1
|
Korula P, Alexander H, John JS, Kirubakaran R, Singh B, Tharyan P, Rupali P. Favipiravir for treating COVID-19. Cochrane Database Syst Rev 2024; 2:CD015219. [PMID: 38314855 PMCID: PMC10840071 DOI: 10.1002/14651858.cd015219.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to challenge the health workforce and societies worldwide. Favipiravir was suggested by some experts to be effective and safe to use in COVID-19. Although this drug has been evaluated in randomized controlled trials (RCTs), it is still unclear if it has a definite role in the treatment of COVID-19. OBJECTIVES To assess the effects of favipiravir compared to no treatment, supportive treatment, or other experimental antiviral treatment in people with acute COVID-19. SEARCH METHODS We searched the Cochrane COVID-19 Study Register, MEDLINE, Embase, the World Health Organization (WHO) COVID-19 Global literature on coronavirus disease, and three other databases, up to 18 July 2023. SELECTION CRITERIA We searched for RCTs evaluating the efficacy of favipiravir in treating people with COVID-19. DATA COLLECTION AND ANALYSIS We used standard Cochrane methodological procedures for data collection and analysis. We used the GRADE approach to assess the certainty of evidence for each outcome. MAIN RESULTS We included 25 trials that randomized 5750 adults (most under 60 years of age). The trials were conducted in Bahrain, Brazil, China, India, Iran, Kuwait, Malaysia, Mexico, Russia, Saudi Arabia, Thailand, the UK, and the USA. Most participants were hospitalized with mild to moderate disease (89%). Twenty-two of the 25 trials investigated the role of favipiravir compared to placebo or standard of care, whilst lopinavir/ritonavir was the comparator in two trials, and umifenovir in one trial. Most trials (24 of 25) initiated favipiravir at 1600 mg or 1800 mg twice daily for the first day, followed by 600 mg to 800 mg twice a day. The duration of treatment varied from five to 14 days. We do not know whether favipiravir reduces all-cause mortality at 28 to 30 days, or in-hospital (risk ratio (RR) 0.84, 95% confidence interval (CI) 0.49 to 1.46; 11 trials, 3459 participants; very low-certainty evidence). We do not know if favipiravir reduces the progression to invasive mechanical ventilation (RR 0.86, 95% CI 0.68 to 1.09; 8 trials, 1383 participants; very low-certainty evidence). Favipiravir may make little to no difference in the need for admission to hospital (if ambulatory) (RR 1.04, 95% CI 0.44 to 2.46; 4 trials, 670 participants; low-certainty evidence). We do not know if favipiravir reduces the time to clinical improvement (defined as time to a 2-point reduction in patients' admission status on the WHO's ordinal scale) (hazard ratio (HR) 1.13, 95% CI 0.69 to 1.83; 4 trials, 721 participants; very low-certainty evidence). Favipiravir may make little to no difference to the progression to oxygen therapy (RR 1.20, 95% CI 0.83 to 1.75; 2 trials, 543 participants; low-certainty evidence). Favipiravir may lead to an overall increased incidence of adverse events (RR 1.27, 95% CI 1.05 to 1.54; 18 trials, 4699 participants; low-certainty evidence), but may result in little to no difference inserious adverse eventsattributable to the drug (RR 1.04, 95% CI 0.76 to 1.42; 12 trials, 3317 participants; low-certainty evidence). AUTHORS' CONCLUSIONS The low- to very low-certainty evidence means that we do not know whether favipiravir is efficacious in people with COVID-19 illness, irrespective of severity or admission status. Treatment with favipiravir may result in an overall increase in the incidence of adverse events but may not result in serious adverse events.
Collapse
Affiliation(s)
- Pritish Korula
- Division of Critical Care Medicine, Christian Medical College, Vellore, India
| | - Hanna Alexander
- Department of Infectious Diseases, Christian Medical College, Vellore, India
| | - Jisha Sara John
- Department of Infectious Diseases, Christian Medical College, Vellore, India
| | - Richard Kirubakaran
- Department of Infectious Diseases, Christian Medical College, Vellore, India
| | - Bhagteshwar Singh
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Prathap Tharyan
- Clinical Epidemiology Unit, Prof. BV Moses Centre for Evidence-Informed Healthcare and Health Policy, Christian Medical College, Vellore, India
| | - Priscilla Rupali
- Department of Infectious Diseases, Christian Medical College, Vellore, India
| |
Collapse
|
2
|
Kreins AY, Roux E, Pang J, Cheng I, Charles O, Roy S, Mohammed R, Owens S, Lowe DM, Brugha R, Williams R, Howley E, Best T, Davies EG, Worth A, Solas C, Standing JF, Goldstein RA, Rocha-Pereira J, Breuer J. Favipiravir induces HuNoV viral mutagenesis and infectivity loss with clinical improvement in immunocompromised patients. Clin Immunol 2024; 259:109901. [PMID: 38218209 DOI: 10.1016/j.clim.2024.109901] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/15/2024]
Abstract
Chronic human norovirus (HuNoV) infections in immunocompromised patients result in severe disease, yet approved antivirals are lacking. RNA-dependent RNA polymerase (RdRp) inhibitors inducing viral mutagenesis display broad-spectrum in vitro antiviral activity, but clinical efficacy in HuNoV infections is anecdotal and the potential emergence of drug-resistant variants is concerning. Upon favipiravir (and nitazoxanide) treatment of four immunocompromised patients with life-threatening HuNoV infections, viral whole-genome sequencing showed accumulation of favipiravir-induced mutations which coincided with clinical improvement although treatment failed to clear HuNoV. Infection of zebrafish larvae demonstrated drug-associated loss of viral infectivity and favipiravir treatment showed efficacy despite occurrence of RdRp variants potentially causing favipiravir resistance. This indicates that within-host resistance evolution did not reverse loss of viral fitness caused by genome-wide accumulation of sequence changes. This off-label approach supports the use of mutagenic antivirals for treating prolonged RNA viral infections and further informs the debate surrounding their impact on virus evolution.
Collapse
Affiliation(s)
- Alexandra Y Kreins
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Department of Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Emma Roux
- KU Leuven - Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Juanita Pang
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Iek Cheng
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Department of Pharmacy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Oscar Charles
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Sunando Roy
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Reem Mohammed
- Department of Pediatrics, Division of Allergy and Immunology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Stephen Owens
- Department of Paediatric Allergy, Immunology and Infectious Diseases, The Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle, United Kingdom
| | - David M Lowe
- Immunology Department, Royal Free Hospital NHS Foundation Trust, London, United Kingdom; Institute of Immunity and Transplantation, University College London, London, UK
| | - Rossa Brugha
- Department of Cardiothoracic Transplantation, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Rachel Williams
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Evey Howley
- Department of Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Timothy Best
- Department of Microbiology, Virology and Infection Control, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - E Graham Davies
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Department of Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Austen Worth
- Department of Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Caroline Solas
- Unité des Virus Émergents IRD 190, INSERM 1207, Aix-Marseille Université, Marseille, France; APHM, Laboratoire de Pharmacocinétique et Toxicologie, Hôpital La Timone, Marseille, France
| | - Joseph F Standing
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Department of Pharmacy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Richard A Goldstein
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Joana Rocha-Pereira
- KU Leuven - Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium.
| | - Judith Breuer
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom; Institute of Immunity and Transplantation, University College London, London, UK.
| |
Collapse
|
3
|
Darres A, Del Bello A, Marion O, de Lamballerie X, Malvy D, Izopet J, Kamar N. Failure of favipiravir to treat chronic norovirus infection in a kidney-transplant patient. Transpl Infect Dis 2024; 26:e14235. [PMID: 38180288 DOI: 10.1111/tid.14235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024]
Affiliation(s)
- Amandine Darres
- Department of Nephrology and Organ Transplantation, Toulouse University Hospital, Toulouse, France
- Department of Vascular Biology, Institute of Metabolic and Cardiovascular Diseases (I2MC), Toulouse, France
| | - Arnaud Del Bello
- Department of Nephrology and Organ Transplantation, Toulouse University Hospital, Toulouse, France
- Department of Vascular Biology, Institute of Metabolic and Cardiovascular Diseases (I2MC), Toulouse, France
| | - Olivier Marion
- Department of Nephrology and Organ Transplantation, Toulouse University Hospital, Toulouse, France
- INSERM UMR1291, CNRS UMR5051, Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Toulouse, France
| | - Xavier de Lamballerie
- Unité des Virus Émergents, INSERM-1207, IRD-190, Aix-Marseille University, Marseille, France
| | - Denis Malvy
- Service des maladies infectieuses et tropicales, CHU de Bordeaux, Bordeaux, France
- Inserm UMR 1219, IRD EMR 271, Bordeaux Population Health, Université de Bordeaux, Bordeaux, France
| | - Jacques Izopet
- INSERM UMR1291, CNRS UMR5051, Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Toulouse, France
- Department of Virology, Toulouse University Hospital, Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| | - Nassim Kamar
- Department of Nephrology and Organ Transplantation, Toulouse University Hospital, Toulouse, France
- INSERM UMR1291, CNRS UMR5051, Université Toulouse III, Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Toulouse, France
- Université Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
4
|
Illingworth CJR, Guerra-Assuncao JA, Gregg S, Charles O, Pang J, Roy S, Abdelnabi R, Neyts J, Breuer J. Genetic consequences of effective and suboptimal dosing with mutagenic drugs in a hamster model of SARS-CoV-2 infection. Virus Evol 2024; 10:veae001. [PMID: 38486802 PMCID: PMC10939363 DOI: 10.1093/ve/veae001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 11/23/2023] [Accepted: 01/03/2024] [Indexed: 03/17/2024] Open
Abstract
Mutagenic antiviral drugs have shown promise against multiple viruses, but concerns have been raised about whether their use might promote the emergence of new and harmful viral variants. Recently, genetic signatures associated with molnupiravir use have been identified in the global SARS-COV-2 population. Here, we examine the consequences of using favipiravir and molnupiravir to treat SARS-CoV-2 infection in a hamster model, comparing viral genome sequence data collected from (1) untreated hamsters, and (2) from hamsters receiving effective and suboptimal doses of treatment. We identify a broadly linear relationship between drug dose and the extent of variation in treated viral populations, with a high proportion of this variation being composed of variants at frequencies of less than 1 per cent, below typical thresholds for variant calling. Treatment with an effective dose of antiviral drug was associated with a gain of between 7 and 10 variants per viral genome relative to drug-free controls: even after a short period of treatment a population founded by a transmitted virus could contain multiple sequence differences to that of the original host. Treatment with a suboptimal dose of drug showed intermediate gains of variants. No dose-dependent signal was identified in the numbers of single-nucleotide variants reaching frequencies in excess of 5 per cent. We did not find evidence to support the emergence of drug resistance or of novel immune phenotypes. Our study suggests that where onward transmission occurs, a short period of treatment with mutagenic drugs may be sufficient to generate a significant increase in the number of viral variants transmitted.
Collapse
Affiliation(s)
| | - Jose A Guerra-Assuncao
- Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Infection, Immunity and Inflammation Research and Teaching Department, University College London, Gower Street, London WC1E 6BT, UK
| | - Samuel Gregg
- Infection, Immunity and Inflammation Research and Teaching Department, University College London, Gower Street, London WC1E 6BT, UK
| | - Oscar Charles
- Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Infection, Immunity and Inflammation Research and Teaching Department, University College London, Gower Street, London WC1E 6BT, UK
| | - Juanita Pang
- Infection, Immunity and Inflammation Research and Teaching Department, University College London, Gower Street, London WC1E 6BT, UK
| | - Sunando Roy
- Infection, Immunity and Inflammation Research and Teaching Department, University College London, Gower Street, London WC1E 6BT, UK
| | - Rana Abdelnabi
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, Leuven B-3000, Belgium
- The VirusBank Platform, Gaston Geenslaan, Leuven B-3000, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, Leuven B-3000, Belgium
- The VirusBank Platform, Gaston Geenslaan, Leuven B-3000, Belgium
| | - Judith Breuer
- Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street, London WC1N 3JH, UK
- Infection, Immunity and Inflammation Research and Teaching Department, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
5
|
Chaimongkol N, Dábilla N, Tohma K, Matsushima Y, Yardley AB, Levenson EA, Johnson JA, Ahorrio C, Oler AJ, Kim DY, Souza M, Sosnovtsev SV, Parra GI, Green KY. Norovirus evolves as one or more distinct clonal populations in immunocompromised hosts. mBio 2023; 14:e0217723. [PMID: 37905910 PMCID: PMC10746188 DOI: 10.1128/mbio.02177-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 11/02/2023] Open
Abstract
IMPORTANCE Noroviruses are an important cause of chronic diarrhea in patients with compromised immune systems. Presently, there are no effective therapies to clear the virus, which can persist for years in the intestinal tract. The goal of our study was to develop a better understanding of the norovirus strains that are associated with these long-term infections. With the remarkable diversity of norovirus strains detected in the immunocompromised patient cohort we studied, it appears that most, if not all, noroviruses circulating in nature may have the capacity to establish a chronic infection when a person is unable to mount an effective immune response. Our work is the most comprehensive genetic data set generated to date in which near full-length genomes from noroviruses associated with chronic infection were analyzed by high-resolution next-generation sequencing. Analysis of this data set led to our discovery that certain patients in our cohort were shedding noroviruses that could be subdivided into distinct haplotypes or populations of viruses that were co-evolving independently. The ability to track haplotypes of noroviruses during chronic infection will allow us to fine-tune our understanding of how the virus adapts and maintains itself in the human host, and how selective pressures such as antiviral drugs can affect these distinct populations.
Collapse
Affiliation(s)
- Natthawan Chaimongkol
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Nathânia Dábilla
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Laboratory of Virology and Cell Culture, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Kentaro Tohma
- Division of Viral Products, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yuki Matsushima
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Allison Behrle Yardley
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Eric A. Levenson
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jordan A. Johnson
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Courtney Ahorrio
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew J. Oler
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel Y. Kim
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Menira Souza
- Laboratory of Virology and Cell Culture, Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, Goiás, Brazil
| | - Stanislav V. Sosnovtsev
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Gabriel I. Parra
- Division of Viral Products, Food and Drug Administration, Silver Spring, Maryland, USA
| | - Kim Y. Green
- Caliciviruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
6
|
Reddy NA, Raj K, Lashkari HP. Norovirus Infection in Children with Acute Lymphoblastic Leukemia. Infect Chemother 2023; 55:403-407. [PMID: 37794579 PMCID: PMC10551709 DOI: 10.3947/ic.2023.0058] [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: 05/26/2023] [Accepted: 08/28/2023] [Indexed: 10/06/2023] Open
Abstract
Norovirus infection in children on treatment for acute lymphoblastic leukemia can lead to severe morbidity due to chronic viral shedding, malabsorption, failure to thrive, and interruption of chemotherapy. We had four children with norovirus diarrhoea in eight years period in our pediatric oncology unit. Three children under two years of age had chronic noroviral shedding and persistent diarrhoea, probably due to poor adaptive immune responses. Two of those children didn't respond to nitazoxanide and succumbed to the illness. The third patient who received nitazoxanide and favipiravir, is currently well on chemotherapy.
Collapse
Affiliation(s)
- Nalla Anuraag Reddy
- Department of Pediatric Hematology and Oncology, KMC Hospital, Ambedkar circle, Mangalore, India
| | - Keerthi Raj
- Department of Pediatrics, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Harsha Prasada Lashkari
- Department of Pediatrics, Kasturba Medical College, Manipal Academy of Higher Education (MAHE), Manipal, India.
| |
Collapse
|
7
|
Bekegnran CP, Driouich JS, Breuer J, Barthelemy K, Giocanti M, de Lamballerie X, Kreins AY, Nougairede A, Solas C. Simultaneous quantitation of favipiravir and its hydroxide metabolite in human plasma and hamster matrices using a UPLC-MS/MS method. Biomed Chromatogr 2023; 37:e5689. [PMID: 37349975 DOI: 10.1002/bmc.5689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 05/19/2023] [Accepted: 05/31/2023] [Indexed: 06/24/2023]
Abstract
Favipiravir, a broad-spectrum RNA-dependent RNA polymerase inhibitor, is currently being evaluated in preclinical and clinical studies for the treatment of various infectious diseases including COVID-19. We developed an ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) assay for the quantification of favipiravir and its hydroxide metabolite (M1), in human and hamster biological matrices. Analytes were separated on an Acquity UPLC HSS T3 column (2.1 × 100 mm, 1.8 μm) after a simple protein precipitation with acetonitrile. The mobile phase consisted of water and methanol, each containing 0.05% formic acid. Experiments were performed using electrospray ionization in the positive and negative ion mode, with protonated molecules used as the precursor ion and a total run time of 6 min. The MS/MS response was linear over the concentration ranges from 0.5-100 μg/ml for favipiravir and 0.25-30 μg/ml for M1. Intra- and inter-day accuracy and precision were within the recommended limits of the European Medicines Agency guidelines. No significant matrix effect was observed, and the method was successfully applied to inform favipiravir dose adjustments in six immunocompromised children with severe RNA viral infections. In conclusion, the UPLC-MS/MS assay is suitable for quantification of favipiravir over a wide range of dosing regimens, and can easily be adapted to other matrices and species.
Collapse
Affiliation(s)
- Cesar P Bekegnran
- APHM, Laboratoire de Pharmacocinétique et Toxicologie, Hôpital La Timone, Marseille, France
- Unité des Virus Émergents (UVE: Aix-Marseille University-IRD 190-Inserm 1207), Marseille, France
| | - Jean-Selim Driouich
- Unité des Virus Émergents (UVE: Aix-Marseille University-IRD 190-Inserm 1207), Marseille, France
| | - Judith Breuer
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Karine Barthelemy
- Unité des Virus Émergents (UVE: Aix-Marseille University-IRD 190-Inserm 1207), Marseille, France
| | - Madeleine Giocanti
- APHM, Laboratoire de Pharmacocinétique et Toxicologie, Hôpital La Timone, Marseille, France
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE: Aix-Marseille University-IRD 190-Inserm 1207), Marseille, France
| | - Alexandra Y Kreins
- UCL Great Ormond Street Institute of Child Health, London, UK
- Department of Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Trust Foundation, London, UK
| | - Antoine Nougairede
- Unité des Virus Émergents (UVE: Aix-Marseille University-IRD 190-Inserm 1207), Marseille, France
| | - Caroline Solas
- APHM, Laboratoire de Pharmacocinétique et Toxicologie, Hôpital La Timone, Marseille, France
- Unité des Virus Émergents (UVE: Aix-Marseille University-IRD 190-Inserm 1207), Marseille, France
| |
Collapse
|
8
|
Chakraborty C, Bhattacharya M, Saha A, Alshammari A, Alharbi M, Saikumar G, Pal S, Dhama K, Lee SS. Revealing the structural and molecular interaction landscape of the favipiravir-RTP and SARS-CoV-2 RdRp complex through integrative bioinformatics: Insights for developing potent drugs targeting SARS-CoV-2 and other viruses. J Infect Public Health 2023; 16:1048-1056. [PMID: 37196368 DOI: 10.1016/j.jiph.2023.05.010] [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: 02/01/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND The global research community has made considerable progress in therapeutic and vaccine research during the COVID-19 pandemic. Several therapeutics have been repurposed for the treatment of COVID-19. One such compound is, favipiravir, which was approved for the treatment of influenza viruses, including drug-resistant influenza. Despite the limited information on its molecular activity, clinical trials have attempted to determine the effectiveness of favipiravir in patients with mild to moderate COVID-19. Here, we report the structural and molecular interaction landscape of the macromolecular complex of favipiravir-RTP and SARS-CoV-2 RdRp with the RNA chain. METHODS Integrative bioinformatics was used to reveal the structural and molecular interaction landscapes of two macromolecular complexes retrieved from RCSB PDB. RESULTS We analyzed the interactive residues, H-bonds, and interaction interfaces to evaluate the structural and molecular interaction landscapes of the two macromolecular complexes. We found seven and six H-bonds in the first and second interaction landscapes, respectively. The maximum bond length is 3.79 Å. In the hydrophobic interactions, five residues (Asp618, Asp760, Thr687, Asp623, and Val557) were associated with the first complex and two residues (Lys73 and Tyr217) were associated with the second complex. The mobilities, collective motion, and B-factor of the two macromolecular complexes were analyzed. Finally, we developed different models, including trees, clusters, and heat maps of antiviral molecules, to evaluate the therapeutic status of favipiravir as an antiviral drug. CONCLUSIONS The results revealed the structural and molecular interaction landscape of the binding mode of favipiravir with the nsp7-nsp8-nsp12-RNA SARS-CoV-2 RdRp complex. Our findings can help future researchers in understanding the mechanism underlying viral action and guide the design of nucleotide analogs that mimic favipiravir and exhibit greater potency as antiviral drugs against SARS-CoV-2 and other infectious viruses. Thus, our work can help in preparing for future epidemics and pandemics.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India.
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore 756020, Odisha, India
| | - Abinit Saha
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - G Saikumar
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Soumen Pal
- School of Mechanical Engineering, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopaedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Republic of Korea
| |
Collapse
|
9
|
Chadwick PR, Trainor E, Marsden GL, Mills S, Chadwick C, O'Brien SJ, Evans CM, Mullender C, Strazds P, Turner S, Weston V, Toleman MS, de Barros C, Kontkowski G, Bak A. Guidelines for the management of norovirus outbreaks in acute and community health and social care settings. J Hosp Infect 2023:S0195-6701(23)00043-9. [PMID: 36796728 DOI: 10.1016/j.jhin.2023.01.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/19/2023] [Accepted: 01/22/2023] [Indexed: 02/17/2023]
Affiliation(s)
| | - Eamonn Trainor
- Northern Care Alliance NHS Foundation Trust, Greater Manchester, UK.
| | - Gemma L Marsden
- Healthcare Infection Society, London, UK; Royal College of General Practitioners, London, UK
| | - Samuel Mills
- British Infection Association, Seafield, West Lothian, UK; Oxford University NHS Foundation Trust, Oxford, UK
| | | | | | - Cariad M Evans
- Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | | | - Pixy Strazds
- Infection Prevention Society, London, UK; St Andrew's Healthcare, Northampton, UK
| | - Sarah Turner
- Infection Prevention Society, London, UK; Stockport Council, Stockport, UK
| | - Valya Weston
- Healthcare Infection Society, London, UK; Infection Prevention Society, London, UK; NHS England, London, UK
| | - Michelle S Toleman
- Healthcare Infection Society, London, UK; Cambridge University Hospitals NHS Trust, Cambridge, UK
| | | | | | - Aggie Bak
- Healthcare Infection Society, London, UK
| |
Collapse
|
10
|
Venturini C, Pang J, Tamuri AU, Roy S, Atkinson C, Griffiths P, Breuer J, Goldstein RA. Haplotype assignment of longitudinal viral deep sequencing data using covariation of variant frequencies. Virus Evol 2022; 8:veac093. [PMID: 36478783 PMCID: PMC9719071 DOI: 10.1093/ve/veac093] [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: 10/07/2021] [Revised: 09/15/2022] [Accepted: 10/05/2022] [Indexed: 11/13/2022] Open
Abstract
Longitudinal deep sequencing of viruses can provide detailed information about intra-host evolutionary dynamics including how viruses interact with and transmit between hosts. Many analyses require haplotype reconstruction, identifying which variants are co-located on the same genomic element. Most current methods to perform this reconstruction are based on a high density of variants and cannot perform this reconstruction for slowly evolving viruses. We present a new approach, HaROLD (HAplotype Reconstruction Of Longitudinal Deep sequencing data), which performs this reconstruction based on identifying co-varying variant frequencies using a probabilistic framework. We illustrate HaROLD on both RNA and DNA viruses with synthetic Illumina paired read data created from mixed human cytomegalovirus (HCMV) and norovirus genomes, and clinical datasets of HCMV and norovirus samples, demonstrating high accuracy, especially when longitudinal samples are available.
Collapse
Affiliation(s)
- Cristina Venturini
- Infection, Immunity, Inflammation, Institute of Child Health, University College London, London WC1E 6BT, UK
| | - Juanita Pang
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Asif U Tamuri
- Research IT Services, University College London, London WC1E 6BT, UK
| | - Sunando Roy
- Infection, Immunity, Inflammation, Institute of Child Health, University College London, London WC1E 6BT, UK
| | - Claire Atkinson
- Institute for Immunity and Transplantation, University College London, London NW3 2PP, UK
| | - Paul Griffiths
- Institute for Immunity and Transplantation, University College London, London NW3 2PP, UK
| | - Judith Breuer
- Infection, Immunity, Inflammation, Institute of Child Health, University College London, London WC1E 6BT, UK
- Great Ormond Street Hospital for Children, London WC1N 3JH, UK
| | - Richard A Goldstein
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
- Infection, Immunity, Inflammation, Institute of Child Health, University College London, London WC1E 6BT, UK
| |
Collapse
|
11
|
Smith T, Hoyo-Vadillo C, Adom AA, Favari-Perozzi L, Gastine S, Dehbi HM, Villegas-Lara B, Mateos E, González YSP, Navarro-Gualito MD, Cruz-Carbajal AS, Cortes-Vazquez MA, Bekker-Méndez C, Aguirre-Alvarado C, Aguirre-Gil G, Delgado-Pastelin L, Owen A, Lowe D, Standing J, Escobedo J. Favipiravir and/or nitazoxanide: a randomized, double-blind, 2×2 design, placebo-controlled trial of early therapy in COVID-19 in health workers, their household members, and patients treated at IMSS (FANTAZE). Trials 2022; 23:583. [PMID: 35869526 PMCID: PMC9306230 DOI: 10.1186/s13063-022-06533-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/08/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The 2020 pandemic of SARS-CoV-2 causing COVID-19 disease is an unprecedented global emergency. COVID-19 appears to be a disease with an early phase where the virus replicates, coinciding with the first presentation of symptoms, followed by a later 'inflammatory' phase which results in severe disease in some individuals. It is known from other rapidly progressive infections such as sepsis and influenza that early treatment with antimicrobials is associated with a better outcome. The hypothesis is that this holds for COVID-19 and that early antiviral treatment may prevent progression to the later phase of the disease. METHODS Trial design: Phase IIA randomised, double-blind, 2 × 2 design, placebo-controlled, interventional trial. RANDOMISATION Participants will be randomised 1:1 by stratification, with the following factors: gender, obesity, symptomatic or asymptomatic, current smoking status presence or absence of comorbidity, and if the participant has or has not been vaccinated. BLINDING Participants and investigators will both be blinded to treatment allocation (double-blind). DISCUSSION We propose to conduct a proof-of-principle placebo-controlled clinical trial of favipiravir plus or minus nitazoxanide in health workers, their household members and patients treated at the Mexican Social Security Institute (IMSS) facilities. Participants with or without symptomatic COVID-19 or who tested positive will be assigned to receive favipiravir plus nitazoxanide or favipiravir plus nitazoxanide placebo. The primary outcome will be the difference in the amount of virus ('viral load') in the upper respiratory tract after 5 days of therapy. Secondary outcomes will include hospitalization, major morbidity and mortality, pharmacokinetics, and impact of antiviral therapy on viral genetic mutation rate. If favipiravir with nitazoxanide demonstrates important antiviral effects without significant toxicity, there will be a strong case for a larger trial in people at high risk of hospitalization or intensive care admission, for example older patients and/or those with comorbidities and with early disease. TRIAL REGISTRATION ClinicalTrials.gov NCT04918927 . Registered on June 9, 2021.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - David Lowe
- Division of Infection and Immunity, UCL, London, UK
| | | | | |
Collapse
|
12
|
Lu MC, Lin SC, Hsu YH, Chen SY. Epidemiology, Clinical Features, and Unusual Complications of Norovirus Infection in Taiwan: What We Know after Rotavirus Vaccines. Pathogens 2022; 11:pathogens11040451. [PMID: 35456126 PMCID: PMC9026459 DOI: 10.3390/pathogens11040451] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 02/01/2023] Open
Abstract
Noroviruses (NoVs) are one of the emerging and rapidly spreading groups of pathogens threatening human health. A reduction in sporadic NoV infections was noted following the start of the COVID-19 pandemic, but the return of NoV gastroenteritis during the COVID-19 pandemic has been noted recently. Research in recent years has shown that different virus strains are associated with different clinical characteristics; moreover, there is a paucity of research into extraintestinal or unusual complications that may be associated with NoV. The genomic diversity of circulating NoVs is also complex and may vary significantly. Therefore, this short narrative review focuses on sharing the Taiwan experience of NoV infection including epidemiology, clinical features, and complications following suboptimal rotavirus immunization in Taiwan (after October 2006). We also highlight the unusual complications associated with NoV infections and the impacts of NoV infection during the COVID-19 pandemic in the literature for possible future research directions. To conclude, further research is needed to quantify the burden of NoV across the spectrum of disease severity in Taiwan. The evidence of the connection between NoV and the unusual complications is still lacking.
Collapse
Affiliation(s)
- Meng-Che Lu
- Division of Allergy, Asthma and Immunology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (M.-C.L.); (S.-C.L.)
| | - Sheng-Chieh Lin
- Division of Allergy, Asthma and Immunology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; (M.-C.L.); (S.-C.L.)
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei city 11031, Taiwan
| | - Yi-Hsiang Hsu
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA;
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shih-Yen Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei city 11031, Taiwan
- Division of Pediatric Gastroenterology and Hepatology, Department of Pediatrics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
- Correspondence:
| |
Collapse
|
13
|
Boshier FAT, Pang J, Penner J, Parker M, Alders N, Bamford A, Grandjean L, Grunewald S, Hatcher J, Best T, Dalton C, Bynoe PD, Frauenfelder C, Köeglmeier J, Myerson P, Roy S, Williams R, de Silva TI, Goldstein RA, Breuer J. Evolution of viral variants in remdesivir-treated and untreated SARS-CoV-2-infected pediatrics patients. J Med Virol 2022; 94:161-172. [PMID: 34415583 PMCID: PMC8426849 DOI: 10.1002/jmv.27285] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/04/2021] [Accepted: 08/18/2021] [Indexed: 11/07/2022]
Abstract
Detailed information on intrahost viral evolution in SARS-CoV-2 with and without treatment is limited. Sequential viral loads and deep sequencing of SARS-CoV-2 from the upper respiratory tract of nine hospitalized children, three of whom were treated with remdesivir, revealed that remdesivir treatment suppressed viral load in one patient but not in a second infected with an identical strain without any evidence of drug resistance found. Reduced levels of subgenomic RNA during treatment of the second patient, suggest an additional effect of remdesivir on viral replication. Haplotype reconstruction uncovered persistent SARS-CoV-2 variant genotypes in four patients. These likely arose from within-host evolution, although superinfection cannot be excluded in one case. Although our dataset is small, observed sample-to-sample heterogeneity in variant frequencies across four of nine patients suggests the presence of discrete viral populations in the lung with incomplete population sampling in diagnostic swabs. Such compartmentalization could compromise the penetration of remdesivir into the lung, limiting the drugs in vivo efficacy, as has been observed in other lung infections.
Collapse
Affiliation(s)
- Florencia A. T. Boshier
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Juanita Pang
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
- Division of Infection and ImmunityUniversity College LondonLondonUK
| | - Justin Penner
- Department of Infectious DiseaseGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Matthew Parker
- Department of Infection, Immunity and Cardiovascular Diseases, The Florey InstituteUniversity of SheffieldSheffieldUK
| | - Nele Alders
- Department of Infectious DiseaseGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Alasdair Bamford
- Department of Infectious DiseaseGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Louis Grandjean
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Stephanie Grunewald
- Department of Metabolic MedicineUCL Great Ormond Street Institute of Child HealthLondonUK
| | - James Hatcher
- Department of MicrobiologyGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Timothy Best
- Department of MicrobiologyGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Caroline Dalton
- Department of PharmacyGreat Ormond Street Hospital for Children NHS TrustLondonUK
| | - Patricia Dyal Bynoe
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Claire Frauenfelder
- Department of EarsNose and Throat, Great Ormond Street Hospital for Children NHS Foundation TrustLondonUK
- Division of SurgeryUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Jutta Köeglmeier
- Department of GastroenterologyGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | - Phoebe Myerson
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Sunando Roy
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Rachel Williams
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Thushan I. de Silva
- Department of Infection, Immunity and Cardiovascular Diseases, The Florey InstituteUniversity of SheffieldSheffieldUK
| | | | - Judith Breuer
- Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
- Department of MicrobiologyGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| | | |
Collapse
|
14
|
Current and Future Antiviral Strategies to Tackle Gastrointestinal Viral Infections. Microorganisms 2021; 9:microorganisms9081599. [PMID: 34442677 PMCID: PMC8399003 DOI: 10.3390/microorganisms9081599] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 01/16/2023] Open
Abstract
Acute gastroenteritis caused by virus has a major impact on public health worldwide in terms of morbidity, mortality, and economic burden. The main culprits are rotaviruses, noroviruses, sapoviruses, astroviruses, and enteric adenoviruses. Currently, there are no antiviral drugs available for the prevention or treatment of viral gastroenteritis. Here, we describe the antivirals that were identified as having in vitro and/or in vivo activity against these viruses, originating from in silico design or library screening, natural sources or being repurposed drugs. We also highlight recent advances in model systems available for this (hard to cultivate) group of viruses, such as organoid technologies, and that will facilitate antiviral studies as well as fill some of current knowledge gaps that hamper the development of highly efficient therapies against gastroenteric viruses.
Collapse
|
15
|
Kondapi DS, Ramani S, Estes MK, Atmar RL, Okhuysen PC. Norovirus in Cancer Patients: A Review. Open Forum Infect Dis 2021; 8:ofab126. [PMID: 34189156 PMCID: PMC8232388 DOI: 10.1093/ofid/ofab126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022] Open
Abstract
Norovirus (NoV) is the leading cause of viral-related diarrhea in cancer patients, in whom it can be chronic, contributing to decreased quality of life, interruption of cancer care, malnutrition, and altered mucosal barrier function. Immunosuppressed cancer patients shed NoV for longer periods of time than immunocompetent hosts, favoring quasispecies development and emergence of novel NoV variants. While nucleic acid amplification tests (NAATs) for NoV diagnosis have revolutionized our understanding of NoV burden of disease, not all NAATs provide information on viral load or infecting genotype. There is currently no effective antiviral or vaccine for chronic NoV infections. Screening for inhibitors of NoV replication in intestinal organoid culture models and creation of NoV-specific adoptive T cells are promising new strategies to develop treatments for chronic NoV in immunosuppressed patients. Herein we summarize data on the epidemiology, clinical manifestations, diagnostic challenges, and treatment of NoV infection in patients with cancer.
Collapse
Affiliation(s)
- Divya Samantha Kondapi
- Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Section of Infectious Diseases, Baylor College of Medicine, Houston, Texas, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Robert L Atmar
- Section of Infectious Diseases, Baylor College of Medicine, Houston, Texas, USA
| | - Pablo C Okhuysen
- Infection Control and Employee Health, Division of Internal Medicine, Department of Infectious Diseases, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
16
|
Lumby CK, Zhao L, Oporto M, Best T, Tutill H, Shah D, Veys P, Williams R, Worth A, Illingworth CJR, Breuer J. Favipiravir and Zanamivir Cleared Infection with Influenza B in a Severely Immunocompromised Child. Clin Infect Dis 2021; 71:e191-e194. [PMID: 32124919 DOI: 10.1093/cid/ciaa023] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 03/01/2020] [Indexed: 01/08/2023] Open
Abstract
A combination of favipiravir and zanamivir successfully cleared influenza B infection in a child who had undergone bone marrow transplant for X-linked severe combined immunodeficiency, with no recovery of T lymphocytes. Deep sequencing of viral samples illuminated the within-host dynamics of infection, demonstrating the effectiveness of favipiravir in this case.
Collapse
Affiliation(s)
- Casper K Lumby
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Lei Zhao
- Department of Genetics, University of Cambridge, Cambridge, UK
| | | | - Tim Best
- Great Ormond Street Hospital, London, UK
| | - Helena Tutill
- Division of Infection and Immunity, University College London, London, UK
| | - Divya Shah
- Division of Infection and Immunity, University College London, London, UK
| | - Paul Veys
- Great Ormond Street Hospital, London, UK
| | - Rachel Williams
- Division of Infection and Immunity, University College London, London, UK
| | | | - Christopher J R Illingworth
- Department of Genetics, University of Cambridge, Cambridge, UK.,Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge
| | - Judy Breuer
- Great Ormond Street Hospital, London, UK.,Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
17
|
Li H, Jiang XM, Cui N, Yuan C, Zhang SF, Lu QB, Yang ZD, Xin QL, Song YB, Zhang XA, Liu HZ, Du J, Fan XJ, Yuan L, Yuan YM, Wang Z, Wang J, Zhang L, Zhang DN, Wang ZB, Dai K, Bai JY, Hao ZN, Fan H, Fang LQ, Xiao G, Yang Y, Peng K, Wang HQ, Li JX, Zhang LK, Liu W. Clinical effect and antiviral mechanism of T-705 in treating severe fever with thrombocytopenia syndrome. Signal Transduct Target Ther 2021; 6:145. [PMID: 33859168 PMCID: PMC8050330 DOI: 10.1038/s41392-021-00541-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/24/2021] [Accepted: 02/27/2021] [Indexed: 11/08/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) virus (SFTSV) is an emerging tick-borne virus with high fatality and an expanding endemic. Currently, effective anti-SFTSV intervention remains unavailable. Favipiravir (T-705) was recently reported to show in vitro and in animal model antiviral efficacy against SFTSV. Here, we conducted a single-blind, randomized controlled trial to assess the efficacy and safety of T-705 in treating SFTS (Chinese Clinical Trial Registry website, number ChiCTR1900023350). From May to August 2018, laboratory-confirmed SFTS patients were recruited from a designated hospital and randomly assigned to receive oral T-705 in combination with supportive care or supportive care only. Fatal outcome occurred in 9.5% (7/74) of T-705 treated patients and 18.3% (13/71) of controls (odds ratio, 0.466, 95% CI, 0.174-1.247). Cox regression showed a significant reduction in case fatality rate (CFR) with an adjusted hazard ratio of 0.366 (95% CI, 0.142-0.944). Among the low-viral load subgroup (RT-PCR cycle threshold ≥26), T-705 treatment significantly reduced CFR from 11.5 to 1.6% (P = 0.029), while no between-arm difference was observed in the high-viral load subgroup (RT-PCR cycle threshold <26). The T-705-treated group showed shorter viral clearance, lower incidence of hemorrhagic signs, and faster recovery of laboratory abnormities compared with the controls. The in vitro and animal experiments demonstrated that the antiviral efficacies of T-705 were proportionally induced by SFTSV mutation rates, particularly from two transition mutation types. The mutation analyses on T-705-treated serum samples disclosed a partially consistent mutagenesis pattern as those of the in vitro or animal experiments in reducing the SFTSV viral loads, further supporting the anti-SFTSV effect of T-705, especially for the low-viral loads.
Collapse
Affiliation(s)
- Hao Li
- Beijing Institute of Microbiology and Epidemiology, State Key Laboratory of Pathogen and Biosecurity, Beijing, P. R. China
| | - Xia-Ming Jiang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, P. R. China
| | - Ning Cui
- The 154 Hospital, People's Liberation Army, Xinyang, Henan, P. R. China
| | - Chun Yuan
- The 154 Hospital, People's Liberation Army, Xinyang, Henan, P. R. China
| | - Shao-Fei Zhang
- Beijing Institute of Microbiology and Epidemiology, State Key Laboratory of Pathogen and Biosecurity, Beijing, P. R. China
| | - Qing-Bin Lu
- School of Public Health, Peking University, Beijing, P. R. China
| | - Zhen-Dong Yang
- The 154 Hospital, People's Liberation Army, Xinyang, Henan, P. R. China
| | - Qin-Lin Xin
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, P. R. China
| | - Ya-Bin Song
- Beijing Institute of Microbiology and Epidemiology, State Key Laboratory of Pathogen and Biosecurity, Beijing, P. R. China
| | - Xiao-Ai Zhang
- Beijing Institute of Microbiology and Epidemiology, State Key Laboratory of Pathogen and Biosecurity, Beijing, P. R. China
| | - Hai-Zhou Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, P. R. China
| | - Juan Du
- School of Public Health, Peking University, Beijing, P. R. China
| | - Xue-Juan Fan
- The 154 Hospital, People's Liberation Army, Xinyang, Henan, P. R. China
| | - Lan Yuan
- The 154 Hospital, People's Liberation Army, Xinyang, Henan, P. R. China
| | - Yi-Mei Yuan
- The 154 Hospital, People's Liberation Army, Xinyang, Henan, P. R. China
| | - Zhen Wang
- The 154 Hospital, People's Liberation Army, Xinyang, Henan, P. R. China
| | - Juan Wang
- The 154 Hospital, People's Liberation Army, Xinyang, Henan, P. R. China
| | - Lan Zhang
- The 154 Hospital, People's Liberation Army, Xinyang, Henan, P. R. China
| | - Dong-Na Zhang
- Beijing Institute of Microbiology and Epidemiology, State Key Laboratory of Pathogen and Biosecurity, Beijing, P. R. China
| | - Zhi-Bo Wang
- Beijing Institute of Microbiology and Epidemiology, State Key Laboratory of Pathogen and Biosecurity, Beijing, P. R. China
| | - Ke Dai
- Beijing Institute of Microbiology and Epidemiology, State Key Laboratory of Pathogen and Biosecurity, Beijing, P. R. China
| | - Jie-Ying Bai
- Institute of Molecular Medicine, Peking University, Beijing, P. R. China
| | - Zhao-Nian Hao
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Hang Fan
- Beijing Institute of Microbiology and Epidemiology, State Key Laboratory of Pathogen and Biosecurity, Beijing, P. R. China
| | - Li-Qun Fang
- Beijing Institute of Microbiology and Epidemiology, State Key Laboratory of Pathogen and Biosecurity, Beijing, P. R. China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, P. R. China
| | - Yang Yang
- Department of Biostatistics and Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Ke Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, P. R. China
| | - Hong-Quan Wang
- Beijing Institute of Microbiology and Epidemiology, State Key Laboratory of Pathogen and Biosecurity, Beijing, P. R. China
| | - Jian-Xiong Li
- Department of Cancer, People's Liberation Army General Hospital, Beijing, P. R. China.
| | - Lei-Ke Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, P. R. China.
| | - Wei Liu
- Beijing Institute of Microbiology and Epidemiology, State Key Laboratory of Pathogen and Biosecurity, Beijing, P. R. China.
- Beijing Key Laboratory of Vector Borne and Natural Focus Infectious Diseases, Beijing, P. R. China.
| |
Collapse
|
18
|
Ashton G, Shand A, Arnott I, Din S. Profound diarrhoea and weight loss in an immunocompromised patient. BMJ Case Rep 2021; 14:14/1/e236913. [PMID: 33462001 PMCID: PMC7813290 DOI: 10.1136/bcr-2020-236913] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
A 75-year-old man was admitted with a 3-month history of worsening diarrhoea and weight loss. He was on long-term immunosuppression following cardiac transplantation. Investigations revealed herpes simplex oesophagitis and stool samples were positive for norovirus. Treatment with acyclovir and nitazoxanide resulted in a complete resolution of symptoms. Norovirus is a common cause of infectious gastroenteritis, but immunosuppressed patients may present with chronic diarrhoea rather than an acute illness. This case highlights the importance of a low clinical threshold for testing for norovirus infection in immunocompromised patients.
Collapse
Affiliation(s)
| | - Alan Shand
- Gastroenterology Unit, NHS Lothian, Edinburgh, UK
| | - Ian Arnott
- Gastroenterology Unit, NHS Lothian, Edinburgh, UK
| | - Shahida Din
- Gastroenterology Unit, NHS Lothian, Edinburgh, UK
| |
Collapse
|
19
|
Łagocka R, Dziedziejko V, Kłos P, Pawlik A. Favipiravir in Therapy of Viral Infections. J Clin Med 2021; 10:E273. [PMID: 33451007 PMCID: PMC7828521 DOI: 10.3390/jcm10020273] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 02/07/2023] Open
Abstract
Favipiravir (FPV) is a novel antiviral drug acting as a competitive inhibitor of RNA-dependent RNA polymerase (RdRp), preventing viral transcription and replication. FPV was approved in Japan in 2014 for therapy of influenza unresponsive to standard antiviral therapies. FPV was also used in the therapy of Ebola virus disease (EVD) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In this review, we discuss the mechanisms of action, pharmacokinetic parameters, toxicity, and adverse effects of FPV, as well as clinical studies evaluating the use of FPV in the therapy of influenza virus (IV) infection, EVD, and SARS-CoV-2 infection, along with its effectiveness in treating other human RNA infections.
Collapse
Affiliation(s)
- Ryta Łagocka
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, 70-204 Szczecin, Poland;
| | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-204 Szczecin, Poland; (V.D.); (P.K.)
| | - Patrycja Kłos
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-204 Szczecin, Poland; (V.D.); (P.K.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-204 Szczecin, Poland
| |
Collapse
|
20
|
Roberto P, Francesco L, Emanuela C, Giorgia G, Pasquale N, Sara D. Current treatment of COVID-19 in renal patients: hope or hype? Intern Emerg Med 2020; 15:1389-1398. [PMID: 32986137 PMCID: PMC7520511 DOI: 10.1007/s11739-020-02510-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/12/2020] [Indexed: 12/28/2022]
Abstract
To date the severe acute respiratory syndrome coronavirus 2 (SARS- CoV-2), known as COVID-19, is for clinicians the most difficult global therapeutic problem. In this landscape, the management of patients with chronic kidney disease, acute kidney injury or patients undergoing immunosuppressant therapies for kidney transplant or glomerular diseases, represent a clinical challenge for nephrologists, especially in patients with severe acute lung involvement. Therefore in this setting, due to the lack of anti-COVID treatment schedules, tailored management is mandatory to reduce the side effects, as consequence of impaired renal function and drugs interactions. We report the main treatment actually used against SARS-CoV-2, underlining its possible use in the nephropatic patients and the central role of nephrologists to improve the clinical outcome.
Collapse
Affiliation(s)
- Palumbo Roberto
- Nephology and Dialysis Department, Sant'Eugenio Hospital, Rome, Italy
| | | | - Cordova Emanuela
- Nephology and Dialysis Department, Sant'Eugenio Hospital, Rome, Italy
| | | | | | - Dominijanni Sara
- Nephology and Dialysis Department, Sant'Eugenio Hospital, Rome, Italy.
| |
Collapse
|
21
|
Kaptein SJF, Jacobs S, Langendries L, Seldeslachts L, Ter Horst S, Liesenborghs L, Hens B, Vergote V, Heylen E, Barthelemy K, Maas E, De Keyzer C, Bervoets L, Rymenants J, Van Buyten T, Zhang X, Abdelnabi R, Pang J, Williams R, Thibaut HJ, Dallmeier K, Boudewijns R, Wouters J, Augustijns P, Verougstraete N, Cawthorne C, Breuer J, Solas C, Weynand B, Annaert P, Spriet I, Vande Velde G, Neyts J, Rocha-Pereira J, Delang L. Favipiravir at high doses has potent antiviral activity in SARS-CoV-2-infected hamsters, whereas hydroxychloroquine lacks activity. Proc Natl Acad Sci U S A 2020; 117:26955-26965. [PMID: 33037151 PMCID: PMC7604414 DOI: 10.1073/pnas.2014441117] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) rapidly spread around the globe after its emergence in Wuhan in December 2019. With no specific therapeutic and prophylactic options available, the virus has infected millions of people of which more than half a million succumbed to the viral disease, COVID-19. The urgent need for an effective treatment together with a lack of small animal infection models has led to clinical trials using repurposed drugs without preclinical evidence of their in vivo efficacy. We established an infection model in Syrian hamsters to evaluate the efficacy of small molecules on both infection and transmission. Treatment of SARS-CoV-2-infected hamsters with a low dose of favipiravir or hydroxychloroquine with(out) azithromycin resulted in, respectively, a mild or no reduction in virus levels. However, high doses of favipiravir significantly reduced infectious virus titers in the lungs and markedly improved lung histopathology. Moreover, a high dose of favipiravir decreased virus transmission by direct contact, whereas hydroxychloroquine failed as prophylaxis. Pharmacokinetic modeling of hydroxychloroquine suggested that the total lung exposure to the drug did not cause the failure. Our data on hydroxychloroquine (together with previous reports in macaques and ferrets) thus provide no scientific basis for the use of this drug in COVID-19 patients. In contrast, the results with favipiravir demonstrate that an antiviral drug at nontoxic doses exhibits a marked protective effect against SARS-CoV-2 in a small animal model. Clinical studies are required to assess whether a similar antiviral effect is achievable in humans without toxic effects.
Collapse
Affiliation(s)
- Suzanne J F Kaptein
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium;
| | - Sofie Jacobs
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Lana Langendries
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Laura Seldeslachts
- Biomedical MRI and Molecular Small Animal Imaging Centre, Department of Imaging and Pathology, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Sebastiaan Ter Horst
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Laurens Liesenborghs
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Bart Hens
- Drug Delivery & Disposition, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Valentijn Vergote
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Elisabeth Heylen
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Karine Barthelemy
- Unité des Virus Emergents, Aix Marseille University, Institut de Recherche pour le Développement (IRD) 190, Institut National de la Santé et de la Recherche Médicale (INSERM) 1207, 13005 Marseille, France
| | - Elke Maas
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Carolien De Keyzer
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Lindsey Bervoets
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Jasper Rymenants
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Tina Van Buyten
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Xin Zhang
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Rana Abdelnabi
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Juanita Pang
- UCL Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, United Kingdom
| | - Rachel Williams
- UCL Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, United Kingdom
| | - Hendrik Jan Thibaut
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Kai Dallmeier
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Robbert Boudewijns
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Jens Wouters
- Molecular Small Animal Imaging Centre, Department of Imaging and Pathology, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Patrick Augustijns
- Drug Delivery & Disposition, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Nick Verougstraete
- Department of Laboratory Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Christopher Cawthorne
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Judith Breuer
- UCL Great Ormond Street Institute of Child Health, University College London, WC1N 1EH London, United Kingdom
| | - Caroline Solas
- Assistance Publique-Hôpitaux de Marseille, Aix-Marseille University, Unité des Virus Emergents, Institut de Recherche pour le Développement (IRD) 190, Institut National de la Santé et de la Recherche Médicale (INSERM) 1207, Laboratoire de Pharmacocinétique et Toxicologie, 13005 Marseille, France
| | - Birgit Weynand
- Translational Cell and Tissue Research, Department of Imaging and Pathology, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Pieter Annaert
- Drug Delivery & Disposition, Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Isabel Spriet
- Pharmacy Department, University Hospitals Leuven, 3000 Leuven, Belgium
- Department of Pharmaceutical and Pharmacological Sciences, Katholieke Universiteit Leuven-University of Leuven, 3000 Leuven, Belgium
| | - Greetje Vande Velde
- Biomedical MRI and Molecular Small Animal Imaging Centre, Department of Imaging and Pathology, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Johan Neyts
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium;
- Global Virus Network, Baltimore, MD 21201
| | - Joana Rocha-Pereira
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium;
| | - Leen Delang
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium;
| |
Collapse
|
22
|
Dehbi HM, Lowe DM, O'Quigley J. Early phase dose-finding trials in virology. Stat Med 2020; 40:240-253. [PMID: 33053601 DOI: 10.1002/sim.8771] [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: 04/02/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 11/10/2022]
Abstract
Little has been published in terms of dose-finding methodology in virology. Aside from a few papers focusing on HIV, the considerable progress in dose-finding methodology of the last 25 years has focused almost entirely on oncology. While adverse reactions to cytotoxic drugs may be life threatening, for anti-viral agents we anticipate something different: side effects that provoke the cessation of treatment. This would correspond to treatment failure. On the other hand, success would not be yes/no but would correspond to a range of responses, from small, no more than say 20% reduction in viral load to the complete elimination of the virus. Less than total success matters since this may allow the patient to achieve immune-mediated clearance. The motivation for this article is an upcoming dose-finding trial in chronic norovirus infection. We propose a novel methodology whose goal is twofold: first, to identify the dose that provides the most favorable distribution of treatment outcomes, and, second, to do this in a way that maximizes the treatment benefit for the patients included in the study.
Collapse
Affiliation(s)
- Hakim-Moulay Dehbi
- Comprehensive Clinical Trials Unit, University College London, London, UK
| | - David M Lowe
- Institute of Immunity and Transplantation, Royal Free Hospital, London, UK
| | - John O'Quigley
- Department of Statistical Science, University College London, London, UK
| |
Collapse
|
23
|
Yu P, Wang Y, Li Y, Li Y, Miao Z, Peppelenbosch MP, Pan Q. 2'-Fluoro-2'-deoxycytidine inhibits murine norovirus replication and synergizes MPA, ribavirin and T705. Arch Virol 2020; 165:2605-2613. [PMID: 32770483 PMCID: PMC7414258 DOI: 10.1007/s00705-020-04759-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/30/2020] [Indexed: 11/22/2022]
Abstract
Noroviruses are the main causative agents of acute viral gastroenteritis worldwide. However, no vaccine or specific antiviral treatment is available, imposing a heavy global health burden. The nucleoside analogue 2’-fluoro-2’-deoxycytidine (2’-FdC) has been reported to have broad antiviral activity. Here, we report that 2’-FdC significantly inhibits murine norovirus replication in macrophages. This effect was partially reversed by exogenous supplementation of cytidine triphosphate. The combination of 2’-FdC with mycophenolic acid, ribavirin or favipiravir (T705) exerts synergistic antiviral effects. These results indicate that 2’-FdC is a potential candidate for antiviral drug development against norovirus infection.
Collapse
Affiliation(s)
- Peifa Yu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Yining Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Yunlong Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Yang Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Zhijiang Miao
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Room Na-1005, 's-Gravendijkwal 230, 3015 CE, Rotterdam, The Netherlands.
| |
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW The majority of norovirus outbreaks in the United States occur in healthcare facilities. With the growing population of immunocompromised hosts who are in frequent contact with healthcare facilities, norovirus is not only a threat to hospitals and nursing homes but also to these individuals. This review summarizes the impact of norovirus infection on healthcare facilities and immunocompromised hosts. RECENT FINDINGS The natural history of norovirus infection in immunocompromised individuals remains poorly understood. Although host immune responses play a critical role in reducing duration of viral shedding and viral load in norovirus-infected individuals, why some immunocompromised patients spontaneously recover while others develop a chronic and protracted course of illness remains unclear. Norovirus outbreaks occur in healthcare facilities because the virus is highly contagious, resistant to disinfection and efficiently transmitted. The use of real-time metagenomic next-generation sequencing and phylogenetic analyses has provided valuable information on transmission patterns in complex hospital-associated norovirus outbreaks. The development of human intestinal enteroid cultures enables the determination of effectiveness of disinfectants against human noroviruses, circumventing the validity questions with surrogate virus models due to differences in susceptibility to inactivation and disinfectants. SUMMARY Metagenomics next-generation sequencing can enhance our understanding of norovirus transmission and lead to more timely mitigation strategies to curb norovirus outbreaks in healthcare facilities. With new in-vitro cultivation methods for human noroviruses, candidate vaccines and effective antivirals could be available in the near future.
Collapse
|
25
|
Domman D, Ruis C, Dorman MJ, Shakya M, Chain PSG. Novel Insights Into the Spread of Enteric Pathogens Using Genomics. J Infect Dis 2020; 221:S319-S330. [PMID: 31538189 DOI: 10.1093/infdis/jiz220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/19/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Daryl Domman
- Bioscience Division, Los Alamos National Laboratory, New Mexico
| | - Christopher Ruis
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, United Kingdom.,Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Matthew J Dorman
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, United Kingdom
| | - Migun Shakya
- Bioscience Division, Los Alamos National Laboratory, New Mexico
| | | |
Collapse
|
26
|
Kerber R, Lorenz E, Duraffour S, Sissoko D, Rudolf M, Jaeger A, Cisse SD, Camara AM, Miranda O, Castro CM, Akoi Bore J, Raymond Koundouno F, Repits J, Afrough B, Becker-Ziaja B, Hinzmann J, Mertens M, Vitoriano I, Hugh Logue C, Böttcher JP, Pallasch E, Sachse A, Bah A, Cabeza-Cabrerizo M, Nitzsche K, Kuisma E, Michel J, Holm T, Zekeng EG, Cowley LA, Garcia-Dorival I, Hetzelt N, Baum JHJ, Portmann J, Carter L, Yenamaberhan RL, Camino A, Enkirch T, Singethan K, Meisel S, Mazzarelli A, Kosgei A, Kafetzopoulou L, Rickett NY, Patrono LV, Ghebreghiorghis L, Arnold U, Colin G, Juchet S, Marchal CL, Kolie JS, Beavogui AH, Wurr S, Bockholt S, Krumkamp R, May J, Stoecker K, Fleischmann E, Ippolito G, Carroll MW, Koivogui L, Magassouba N, Keita S, Gurry C, Drury P, Diallo B, Formenty P, Wölfel R, Di Caro A, Gabriel M, Anglaret X, Malvy D, Günther S. Laboratory Findings, Compassionate Use of Favipiravir, and Outcome in Patients With Ebola Virus Disease, Guinea, 2015-A Retrospective Observational Study. J Infect Dis 2020; 220:195-202. [PMID: 30788508 PMCID: PMC6581890 DOI: 10.1093/infdis/jiz078] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 02/14/2019] [Indexed: 11/14/2022] Open
Abstract
Background In 2015, the laboratory at the Ebola treatment center in Coyah, Guinea, confirmed Ebola virus disease (EVD) in 286 patients. The cycle threshold (Ct) of an Ebola virus–specific reverse transcription–polymerase chain reaction assay and 13 blood chemistry parameters were measured on admission and during hospitalization. Favipiravir treatment was offered to patients with EVD on a compassionate-use basis. Methods To reduce biases in the raw field data, we carefully selected 163 of 286 patients with EVD for a retrospective study to assess associations between potential risk factors, alterations in blood chemistry findings, favipiravir treatment, and outcome. Results The case-fatality rate in favipiravir-treated patients was lower than in untreated patients (42.5% [31 of 73] vs 57.8% [52 of 90]; P = .053 by univariate analysis). In multivariate regression analysis, a higher Ct and a younger age were associated with survival (P < .001), while favipiravir treatment showed no statistically significant effect (P = .11). However, Kaplan-Meier analysis indicated a longer survival time in the favipiravir-treated group (P = .015). The study also showed characteristic changes in blood chemistry findings in patients who died, compared with survivors. Conclusions Consistent with the JIKI trial, this retrospective study revealed a trend toward improved survival in favipiravir- treated patients; however, the effect of treatment was not statistically significant, except for its influence on survival time.
Collapse
Affiliation(s)
- Romy Kerber
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Eva Lorenz
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,German Center for Infection Research, Braunschweig
| | - Sophie Duraffour
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Daouda Sissoko
- INSERM U1219, Bordeaux University, Bordeaux, France.,Bordeaux University Hospital, Bordeaux, France
| | - Martin Rudolf
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Anna Jaeger
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,German Center for Infection Research, Braunschweig
| | | | | | | | | | - Joseph Akoi Bore
- European Mobile Laboratory Consortium, Hamburg.,Ministry of Health Guinea, Guinea
| | | | - Johanna Repits
- European Mobile Laboratory Consortium, Hamburg.,Janssen-Cilag, Sollentuna, Sweden
| | - Babak Afrough
- European Mobile Laboratory Consortium, Hamburg.,Public Health England, Porton Down
| | - Beate Becker-Ziaja
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Julia Hinzmann
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Marc Mertens
- European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig.,Friedrich Loeffler Institute, Federal Research Institute for Animal Health, Greifswald
| | - Ines Vitoriano
- European Mobile Laboratory Consortium, Hamburg.,Public Health England, Porton Down
| | | | - Jan-Peter Böttcher
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Elisa Pallasch
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Andreas Sachse
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Amadou Bah
- European Mobile Laboratory Consortium, Hamburg.,Swiss Tropical and Public Health Institute, Basel
| | | | | | - Eeva Kuisma
- European Mobile Laboratory Consortium, Hamburg.,Public Health England, Porton Down
| | - Janine Michel
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Tobias Holm
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | | | - Lauren A Cowley
- European Mobile Laboratory Consortium, Hamburg.,Public Health England, London.,Milner Centre for Evolution, University of Bath, Bath
| | - Isabel Garcia-Dorival
- European Mobile Laboratory Consortium, Hamburg.,Institute of Infection and Global Health, University of Liverpool, Liverpool
| | - Nicole Hetzelt
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Jonathan Hans Josef Baum
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg
| | - Jasmine Portmann
- European Mobile Laboratory Consortium, Hamburg.,Federal Office for Civil Protection, Spiez Laboratory, Spiez
| | - Lisa Carter
- European Mobile Laboratory Consortium, Hamburg.,University College London, London.,World Health Organization, Geneva, Switzerland
| | - Rahel Lemma Yenamaberhan
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg
| | | | - Theresa Enkirch
- European Mobile Laboratory Consortium, Hamburg.,Division of Veterinary Medicine, Paul-Ehrlich-Institut, Langen, Germany
| | - Katrin Singethan
- European Mobile Laboratory Consortium, Hamburg.,Institute of Virology, Technische Universität München, Munich, Germany
| | - Sarah Meisel
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg
| | - Antonio Mazzarelli
- European Mobile Laboratory Consortium, Hamburg.,National Institute for Infectious Diseases "Lazzaro Spallanzani," Rome, Italy
| | - Abigail Kosgei
- European Mobile Laboratory Consortium, Hamburg.,Kenya Medical Research Institute, Nairobi, Kenya
| | - Liana Kafetzopoulou
- European Mobile Laboratory Consortium, Hamburg.,KU Leuven-University of Leuven, Rega Institute for Medical Research, Leuven, Belgium
| | - Natasha Y Rickett
- European Mobile Laboratory Consortium, Hamburg.,Institute of Infection and Global Health, University of Liverpool, Liverpool
| | - Livia Victoria Patrono
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg
| | | | - Ulrike Arnold
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Géraldine Colin
- INSERM U1219, Bordeaux University, Bordeaux, France.,Bordeaux University Hospital, Bordeaux, France.,PAC-CI, ANRS Research Site, Treichville University Hospital, Abidjan, Côte d'Ivoire
| | - Sylvain Juchet
- INSERM U1219, Bordeaux University, Bordeaux, France.,Bordeaux University Hospital, Bordeaux, France.,PAC-CI, ANRS Research Site, Treichville University Hospital, Abidjan, Côte d'Ivoire
| | | | | | | | - Stephanie Wurr
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Sabrina Bockholt
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Ralf Krumkamp
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,German Center for Infection Research, Braunschweig
| | - Jürgen May
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,German Center for Infection Research, Braunschweig
| | - Kilian Stoecker
- European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig.,Bundeswehr Institute of Microbiology, Munich, Germany
| | - Erna Fleischmann
- European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig.,Bundeswehr Institute of Microbiology, Munich, Germany
| | - Giuseppe Ippolito
- European Mobile Laboratory Consortium, Hamburg.,National Institute for Infectious Diseases "Lazzaro Spallanzani," Rome, Italy
| | - Miles W Carroll
- European Mobile Laboratory Consortium, Hamburg.,Public Health England, Porton Down.,University of Southampton, South General Hospital, Southampton, United Kingdom
| | | | - N'Faly Magassouba
- Laboratoire des Fièvres Hémorragiques en Guinée, Université Gamal Abdel Nasser de Conakry, Guinea
| | | | | | | | | | | | - Roman Wölfel
- European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig.,Bundeswehr Institute of Microbiology, Munich, Germany
| | - Antonino Di Caro
- European Mobile Laboratory Consortium, Hamburg.,National Institute for Infectious Diseases "Lazzaro Spallanzani," Rome, Italy
| | - Martin Gabriel
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Xavier Anglaret
- INSERM U1219, Bordeaux University, Bordeaux, France.,Bordeaux University Hospital, Bordeaux, France.,PAC-CI, ANRS Research Site, Treichville University Hospital, Abidjan, Côte d'Ivoire
| | - Denis Malvy
- INSERM U1219, Bordeaux University, Bordeaux, France.,Bordeaux University Hospital, Bordeaux, France
| | - Stephan Günther
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| |
Collapse
|
27
|
Shiraki K, Daikoku T. Favipiravir, an anti-influenza drug against life-threatening RNA virus infections. Pharmacol Ther 2020; 209:107512. [PMID: 32097670 PMCID: PMC7102570 DOI: 10.1016/j.pharmthera.2020.107512] [Citation(s) in RCA: 292] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/14/2020] [Indexed: 12/16/2022]
Abstract
Favipiravir has been developed as an anti-influenza drug and licensed as an anti-influenza drug in Japan. Additionally, favipiravir is being stockpiled for 2 million people as a countermeasure for novel influenza strains. This drug functions as a chain terminator at the site of incorporation of the viral RNA and reduces the viral load. Favipiravir cures all mice in a lethal influenza infection model, while oseltamivir fails to cure the animals. Thus, favipiravir contributes to curing animals with lethal infection. In addition to influenza, favipiravir has a broad spectrum of anti-RNA virus activities in vitro and efficacies in animal models with lethal RNA viruses and has been used for treatment of human infection with life-threatening Ebola virus, Lassa virus, rabies, and severe fever with thrombocytopenia syndrome. The best feature of favipiravir as an antiviral agent is the apparent lack of generation of favipiravir-resistant viruses. Favipiravir alone maintains its therapeutic efficacy from the first to the last patient in an influenza pandemic or an epidemic lethal RNA virus infection. Favipiravir is expected to be an important therapeutic agent for severe influenza, the next pandemic influenza strain, and other severe RNA virus infections for which standard treatments are not available.
Collapse
Affiliation(s)
- Kimiyasu Shiraki
- Senri Kinran University and Department of Virology, University of Toyama, Japan.
| | - Tohru Daikoku
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Hokuriku University, Japan
| |
Collapse
|
28
|
Brown LAK, Ruis C, Clark I, Roy S, Brown JR, Albuquerque AS, Patel SY, Miller J, Karim MY, Dervisevic S, Moore J, Williams CA, Cudini J, Moreira F, Neild P, Seneviratne SL, Workman S, Toumpanakis C, Atkinson C, Burns SO, Breuer J, Lowe DM. A comprehensive characterization of chronic norovirus infection in immunodeficient hosts. J Allergy Clin Immunol 2019; 144:1450-1453. [PMID: 31415785 PMCID: PMC6843911 DOI: 10.1016/j.jaci.2019.07.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/11/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Li-An K Brown
- Department of Infectious Diseases, Royal Free London NHS Foundation Trust, London, United Kingdom; Department of Microbiology, Whittington Health NHS Trust, London, United Kingdom
| | - Christopher Ruis
- Division of Infection and Immunity, University College London, London, United Kingdom; Molecular Immunity Unit, Department of Medicine, University of Cambridge, Medical Research Council (MRC)-Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Ian Clark
- Department of Histopathology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Sunando Roy
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Julianne R Brown
- Department of Microbiology, Virology and Infection Prevention and Control, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Adriana S Albuquerque
- Institute of Immunity and Transplantation, University College London, Royal Free Campus, London, United Kingdom
| | - Smita Y Patel
- Oxford University Hospitals NHS Trust and NIHR Oxford Biomedical Research Centre, Oxford, United Kingdom
| | - Joanne Miller
- Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, Surrey, United Kingdom
| | - Mohammed Yousuf Karim
- Royal Surrey County Hospital NHS Foundation Trust, Egerton Road, Guildford, Surrey, United Kingdom; Pathology, Sidra Medicine, Doha, Qatar
| | - Samir Dervisevic
- Norfolk and Norwich University Hospital, Norwich, Norfolk, United Kingdom
| | - Jennifer Moore
- Norfolk and Norwich University Hospital, Norwich, Norfolk, United Kingdom
| | - Charlotte A Williams
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Juliana Cudini
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Fernando Moreira
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Penny Neild
- St George's University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Suranjith L Seneviratne
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Sarita Workman
- Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Christos Toumpanakis
- Department of Gastroenterology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Claire Atkinson
- Institute of Immunity and Transplantation, University College London, Royal Free Campus, London, United Kingdom
| | - Siobhan O Burns
- Institute of Immunity and Transplantation, University College London, Royal Free Campus, London, United Kingdom; Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Judith Breuer
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - David M Lowe
- Institute of Immunity and Transplantation, University College London, Royal Free Campus, London, United Kingdom; Department of Clinical Immunology, Royal Free London NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
29
|
Sempere RN, Arias A. Establishment of a Cell Culture Model of Persistent Flaviviral Infection: Usutu Virus Shows Sustained Replication during Passages and Resistance to Extinction by Antiviral Nucleosides. Viruses 2019; 11:E560. [PMID: 31212939 PMCID: PMC6630443 DOI: 10.3390/v11060560] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 06/02/2019] [Accepted: 06/15/2019] [Indexed: 12/30/2022] Open
Abstract
Chronic viral disease constitutes a major global health problem, with several hundred million people affected and an associated elevated number of deaths. An increasing number of disorders caused by human flaviviruses are related to their capacity to establish a persistent infection. Here we show that Usutu virus (USUV), an emerging zoonotic flavivirus linked to sporadic neurologic disease in humans, can establish a persistent infection in cell culture. Two independent lineages of Vero cells surviving USUV lytic infection were cultured over 82 days (41 cell transfers) without any apparent cytopathology crisis associated. We found elevated titers in the supernatant of these cells, with modest fluctuations during passages but no overall tendency towards increased or decreased infectivity. In addition to full-length genomes, viral RNA isolated from these cells at passage 40 revealed the presence of defective genomes, containing different deletions at the 5' end. These truncated transcripts were all predicted to encode shorter polyprotein products lacking membrane and envelope structural proteins, and most of non-structural protein 1. Treatment with different broad-range antiviral nucleosides revealed that USUV is sensitive to these compounds in the context of a persistent infection, in agreement with previous observations during lytic infections. The exposure of infected cells to prolonged treatment (10 days) with favipiravir and/or ribavirin resulted in the complete clearance of infectivity in the cellular supernatants (decrease of ~5 log10 in virus titers and RNA levels), although modest changes in intracellular viral RNA levels were recorded (<2 log10 decrease). Drug withdrawal after treatment day 10 resulted in a relapse in virus titers. These results encourage the use of persistently-infected cultures as a surrogate system in the identification of improved antivirals against flaviviral chronic disease.
Collapse
Affiliation(s)
- Raquel Navarro Sempere
- Life Science & Bioengineering Building, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
- Abiopep Sociedad Limitada, Parque Científico de Murcia, 30100 Murcia, Spain.
| | - Armando Arias
- Life Science & Bioengineering Building, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| |
Collapse
|
30
|
Smertina E, Urakova N, Strive T, Frese M. Calicivirus RNA-Dependent RNA Polymerases: Evolution, Structure, Protein Dynamics, and Function. Front Microbiol 2019; 10:1280. [PMID: 31244803 PMCID: PMC6563846 DOI: 10.3389/fmicb.2019.01280] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/22/2019] [Indexed: 12/11/2022] Open
Abstract
The Caliciviridae are viruses with a positive-sense, single-stranded RNA genome that is packaged into an icosahedral, environmentally stable protein capsid. The family contains five genera (Norovirus, Nebovirus, Sapovirus, Lagovirus, and Vesivirus) that infect vertebrates including amphibians, reptiles, birds, and mammals. The RNA-dependent RNA polymerase (RdRp) replicates the genome of RNA viruses and can speed up evolution due to its error-prone nature. Studying calicivirus RdRps in the context of genuine virus replication is often hampered by a lack of suitable model systems. Enteric caliciviruses and RHDV in particular are notoriously difficult to propagate in cell culture; therefore, molecular studies of replication mechanisms are challenging. Nevertheless, research on recombinant proteins has revealed several unexpected characteristics of calicivirus RdRps. For example, the RdRps of RHDV and related lagoviruses possess the ability to expose a hydrophobic motif, to rearrange Golgi membranes, and to copy RNA at unusually high temperatures. This review is focused on the structural dynamics, biochemical properties, kinetics, and putative interaction partners of these RdRps. In addition, we discuss the possible existence of a conserved but as yet undescribed structural element that is shared amongst the RdRps of all caliciviruses.
Collapse
Affiliation(s)
- Elena Smertina
- Commonwealth Scientific and Industrial Research Organisation, Health and Biosecurity, Canberra, ACT, Australia
- Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Nadya Urakova
- Department of Entomology, Pennsylvania State University, University Park, PA, United States
| | - Tanja Strive
- Commonwealth Scientific and Industrial Research Organisation, Health and Biosecurity, Canberra, ACT, Australia
- Invasive Animals Cooperative Research Centre, University of Canberra, Canberra, ACT, Australia
| | - Michael Frese
- Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| |
Collapse
|
31
|
Enosi Tuipulotu D, Fumian TM, Netzler NE, Mackenzie JM, White PA. The Adenosine Analogue NITD008 has Potent Antiviral Activity against Human and Animal Caliciviruses. Viruses 2019; 11:v11060496. [PMID: 31151251 PMCID: PMC6631109 DOI: 10.3390/v11060496] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/24/2019] [Accepted: 05/25/2019] [Indexed: 12/27/2022] Open
Abstract
The widespread nature of calicivirus infections globally has a substantial impact on the health and well-being of humans and animals alike. Currently, the only vaccines approved against caliciviruses are for feline and rabbit-specific members of this group, and thus there is a growing effort towards the development of broad-spectrum antivirals for calicivirus infections. In this study, we evaluated the antiviral activity of the adenosine analogue NITD008 in vitro using three calicivirus model systems namely; feline calicivirus (FCV), murine norovirus (MNV), and the human norovirus replicon. We show that the nucleoside analogue (NA), NITD008, has limited toxicity and inhibits calicivirus replication in all three model systems with EC50 values of 0.94 μM, 0.91 µM, and 0.21 µM for MNV, FCV, and the Norwalk replicon, respectively. NITD008 has a similar level of potency to the most well-studied NA 2′-C-methylcytidine in vitro. Significantly, we also show that continual NITD008 treatment effectively cleared the Norwalk replicon from cells and treatment with 5 µM NITD008 was sufficient to completely prevent rebound. Given the potency displayed by NITD008 against several caliciviruses, we propose that this compound should be interrogated further to assess its effectiveness in vivo. In summary, we have added a potent NA to the current suite of antiviral compounds and provide a NA scaffold that could be further modified for therapeutic use against calicivirus infections.
Collapse
Affiliation(s)
- Daniel Enosi Tuipulotu
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Tulio M Fumian
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
- Laboratório de Virologia Comparada e Ambiental, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-900, Brazil.
| | - Natalie E Netzler
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Jason M Mackenzie
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VC 3010, Australia.
| | - Peter A White
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
32
|
Jones TPW, Buckland M, Breuer J, Lowe DM. Viral infection in primary antibody deficiency syndromes. Rev Med Virol 2019; 29:e2049. [PMID: 31016825 DOI: 10.1002/rmv.2049] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/24/2019] [Accepted: 03/25/2019] [Indexed: 12/22/2022]
Abstract
Patients with primary antibody deficiency syndromes such as X-linked agammaglobulinemia (XLA) and common variable immunodeficiency (CVID) are at increased risk of severe and invasive infection. Viral infection in these populations has been of increasing interest as evidence mounts that viruses contribute significant morbidity and mortality: this is mediated both directly and via aberrant immune responses. We explain the importance of the humoral immune system in defence against viral pathogens before highlighting several significant viral syndromes in patients with antibody deficiency. We explore historical cases of hepatitis C via contaminated immunoglobulin products, the predisposition to invasive enteroviral infections, prolonged excretion of vaccine-derived poliovirus, the morbidity of chronic norovirus infection, and recent literature revealing the importance of respiratory viral infections. We discuss evidence that herpesviruses may play a role in driving the inflammatory disease seen in a subset of patients. We explore the phenomenon of within-host evolution during chronic viral infection and the potential emergence of new pathogenic strains. We highlight novel and emerging viruses identified via deep sequencing techniques. We describe the treatment strategies that have been attempted in all these scenarios and the urgent outstanding questions for research.
Collapse
Affiliation(s)
- Timothy P W Jones
- Department of Infectious Disease and Microbiology, Royal Free Hospital, London, UK
| | - Matthew Buckland
- Institute of Immunity and Transplantation, Royal Free Campus, University College, London, UK
| | - Judith Breuer
- Division of Infection and Immunity, University College London, London, UK
| | - David M Lowe
- Institute of Immunity and Transplantation, Royal Free Campus, University College, London, UK
| |
Collapse
|
33
|
Bassetto M, Van Dycke J, Neyts J, Brancale A, Rocha-Pereira J. Targeting the Viral Polymerase of Diarrhea-Causing Viruses as a Strategy to Develop a Single Broad-Spectrum Antiviral Therapy. Viruses 2019; 11:v11020173. [PMID: 30791582 PMCID: PMC6409847 DOI: 10.3390/v11020173] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 01/01/2023] Open
Abstract
Viral gastroenteritis is an important cause of morbidity and mortality worldwide, being particularly severe for children under the age of five. The most common viral agents of gastroenteritis are noroviruses, rotaviruses, sapoviruses, astroviruses and adenoviruses, however, no specific antiviral treatment exists today against any of these pathogens. We here discuss the feasibility of developing a broad-spectrum antiviral treatment against these diarrhea-causing viruses. This review focuses on the viral polymerase as an antiviral target, as this is the most conserved viral protein among the diverse viral families to which these viruses belong to. We describe the functional and structural similarities of the different viral polymerases, the antiviral effect of reported polymerase inhibitors and highlight common features that might be exploited in an attempt of designing such pan-polymerase inhibitor.
Collapse
Affiliation(s)
- Marcella Bassetto
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, CF10 3NB Cardiff, UK.
| | - Jana Van Dycke
- KU Leuven-Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven 3000, Belgium.
| | - Johan Neyts
- KU Leuven-Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven 3000, Belgium.
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, CF10 3NB Cardiff, UK.
| | - Joana Rocha-Pereira
- KU Leuven-Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, University of Leuven, Leuven 3000, Belgium.
| |
Collapse
|