and lower-income households, and those with obesity are at increased risk of contracting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The immune response in children leads to milder symptoms compared to adults, with fever and cough being most frequent; tough symptoms vary by SARS-CoV-2 variant and age. Diagnostic methods to confirm current or past infection include reverse transcription PCR, rapid antigen tests, and serology. Treatment is mainly supportive, with antivirals and glucocorticoids reserved for severe cases. While serious conditions like multisystem inflammatory syndrome in children and other post-COVID-19 conditions are rare, they require careful management. Vaccination has proven effective in reducing severe disease and protecting against post-COVID-19 conditions. Continued surveillance, including wastewater monitoring and universal or pooled testing, remains crucial for controlling community spread. Key questions remain regarding the duration and quality of immunity after reinfection or vaccination, the impact of coinfections, and optimal treatment protocols for different pediatric populations.
Collapse
Affiliation(s)
- Juliane Wurm
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland; Department of Health Science and Medicine, University Lucerne, Lucerne, Switzerland
| | - Nicole Ritz
- Department of Health Science and Medicine, University Lucerne, Lucerne, Switzerland; Paediatric Infectious Diseases Unit, Department of Paediatrics, Children's Hospital, Cantonal Hospital Lucerne, Lucerne, Switzerland; Mycobacterial and Migrant Health Research, University Children's Hospital Basel and Department for Clinical Research, University of Basel, Basel, Switzerland
| | - Petra Zimmermann
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland; Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Department of Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
38
|
Hickey AJ, Greendyk R, Cummings MJ, Abrams D, O'Donnell MR, Rackley CR, Barbaro RP, Brodie D, Agerstrand C. Extracorporeal Membrane Oxygenation for COVID-19 During the Delta and Omicron Waves in North America. ASAIO J 2025; 71:325-331. [PMID: 39437129 DOI: 10.1097/mat.0000000000002334] [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: 10/25/2024] Open
Abstract
Clinical outcomes for patients with severe acute respiratory failure caused by different variants of the coronavirus disease 2019 (COVID-19) supported with extracorporeal membrane oxygenation (ECMO) are incompletely understood. Clinical characteristics, pre-ECMO management, and hospital mortality at 90 days for adults with COVID-19 who received venovenous ECMO (VV-ECMO) at North American centers during waves predominated by Delta (August 16 to December 12, 2021) and Omicron (January 31 to May 31, 2022) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants were compared in a competing risks framework. One thousand seven hundred and sixty-six patients (1,580 Delta, 186 Omicron) received VV-ECMO for COVID-19 during the Delta- and Omicron-predominant waves in North American centers. In the unadjusted competing risks model, no significant difference was observed in risk of hospital mortality at 90 days between patients during the Delta- versus Omicron-predominant wave (subhazard ratio [sHR], 0.94; 95% confidence interval [CI], 0.74-1.19), but patients supported with VV-ECMO during the Omicron-predominant wave had a significantly lower adjusted risk of hospital mortality at 90 days (subhazard ratio, 0.71; 95% CI, 0.51-0.99). Patients receiving VV-ECMO during the Omicron-predominant wave had a similar unadjusted risk of hospital mortality at 90 days, but a significantly lower adjusted risk of hospital mortality at 90 days than those receiving VV-ECMO during the Delta-predominant wave.
Collapse
Affiliation(s)
- Andrew J Hickey
- From the Division of Pulmonology and Sleep Medicine, Department of Medicine, Atrium Health Pulmonology and Sleep Medicine, Atrium Health, Charlotte, North Carolina
| | - Richard Greendyk
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Irving Medical Center, New York, New York
| | - Matthew J Cummings
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Irving Medical Center, New York, New York
| | - Darryl Abrams
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Irving Medical Center, New York, New York
| | - Max R O'Donnell
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Irving Medical Center, New York, New York
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York
| | - Craig R Rackley
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina
| | - Ryan P Barbaro
- Division of Pediatric Critical Care Medicine, University of Michigan, Ann Arbor, Michigan
| | - Daniel Brodie
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Cara Agerstrand
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
39
|
Hernandez G, Osinski A, Majumdar A, Eitson JL, Antczak M, Pawłowski K, Niederstrasser H, Servage KA, Posner B, Schoggins JW, Ready JM, Tagliabracci VS. Covalent inhibition of the SARS-CoV-2 NiRAN domain via an active-site cysteine. J Biol Chem 2025; 301:108378. [PMID: 40049411 PMCID: PMC12013494 DOI: 10.1016/j.jbc.2025.108378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/07/2025] [Accepted: 02/25/2025] [Indexed: 03/12/2025] Open
Abstract
The kinase-like nidovirus RdRp-associated nucleotidyl transferase (NiRAN) domain of nsp12 in SARS-CoV-2 catalyzes the formation of the 5' RNA cap structure. This activity is required for viral replication, offering a new target for the development of antivirals. Here, we develop a high-throughput assay to screen for small molecule inhibitors targeting the SARS-CoV-2 NiRAN domain. We identified NiRAN covalent inhibitor 2 (NCI-2), a compound with a reactive chloromethyl group that covalently binds to an active site cysteine (Cys53) in the NiRAN domain, inhibiting its activity. NCI-2 can enter cells, bind to, and inactivate ectopically expressed nsp12. A cryo-EM reconstruction of the SARS-CoV-2 replication-transcription complex bound to NCI-2 offers a detailed structural blueprint for rational drug design. Although NCI-2 showed limited potency against SARS-CoV-2 replication in cells, our work lays the groundwork for developing more potent and selective inhibitors targeting the NiRAN domain. This approach presents a promising therapeutic strategy for effectively combating COVID-19 and potentially mitigating future coronavirus outbreaks.
Collapse
Affiliation(s)
- Genaro Hernandez
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Adam Osinski
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Abir Majumdar
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jennifer L Eitson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Monika Antczak
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Krzysztof Pawłowski
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Kelly A Servage
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bruce Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John W Schoggins
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Vincent S Tagliabracci
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
40
|
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
|
41
|
Tomos I, Grigoropoulos I, Kosti C, Chrysikos S, Digalaki A, Thomas K, Hillas G, Kazakou P, Antoniadou A, Kavatha D, Dimakou K. Comparison of effectiveness and safety between baricitinib and tocilizumab in severe COVID-19: a retrospective study. Expert Rev Respir Med 2025; 19:389-397. [PMID: 40017107 DOI: 10.1080/17476348.2025.2473486] [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: 12/02/2024] [Revised: 02/16/2025] [Accepted: 02/25/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Immunomodulators tocilizumab and baricitinib have been used for the treatment of severe COVID-19, however, there are only few published studies comparing their efficacy. RESEARCH DESIGN AND METHODS All consecutive non-ICU hospitalized severe COVID-19 patients who received baricitinib or tocilizumab, were included retrospectively. Primary outcomes were mortality or intubation on day 14, time to oxygen therapy weaning and duration of hospitalization. Safety was measured as treatment-related adverse events. RESULTS 321 hospitalized patients with severe COVID-19 were included (mean age 62.4 years ± 14.7); 241 (75.1%) received baricitinib (mean age 64.2 years ± 15.2) and 80 (24.9%) tocilizumab (mean age 57.3 ± 11.7). Patients who received baricitinib presented significantly lower risk of mortality or intubation on day 14, compared to the tocilizumab group after adjusting for age, sex, vaccination, Charlson comorbidity index, body mass index, remdesivir administration and WHO ordinal scale at enrollment (OR: 0.42, 95% CI: 0.20-0.86). In the augmented inverse-probability weighting regression, the protective role of baricitinib remained statistically significant (OR: 0.76, 95% CI: 0.66-0.88). No difference in secondary bacterial infections was detected, but tocilizumab was associated with significant higher rate of liver injury (Odds Ratio, 95%CI, p < 0.001). CONCLUSIONS Our study suggests survival and safety are significantly better for baricitinib compared to tocilizumab in severe COVID-19. Clinical randomized trials are needed for confirmation.
Collapse
Affiliation(s)
- Ioannis Tomos
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Ioannis Grigoropoulos
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Chrysavgi Kosti
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Serafeim Chrysikos
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Antonia Digalaki
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Konstantinos Thomas
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Georgios Hillas
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| | - Pinelopi Kazakou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Anastasia Antoniadou
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Dimitra Kavatha
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens School of Medicine, Haidari, Greece
| | - Katerina Dimakou
- 5th Department of Respiratory Medicine, 'SOTIRIA' Chest Diseases Hospital of Athens, Athens, Greece
| |
Collapse
|
42
|
Cilloniz C, Videla AJ, Pulido L, Uy-King MJ. Viral community-acquired pneumonia: what's new since COVID-19 emerged? Expert Rev Respir Med 2025; 19:347-362. [PMID: 40077864 DOI: 10.1080/17476348.2025.2479611] [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: 01/17/2025] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 03/14/2025]
Abstract
INTRODUCTION All over the world, viral pneumonia has a significant impact on morbidity and mortality, especially among vulnerable populations. The most common respiratory viruses causing pneumonia include influenza virus, respiratory syncytial virus, adenoviruses and rhinovirus. The COVID-19 pandemic has changed the landscape of viral pneumonia and has reshaped our understanding of the role of viruses in this disease. We are now more aware of the importance of early diagnosis, the impact of co-infections, the effects of viral variants, and the long-term consequences of post-viral pneumonia. AREAS COVERED We discuss the latest scientific evidence regarding epidemiology, diagnosis, treatment, and prevention of viral pneumonia. This review summarizes findings from a PubMed search on respiratory viruses in community-acquired pneumonia. EXPERT OPINION Our experience during the COVID-19 pandemic has changed our perspective on respiratory viruses and their role in viral pneumonia. Diagnostic advances have been made, co-infections have received greater recognition, immune responses to viral infections are better understood, and approaches to treating viral pneumonia have expanded. Despite this progress, however, research on the impact of respiratory viruses on pneumonia must continue to pursue the development of new antivirals and vaccines, and investigate the long-term sequelae, especially in cases of severe viral pneumonia.
Collapse
Affiliation(s)
- Catia Cilloniz
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Health Sciences, Continental University, Huancayo, Peru
| | - Alejandro J Videla
- Pulmonology Department, University Austral Hospital, Austral University, Buenos Aires, Argentina
| | - Laura Pulido
- Pulmonology Department, Italian Hospital of Rosario, Rosario, Argentina
| | - Mary Joy Uy-King
- Chairman Medical Research and Training Committee, Healthway QualiMed Hospital, San Jose Del Monte, Philippines
| |
Collapse
|
43
|
Okoli A, Ogbuagu O. Drug interactions in people with HIV treated with antivirals for other viral illnesses. Expert Opin Drug Metab Toxicol 2025; 21:383-397. [PMID: 39836520 DOI: 10.1080/17425255.2025.2455401] [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: 01/11/2024] [Revised: 12/13/2024] [Accepted: 01/15/2025] [Indexed: 01/23/2025]
Abstract
INTRODUCTION Background: People with HIV (PWH) have benefited tremendously from effective antiretroviral(ARV) treatments. However, PWH are at increased risk for other viral infections transmitted in the same way as HIV (such as hepatitis C and MPox) or that are opportunistic (e.g. cytomegalovirus). These coinfections contribute significantly to morbidity and mortality among PWH and require effective treatments to optimize patient outcomes. However, their management is complicated by drug-drug interactions (DDIs) with ARVs. AREAS COVERED Metabolism pathways and DDIs between approved ARVs and selected antiviral agents used for the treatment of common and clinically relevant viral coinfections are discussed. Literature review included search of published papers, conference abstracts (IAS, CROI, IDWeek, EACS, Glasgow) as well as unpublished data from approved drug prescribing information and regulatory submissions sourced from PubMed, Google, and Google Scholar available between June 30 1981 through June 1, 2024. EXPERT OPINION Management of drug interactions is essential for maintaining efficacy and safety of ARV and other co-administered antiviral therapies. Longer acting agents are now available for treatment of HIV and this lengthens the period during which drug interactions may occur. Emerging novel nanoparticle-carrier targeted hepatitis C and HIV treatments may mitigate, if not eliminate, their propensity for drug-drug interactions.
Collapse
Affiliation(s)
- Adaora Okoli
- Department of Infectious Diseases, Marshfield Clinic Health System, Marshfield, Wisconsin, USA
| | - Onyema Ogbuagu
- Department of Infectious Diseases, Marshfield Clinic Health System, Marshfield, Wisconsin, USA
- Yale AIDS Program, New Haven, CT, USA
| |
Collapse
|
44
|
Safi D, Khouri F, Zareef R, Arabi M. Antivirals in COVID-19: A Focus on Pediatric Cardiac Patients. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2025; 2025:4573096. [PMID: 40196380 PMCID: PMC11972864 DOI: 10.1155/cjid/4573096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/17/2025] [Indexed: 04/09/2025]
Abstract
The COVID-19 pandemic created an unprecedented public health crisis, driven by its rapid global spread and the urgent need for worldwide collaborative interventions to contain it. This urgency spurred the search for therapeutic agents to prevent or manage the infection. Among these, various types of antivirals emerged as a prominent treatment option, supported by a wealth of observational studies and randomized controlled trials. The results from such studies conflict, with some concluding efficacy and others the lack thereof, with variability also occurring depending on the severity of COVID-19 in the studied population. In addition, many agents have been explored using randomized controlled trials-the gold standard in evaluating the efficacy of an intervention-to only a limited degree, with most of the evidence behind their use concluded using observational studies. Thus, the sheer volume of data has made it challenging to resolve inconsistencies and determine true efficacy. Furthermore, there is a paucity in the literature regarding the use of antivirals in the pediatric population infected with COVID-19, with their use being extrapolated from the results of studies done on adult patients. As such, additional trials are needed to solidify the effectiveness of antivirals in managing COVID-19, particularly in the underexplored and especially vulnerable pediatric cardiac patients. Therefore, utilizing the results from randomized controlled trials, this narrative review evaluates the rationale behind the use of antivirals, summarizes the findings from the literature, and concludes with a focused discussion on their application in pediatric cardiac patients.
Collapse
Affiliation(s)
- Dalia Safi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Farah Khouri
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rana Zareef
- Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mariam Arabi
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
45
|
Delgado C, Nogara PA, Miranda MD, Rosa AS, Ferreira VNS, Batista LT, Oliveira TKF, Omage FB, Motta F, Bastos IM, Orian L, Rocha JBT. In Silico and In Vitro Studies of the Approved Antibiotic Ceftaroline Fosamil and Its Metabolites as Inhibitors of SARS-CoV-2 Replication. Viruses 2025; 17:491. [PMID: 40284934 PMCID: PMC12031345 DOI: 10.3390/v17040491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/14/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
The SARS-CoV-2 proteases Mpro and PLpro are critical targets for antiviral drug development for the treatment of COVID-19. The 1,2,4-thiadiazole functional group is an inhibitor of cysteine proteases, such as papain and cathepsins. This chemical moiety is also present in ceftaroline fosamil (CF), an FDA-approved fifth-generation cephalosporin antibiotic. This study investigates the interactions between CF, its primary metabolites (M1 is dephosphorylated CF and M2 is an opened β-lactam ring) and derivatives (protonated M1H and M2H), and its open 1,2,4-thiadiazole rings derivatives (open-M1H and open-M2H) with SARS-CoV-2 proteases and evaluates CF's effects on in vitro viral replication. In silico analyses (molecular docking and molecular dynamics (MD) simulations) demonstrated that CF and its metabolites are potential inhibitors of PLpro and Mpro. Docking analysis indicated that the majority of the ligands were more stable with Mpro than PLpro; however, in vitro biochemical analysis indicated PLpro as the preferred target for CF. CF inhibited viral replication in the human Calu-3 cell model at submicromolar concentrations when added to cell culture medium at 12 h. Our results suggest that CF should be evaluated as a potential repurposing agent for COVID-19, considering not only viral proteases but also other viral targets and relevant cellular pathways. Additionally, the reactivity of sulfur in the 1,2,4-thiadiazole moiety warrants further exploration for the development of viral protease inhibitors.
Collapse
Affiliation(s)
- Cássia Delgado
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria 97000-000, RS, Brazil; (C.D.); (J.B.T.R.)
| | - Pablo Andrei Nogara
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria 97000-000, RS, Brazil; (C.D.); (J.B.T.R.)
- Instituto Federal de Educação, Ciência e Tecnologia Sul-rio-grandense (IFSul), Bagé 96400-000, RS, Brazil
| | - Milene Dias Miranda
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.S.R.); (V.N.S.F.); (L.T.B.); (T.K.F.O.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Alice Santos Rosa
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.S.R.); (V.N.S.F.); (L.T.B.); (T.K.F.O.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Vivian Neuza Santos Ferreira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.S.R.); (V.N.S.F.); (L.T.B.); (T.K.F.O.)
| | - Luisa Tozatto Batista
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.S.R.); (V.N.S.F.); (L.T.B.); (T.K.F.O.)
| | - Thamara Kelcya Fonseca Oliveira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.S.R.); (V.N.S.F.); (L.T.B.); (T.K.F.O.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Folorunsho Bright Omage
- Biological Chemistry Laboratory, Department of Organic Chemistry, Institute of Chemistry, University of Campinas (UNICAMP), Campinas 13000-000, SP, Brazil;
| | - Flávia Motta
- Laboratório de interface patógeno-hospedeiro, Departamento de Biologia Celular, Universidade de Brasília (UnB), Brasília 70910-900, DF, Brazil; (F.M.); (I.M.B.)
| | - Izabela Marques Bastos
- Laboratório de interface patógeno-hospedeiro, Departamento de Biologia Celular, Universidade de Brasília (UnB), Brasília 70910-900, DF, Brazil; (F.M.); (I.M.B.)
| | - Laura Orian
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, Via Marzolo 1, 35129 Padova, Italy;
| | - João Batista Teixeira Rocha
- Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria 97000-000, RS, Brazil; (C.D.); (J.B.T.R.)
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 90000-000, RS, Brazil
| |
Collapse
|
46
|
Ferková S, Lepage M, Désilets A, Assouvie K, Lemieux G, Brochu I, Froehlich U, Gravel-Trudeau A, Vastra J, Jean F, Sarret P, Leduc R, Boudreault PL. Optimizing the pharmacokinetics and selectivity of TMPRSS2 inhibitors. Eur J Med Chem 2025; 294:117579. [PMID: 40382841 DOI: 10.1016/j.ejmech.2025.117579] [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: 02/18/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 05/20/2025]
Abstract
Since 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has undergone significant genomic mutations, contributing to resistance against existing 2019 coronavirus disease (COVID-19) treatments. In a previous study, we identified N-0385, a potent host-directed inhibitor of transmembrane serine protease 2 (TMPRSS2), which has therapeutic efficacy towards SARS-CoV-2 infection. However, in further evaluation of its preclinical druggability, N-0385 displayed unfavorable pharmacokinetic properties, including high bioavailability (99 %) following intranasal (IN) administration. This can lead to substantial systemic exposure and potential adverse effects due to off-target interactions. Here, we designed a library of peptidomimetic compounds with P3 site modifications on an optimized scaffold. We sought to maintain sub-nanomolar potency against TMPRSS2 (Kis < 2 nM), reduce pseudovirus infection, while addressing the lack of selectivity and excessive lung uptake. Notably, inhibitor 9, which contains Asp at the P3 position, achieved a two-fold increase in TMPRSS2 inhibitory potency (Ki = 0.13 ± 0.03 nM), a >700-fold selectivity over Factor Xa (FXa), and showed superior selectivity against other proteases (matriptase, transmembrane serine protease 6 (TMPRSS6), thrombin, furin, and tPA). Despite concerns about the role of FXa in the coagulation cascade, compound 9 had no impact on coagulation or thrombolysis 2 h after in vitro treatment. In the air-liquid interface (ALI) model of the lung epithelium, compound 9 displayed a 1.5-fold decrease in permeability compared to N-0385 and demonstrated sustained stability in lungs (11 h) and plasma (13 h). Taken together, our data demonstrate that continued optimization of this type of inhibitors will lead to improved therapeutics for the treatment of SARS-CoV-2 infection by IN administration.
Collapse
Affiliation(s)
- Sára Ferková
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Matthieu Lepage
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Antoine Désilets
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Kevin Assouvie
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Gabriel Lemieux
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Isabelle Brochu
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Ulrike Froehlich
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Alice Gravel-Trudeau
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jules Vastra
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - François Jean
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Philippe Sarret
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Richard Leduc
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke (IPS), Université de Sherbrooke, Sherbrooke, Québec, Canada.
| |
Collapse
|
47
|
Flisiak R, Jaroszewicz J, Kozielewicz D, Kuchar E, Parczewski M, Pawłowska M, Piekarska A, Rzymski P, Simon K, Tomasiewicz K, Zarębska-Michaluk D. Management of SARS-CoV-2 Infection-Clinical Practice Guidelines of the Polish Association of Epidemiologists and Infectiologists, for 2025. J Clin Med 2025; 14:2305. [PMID: 40217755 PMCID: PMC11989246 DOI: 10.3390/jcm14072305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/17/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
The first Polish recommendations for the management of COVID-19 were published by the Polish Society of Epidemiologists and Infectiologists (PTEiLChZ) on 31 March 2020, and the last three years ago. The emergence of new SARS-CoV-2 variants, a different course of the disease, as well as new knowledge about therapies and vaccines, requires updating diagnostic, therapeutic, and prophylactic guidelines. Despite the reduction in the threat associated with COVID-19, there is a risk of another epidemic caused by coronaviruses, which was an additional reason for developing a new version of the guidelines. In preparing these recommendations, the Delphi method was used, reaching a consensus after three survey cycles. Compared to the 2022 version, the names of the individual stages of the disease have been changed, adapting them to the realities of clinical practice, and attention was paid to the differences observed in immunosuppressed patients and in children. Some previously recommended drugs have been discontinued, including monoclonal antibodies. In addition, general principles of vaccination were presented, as well as issues related to the post-COVID syndrome.
Collapse
Affiliation(s)
- Robert Flisiak
- Department of Infectious Diseases and Hepatology, Medical University in Białystok, 15-540 Białystok, Poland
| | - Jerzy Jaroszewicz
- Department of Infectious Diseases and Hepatology, Medical University of Silesia, 40-635 Katowice, Poland;
| | - Dorota Kozielewicz
- Department of Infectious Diseases and Hepatology, Faculty of Medicine, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University, 87-100 Toruń, Poland; (D.K.); (M.P.)
| | - Ernest Kuchar
- Pediatric and Observation Department, Medical University of Warsaw, 02-091 Warszawa, Poland;
| | - Miłosz Parczewski
- Department of Infectious and Tropical Diseases and Acquired Immunodeficiency, Pomeranian Medical University, 70-204 Szczecin, Poland;
| | - Małgorzata Pawłowska
- Department of Infectious Diseases and Hepatology, Faculty of Medicine, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University, 87-100 Toruń, Poland; (D.K.); (M.P.)
| | - Anna Piekarska
- Department of Infectious Diseases and Hepatology, Medical University of Łódź, 90–419 Łódź, Poland;
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznań University of Medical Sciences, 60-806 Poznań, Poland;
| | - Krzysztof Simon
- Department of Infectious Diseases and Hepatology, Medical University of Wrocław, 51-149 Wrocław, Poland;
| | - Krzysztof Tomasiewicz
- Department of Infectious Diseases and Hepatology, Medical University of Lublin, 20-081 Lublin, Poland;
| | - Dorota Zarębska-Michaluk
- Department of Infectious Diseases and Allergology, Jan Kochanowski University, 25-317 Kielce, Poland;
| |
Collapse
|
48
|
Omange RW, Kim SC, Kolhatkar NS, Plott T, Van Trump W, Zhang K, O’Donnell H, Chen D, Hosny A, Wiest M, Barry Z, Addiego EC, Mengistu M, Odorizzi PM, Cai Y, Jacobson R, Wallin JJ. AI discovery of TLR agonist-driven phenotypes reveals unique features of peripheral cells from healthy donors and ART-suppressed people living with HIV. Front Immunol 2025; 16:1541152. [PMID: 40201178 PMCID: PMC11975909 DOI: 10.3389/fimmu.2025.1541152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Abstract
Background Selective and potent Toll-like receptor (TLR) agonists are currently under evaluation in preclinical models and clinical studies to understand how the innate immune system can be harnessed for therapeutic potential. These molecules are designed to modulate innate and adaptive immune responses, making them promising therapeutic candidates for treating diseases such as cancer or chronic viral infections. Much is known about the expression and signaling of TLRs which varies based on cell type, cellular localization, and tissue distribution. However, the downstream effects of different TLR agonists on cellular populations and phenotypes are not well understood. This study aimed to investigate the impact of TLR pathway stimulation on peripheral blood mononuclear cell (PBMC) cultures from people living with HIV (PLWH) and healthy donors. Methods The effects of TLR4, TLR7, TLR7/8, TLR8 and TLR9 agonists were evaluated on cytokine production, cell population frequencies, and morphological characteristics of PBMC cultures over time. Changes in the proportions of different cell populations in blood and morphological features were assessed using high-content imaging and analyzed using an AI-driven approach. Results TLR4 and TLR8 agonists promoted a compositional shift and accumulation of small round (lymphocyte-like) PBMCs, whereas TLR9 agonists led to an accumulation of large round (myeloid-like) PBMCs. A related increase was observed in markers of cell death, most prominently with TLR4 and TLR8 agonists. All TLR agonists were shown to promote some features associated with cellular migration. Furthermore, a comparison of TLR agonist responses in healthy and HIV-positive PBMCs revealed pronounced differences in cytokine/chemokine responses and morphological cellular features. Most notably, higher actin contraction and nuclear fragmentation was observed in response to TLR4, TLR7, TLR7/8 and TLR9 agonists for antiretroviral therapy (ART)-suppressed PLWH versus healthy PBMCs. Conclusions These data suggest that machine learning, combined with cell imaging and cytokine quantification, can be used to better understand the cytological and soluble immune responses following treatments with immunomodulatory agents in vitro. In addition, comparisons of these responses between disease states are possible with the appropriate patient samples.
Collapse
Affiliation(s)
- Robert Were Omange
- Biomarker Sciences and Diagnostics, Gilead Sciences, Inc., Foster City, CA, United States
| | - Samuel C. Kim
- Biomarker Sciences and Diagnostics, Gilead Sciences, Inc., Foster City, CA, United States
| | - Nikita S. Kolhatkar
- Biomarker Sciences and Diagnostics, Gilead Sciences, Inc., Foster City, CA, United States
| | | | | | | | | | - Daniel Chen
- Spring Science, San Carlos, CA, United States
| | - Ahmed Hosny
- Spring Science, San Carlos, CA, United States
| | | | - Zach Barry
- Spring Science, San Carlos, CA, United States
| | | | - Meron Mengistu
- Biomarker Sciences and Diagnostics, Gilead Sciences, Inc., Foster City, CA, United States
| | - Pamela M. Odorizzi
- Biomarker Sciences and Diagnostics, Gilead Sciences, Inc., Foster City, CA, United States
| | - Yanhui Cai
- Biomarker Sciences and Diagnostics, Gilead Sciences, Inc., Foster City, CA, United States
| | | | - Jeffrey J. Wallin
- Biomarker Sciences and Diagnostics, Gilead Sciences, Inc., Foster City, CA, United States
| |
Collapse
|
49
|
Wong YC, Ng CJ, Huang YB, Chen SY. Effectiveness and Safety of Remdesivir for the Treatment of COVID-19 Patients with Liver Cirrhosis: A Retrospective Cohort Study. Life (Basel) 2025; 15:512. [PMID: 40283067 PMCID: PMC12028772 DOI: 10.3390/life15040512] [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: 02/07/2025] [Revised: 03/16/2025] [Accepted: 03/19/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Patients with liver cirrhosis are at an increased risk of mortality from coronavirus disease 2019 (COVID-19). Remdesivir, an adenosine analog, exhibits activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and is thus recommended for inpatients with COVID-19. This study evaluated the effectiveness and safety of remdesivir in patients with COVID-19 and liver cirrhosis. METHODS This retrospective study was conducted using data from Taiwan's largest healthcare system. The study cohort comprised adult patients with COVID-19 and liver cirrhosis who visited our emergency department between April 2021 and September 2022. Remdesivir's adverse effects, including bradycardia, anemia, unstable glucose levels, and abnormal liver function test results, were recorded. Treatment outcomes were assessed in terms of hospitalization duration, mortality, intubation, and intensive care unit admission. RESULTS This study included 1368 patients with COVID-19 and liver cirrhosis, of whom 46 received remdesivir. Remdesivir recipients were older (66.5 vs. 62 years; p = 0.042) and had a higher rate of oxygen therapy use (56.52% vs. 32.22%; p = 0.001) than nonrecipients. Common adverse effects of remdesivir included lower heart rates (83 vs. 96 bpm; p < 0.001) and decreased hemoglobin levels (9.5 vs. 10.2 g/dL; p = 0.003) without fatal consequences. No statistically significant difference between remdesivir recipients and nonrecipients in hospitalization duration, intubation rates, or mortality rates was found. CONCLUSIONS Remdesivir is safe for treating COVID-19 in patients with liver cirrhosis. Although remdesivir recipients exhibited trends toward improved outcomes in our study, large-scale studies are required to confirm its efficacy in this population.
Collapse
Affiliation(s)
- Yi-Ching Wong
- Department of Emergency Medicine, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.W.); (C.-J.N.); (Y.-B.H.)
| | - Chip-Jin Ng
- Department of Emergency Medicine, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.W.); (C.-J.N.); (Y.-B.H.)
| | - Yan-Bo Huang
- Department of Emergency Medicine, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.W.); (C.-J.N.); (Y.-B.H.)
| | - Shou-Yen Chen
- Department of Emergency Medicine, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (Y.-C.W.); (C.-J.N.); (Y.-B.H.)
- Graduate Institute of Management, College of Management, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
50
|
Imbiakha B, Ezzatpour S, Buchholz DW, Poosala S, Vadloori B, Sonawane A, Gannabathula S, V V S R, Naik NN, Shanmugasundaram S, Lokireddy SR, Kunchala SR, Thummadi C, Godavarty P, Meher K, Vangala S, Saxena U, Aguilar HC, August A, Sahler J. Novel nanobody drug conjugate as a prophylactic or therapeutic against SARS-CoV-2 infection in mice. Biochem Biophys Res Commun 2025; 753:151480. [PMID: 39970838 DOI: 10.1016/j.bbrc.2025.151480] [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: 02/03/2025] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
The COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, resulted in global health and economic crisis at an exceptional level. The high transmissibility of SARS-CoV-2, a lack of population immunity, and the prevalence of severe clinical outcomes created a need for the rapid development of effective therapeutic countermeasures. Sybodies, or synthetic nanobodies, are a novel and unique class of synthetic antigen-binding fragments ideal for large-scale production. We created a neutralizing sybody directed against the receptor-binding domain (RBD) epitope of the Spike protein of SARS-CoV-2 and conjugated it to remdesivir to create nanobody-drug-conjugate (NDC). We used a mouse model of infection to determine the capacity of the NDC to reduce disease severity and mortality. K18-hACE2 mice treated with NDC prior to, simultaneously with, and/or post-SARS-CoV-2 infection had reduced weight loss, mortality, lung pathology, and viral RNA in their lungs. This protection was found to be dependent on the protein structure of the sybody. Delivery of these novel therapeutic NDCs through nasal inhalation using a nebulizer could offer a convenient method for patients to self-administer treatments or as a prophylaxis. This platform therapy could apply to new variants of the ongoing SARS-CoV-2 epidemics, or to other pathogens of future pandemic potential.
Collapse
Affiliation(s)
- Brian Imbiakha
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Shahrzad Ezzatpour
- Department of Microbiology, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, 14853, USA
| | - David W Buchholz
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | | | | | | | | | - Ramya V V S
- Oncoseekbio Pvt Ltd, Bionest, Hyderabad, India
| | | | | | - Sudarsana Reddy Lokireddy
- Oncosimis Biotech Pvt Ltd, Plot No 3, IDA Uppal, Genpact Rd, Habsiguda, Hyderabad, Telangana, 500007, India
| | - Sridhar Rao Kunchala
- Oncosimis Biotech Pvt Ltd, Plot No 3, IDA Uppal, Genpact Rd, Habsiguda, Hyderabad, Telangana, 500007, India
| | - Chennakesavulu Thummadi
- Oncosimis Biotech Pvt Ltd, Plot No 3, IDA Uppal, Genpact Rd, Habsiguda, Hyderabad, Telangana, 500007, India
| | - Pratyusha Godavarty
- Oncosimis Biotech Pvt Ltd, Plot No 3, IDA Uppal, Genpact Rd, Habsiguda, Hyderabad, Telangana, 500007, India
| | - Kranti Meher
- Reagene Innovations Pvt Ltd, Bionest, Hyderabad, India
| | | | - Uday Saxena
- Reagene Innovations Pvt Ltd, Bionest, Hyderabad, India
| | - Hector C Aguilar
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| | - Julie Sahler
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|