1
|
Chiarelli A, Dobrovolny H. Viral Rebound After Antiviral Treatment: A Mathematical Modeling Study of the Role of Antiviral Mechanism of Action. Interdiscip Sci 2024; 16:844-853. [PMID: 39033482 DOI: 10.1007/s12539-024-00643-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 07/23/2024]
Abstract
The development of antiviral treatments for SARS-CoV-2 was an important turning point for the pandemic. Availability of safe and effective antivirals has allowed people to return back to normal life. While SARS-CoV-2 antivirals are highly effective at preventing severe disease, there have been concerning reports of viral rebound in some patients after cessation of antiviral treatment. In this study, we use a mathematical model of viral infection to study the potential of different antivirals to prevent viral rebound. We find that antivirals that block production are most likely to result in viral rebound if the treatment time course is not sufficiently long. Since these antivirals do not prevent infection of cells, cells continue to be infected during treatment. When treatment is stopped, the infected cells will begin producing virus at the usual rate. Antivirals that prevent infection of cells are less likely to result in viral rebound since cells are not being infected during treatment. This study highlights the role of antiviral mechanism of action in increasing or reducing the probability of viral rebound.
Collapse
Affiliation(s)
- Aubrey Chiarelli
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, 76129, USA
| | - Hana Dobrovolny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, 76129, USA.
| |
Collapse
|
2
|
Madueme PGU, Chirove F. A systematic review of mathematical models of Lassa fever. Math Biosci 2024; 374:109227. [PMID: 38844262 DOI: 10.1016/j.mbs.2024.109227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/21/2024] [Accepted: 05/31/2024] [Indexed: 06/13/2024]
Abstract
This systematic review, conducted following the PRISMA guidelines, scrutinizes mathematical models employed in the study of Lassa fever. The analysis revealed the inherent heterogeneity in both models and data, posing significant challenges to parameter estimation. While health and behavioral interventions exhibit promise in mitigating the disease's spread, their efficacy is contingent upon contextual factors. Identified through this review are critical gaps, limitations, and avenues for future research, necessitating increased harmonization and standardization in modeling approaches. The considerations of seasonal and spatial variations emerge as crucial elements demanding targeted investigation. The perpetual threat of emerging diseases, coupled with the enduring public health impact of Lassa fever, underscores the imperative for sustained research endeavors and investments in mathematical modeling. The conclusion underscored that while mathematical modeling remains an invaluable tool in the combat against Lassa fever, its optimal utilization mandates multidisciplinary collaboration, refined data collection methodologies, and an enriched understanding of the intricate disease dynamics. This comprehensive approach is essential for effectively reducing the burden of Lassa fever and safeguarding the health of vulnerable populations.
Collapse
Affiliation(s)
- Praise-God Uchechukwu Madueme
- Department of Mathematics and Applied Mathematics, University of Johannesburg, Auckland Park, 2006, Johannesburg, South Africa
| | - Faraimunashe Chirove
- Department of Mathematics and Applied Mathematics, University of Johannesburg, Auckland Park, 2006, Johannesburg, South Africa.
| |
Collapse
|
3
|
Nuñez IA, Crane A, Crozier I, Worwa G, Kuhn JH. Treatment of highly virulent mammarenavirus infections-status quo and future directions. Expert Opin Drug Discov 2024; 19:537-551. [PMID: 38606475 PMCID: PMC11069405 DOI: 10.1080/17460441.2024.2340494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
INTRODUCTION Mammarenaviruses are negative-sense bisegmented enveloped RNA viruses that are endemic in Africa, the Americas, and Europe. Several are highly virulent, causing acute human diseases associated with high case fatality rates, and are considered to be significant with respect to public health impact or bioterrorism threat. AREAS COVERED This review summarizes the status quo of treatment development, starting with drugs that are in advanced stages of evaluation in early clinical trials, followed by promising candidate medical countermeasures emerging from bench analyses and investigational animal research. EXPERT OPINION Specific therapeutic treatments for diseases caused by mammarenaviruses remain limited to the off-label use of ribavirin and transfusion of convalescent sera. Progress in identifying novel candidate medical countermeasures against mammarenavirus infection has been slow in part because of the biosafety and biosecurity requirements. However, novel methodologies and tools have enabled increasingly efficient high-throughput molecular screens of regulatory-agency-approved small-molecule drugs and led to the identification of several compounds that could be repurposed for the treatment of infection with several mammarenaviruses. Unfortunately, most of them have not yet been evaluated in vivo. The most promising treatment under development is a monoclonal antibody cocktail that is protective against multiple lineages of the Lassa virus in nonhuman primate disease models.
Collapse
Affiliation(s)
- Ivette A. Nuñez
- Integrated Research Facility at Fort Detrick, Division of
Clinical Research, National Institute of Allergy and Infectious Diseases, National
Institutes of Health, Fort Detrick, Frederick, MD21702, USA
| | - Anya Crane
- Integrated Research Facility at Fort Detrick, Division of
Clinical Research, National Institute of Allergy and Infectious Diseases, National
Institutes of Health, Fort Detrick, Frederick, MD21702, USA
| | - Ian Crozier
- Clinical Monitoring Research Program Directorate, Frederick
National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Gabriella Worwa
- Integrated Research Facility at Fort Detrick, Division of
Clinical Research, National Institute of Allergy and Infectious Diseases, National
Institutes of Health, Fort Detrick, Frederick, MD21702, USA
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick, Division of
Clinical Research, National Institute of Allergy and Infectious Diseases, National
Institutes of Health, Fort Detrick, Frederick, MD21702, USA
| |
Collapse
|
4
|
Reyna RA, Littlefield KE, Shehu N, Makishima T, Maruyama J, Paessler S. The Importance of Lassa Fever and Its Disease Management in West Africa. Viruses 2024; 16:266. [PMID: 38400041 PMCID: PMC10892767 DOI: 10.3390/v16020266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/21/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Lassa virus (LASV) is a zoonotic pathogen endemic throughout western Africa and is responsible for a human disease known as Lassa fever (LF). Historically, LASV has been emphasized as one of the greatest public health threats in West Africa, with up to 300,000 cases and 5000 associated deaths per year. This, and the fact that the disease has been reported in travelers, has driven a rapid production of various vaccine candidates. Several of these vaccines are currently in clinical development, despite limitations in understanding the immune response to infection. Alarmingly, the host immune response has been implicated in the induction of sensorineural hearing loss in LF survivors, legitimately raising safety questions about any future vaccines as well as efficacy in preventing potential hearing loss. The objective of this article is to revisit the importance and prevalence of LF in West Africa, with focus on Nigeria, and discuss current therapeutic approaches and ongoing vaccine development. In addition, we aim to emphasize the need for more scientific studies relating to LF-associated hearing loss, and to promote critical discussion about potential risks and benefits of vaccinating the population in endemic regions of West Africa.
Collapse
Affiliation(s)
- Rachel A. Reyna
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kirsten E. Littlefield
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nathan Shehu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Infectious Disease Unit, Department of Medicine, Jos University Teaching Hospital, Jos P.M.B. 2076, Nigeria
| | - Tomoko Makishima
- Department of Otolaryngology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Junki Maruyama
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
5
|
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
|
6
|
Karim M, Lo CW, Einav S. Preparing for the next viral threat with broad-spectrum antivirals. J Clin Invest 2023; 133:e170236. [PMID: 37259914 PMCID: PMC10232003 DOI: 10.1172/jci170236] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023] Open
Abstract
There is a large global unmet need for the development of countermeasures to combat hundreds of viruses known to cause human disease and for the establishment of a therapeutic portfolio for future pandemic preparedness. Most approved antiviral therapeutics target proteins encoded by a single virus, providing a narrow spectrum of coverage. This, combined with the slow pace and high cost of drug development, limits the scalability of this direct-acting antiviral (DAA) approach. Here, we summarize progress and challenges in the development of broad-spectrum antivirals that target either viral elements (proteins, genome structures, and lipid envelopes) or cellular proviral factors co-opted by multiple viruses via newly discovered compounds or repurposing of approved drugs. These strategies offer new means for developing therapeutics against both existing and emerging viral threats that complement DAAs.
Collapse
Affiliation(s)
- Marwah Karim
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, and
| | - Chieh-Wen Lo
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, and
| | - Shirit Einav
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, and
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
- Chan Zuckerberg Biohub San Francisco, San Francisco, California, USA
| |
Collapse
|
7
|
Saito T, Reyna RA, Taniguchi S, Littlefield K, Paessler S, Maruyama J. Vaccine Candidates against Arenavirus Infections. Vaccines (Basel) 2023; 11:635. [PMID: 36992218 PMCID: PMC10057967 DOI: 10.3390/vaccines11030635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/14/2023] Open
Abstract
The viral family Arenaviridae contains several members that cause severe, and often lethal, diseases in humans. Several highly pathogenic arenaviruses are classified as Risk Group 4 agents and must be handled in the highest biological containment facility, biosafety level-4 (BSL-4). Vaccines and treatments are very limited for these pathogens. The development of vaccines is crucial for the establishment of countermeasures against highly pathogenic arenavirus infections. While several vaccine candidates have been investigated, there are currently no approved vaccines for arenavirus infection except for Candid#1, a live-attenuated Junin virus vaccine only licensed in Argentina. Current platforms under investigation for use include live-attenuated vaccines, recombinant virus-based vaccines, and recombinant proteins. We summarize here the recent updates of vaccine candidates against arenavirus infections.
Collapse
Affiliation(s)
- Takeshi Saito
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Rachel A. Reyna
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Satoshi Taniguchi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kirsten Littlefield
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Junki Maruyama
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
8
|
Design, Synthesis, and Biological Evaluation of Benzimidazole Derivatives as Potential Lassa Virus Inhibitors. Molecules 2023; 28:molecules28041579. [PMID: 36838567 PMCID: PMC9963587 DOI: 10.3390/molecules28041579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
The Lassa virus (LASV) causes Lassa fever, a highly infectious and lethal agent of acute viral hemorrhagic fever. At present, there are still no effective treatments available, creating an urgent need to develop novel therapeutics. Some benzimidazole compounds targeting the arenavirus envelope glycoprotein complex (GPC) are promising inhibitors of LASV. In this study, we synthesized two series of LASV inhibitors based on the benzimidazole structure. Lentiviral pseudotypes bearing the LASV GPC were established to identify virus entry inhibitors. Surface plasmon resonance (SPR) was further used to verify the binding activities of the potential compounds. Compounds 7d-Z, 7h-Z, 13c, 13d, and 13f showed relatively excellent antiviral activities with IC50 values ranging from 7.58 to 15.46 nM and their SI values above 1251. These five representative compounds exhibited stronger binding affinity with low equilibrium dissociation constants (KD < 8.25 × 10-7 M) in SPR study. The compound 7h-Z displayed the most potent antiviral activity (IC50 = 7.58 nM) with a relatively high SI value (2496), which could be further studied as a lead compound. The structure-activity relationship indicated that the compounds with lipophilic and spatially larger substituents might possess higher antiviral activity and a much larger safety margin. This study will provide some good guidance for the development of highly active compounds with a novel skeleton against LASV.
Collapse
|
9
|
Abstract
Lassa virus (LASV) is endemic in the rodent populations of Sierra Leone, Nigeria and other countries in West Africa. Spillover to humans occurs frequently and results in Lassa fever, a viral haemorrhagic fever (VHF) associated with a high case fatality rate. Despite advances, fundamental gaps in knowledge of the immunology, epidemiology, ecology and pathogenesis of Lassa fever persist. More frequent outbreaks, the potential for further geographic expansion of Mastomys natalensis and other rodent reservoirs, the ease of procurement and possible use and weaponization of LASV, the frequent importation of LASV to North America and Europe, and the emergence of novel LASV strains in densely populated West Africa have driven new initiatives to develop countermeasures for LASV. Although promising candidates are being evaluated, as yet there are no approved vaccines or therapeutics for human use. This Review discusses the virology of LASV, the clinical course of Lassa fever and the progress towards developing medical countermeasures.
Collapse
Affiliation(s)
- Robert F Garry
- Department of Microbiology and Immunology, Tulane University, New Orleans, LA, USA.
- Zalgen Labs, Frederick, MD, USA.
- Global Viral Network, Baltimore, MD, USA.
| |
Collapse
|
10
|
Abstract
The emergence of drug resistance during antimicrobial therapy is a major global health problem, especially for chronic infections like human immunodeficiency virus, hepatitis B and C, and tuberculosis. Sub-optimal adherence to long-term treatment is an important contributor to resistance risk. New long-acting drugs are being developed for weekly, monthly or less frequent dosing to improve adherence, but may lead to long-term exposure to intermediate drug levels. In this study, we analyse the effect of dosing frequency on the risk of resistance evolving during time-varying drug levels. We find that long-acting therapies can increase, decrease or have little effect on resistance, depending on the source (pre-existing or de novo) and degree of resistance, and rates of drug absorption and clearance. Long-acting therapies with rapid drug absorption, slow clearance and strong wild-type inhibition tend to reduce resistance caused by partially resistant strains in the early stages of treatment even if they do not improve adherence. However, if subpopulations of microbes persist and can reactivate during sub-optimal treatment, longer-acting therapies may substantially increase the resistance risk. Our results show that drug kinetics affect selection for resistance in a complicated manner, and that pathogen-specific models are needed to evaluate the benefits of new long-acting therapies.
Collapse
Affiliation(s)
- Anjalika Nande
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA 02138, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Alison L. Hill
- Program for Evolutionary Dynamics, Harvard University, Cambridge, MA 02138, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
11
|
Abstract
Lassa Fever (LF) is a viral hemorrhagic fever endemic in West Africa. LF begins with flu-like symptoms that are difficult to distinguish from other common endemic diseases such as malaria, dengue, and yellow fever making it hard to diagnose clinically. Availability of a rapid diagnostic test and other serological and molecular assays facilitates accurate diagnosis of LF. Lassa virus therapeutics are currently in different stages of preclinical development. Arevirumab, a cocktail of monoclonal antibodies, demonstrates a great safety and efficacy profile in non-human primates. Major efforts have been made in the development of a Lassa virus vaccine. Two vaccine candidates, MeV-NP and pLASV-GPC are undergoing evaluation in phase I clinical trials.
Collapse
Affiliation(s)
- Lilia I Melnik
- Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70118, USA.
| |
Collapse
|
12
|
Marlin R, Desjardins D, Contreras V, Lingas G, Solas C, Roques P, Naninck T, Pascal Q, Behillil S, Maisonnasse P, Lemaitre J, Kahlaoui N, Delache B, Pizzorno A, Nougairede A, Ludot C, Terrier O, Dereuddre-Bosquet N, Relouzat F, Chapon C, Ho Tsong Fang R, van der Werf S, Rosa Calatrava M, Malvy D, de Lamballerie X, Guedj J, Le Grand R. Antiviral efficacy of favipiravir against Zika and SARS-CoV-2 viruses in non-human primates. Nat Commun 2022; 13:5108. [PMID: 36042198 PMCID: PMC9427089 DOI: 10.1038/s41467-022-32565-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/05/2022] [Indexed: 12/02/2022] Open
Abstract
The COVID-19 pandemic has exemplified that rigorous evaluation in large animal models is key for translation from promising in vitro results to successful clinical implementation. Among the drugs that have been largely tested in clinical trials but failed so far to bring clear evidence of clinical efficacy is favipiravir, a nucleoside analogue with large spectrum activity against several RNA viruses in vitro and in small animal models. Here, we evaluate the antiviral activity of favipiravir against Zika or SARS-CoV-2 virus in cynomolgus macaques. In both models, high doses of favipiravir are initiated before infection and viral kinetics are evaluated during 7 to 15 days after infection. Favipiravir leads to a statistically significant reduction in plasma Zika viral load compared to untreated animals. However, favipiravir has no effects on SARS-CoV-2 viral kinetics, and 4 treated animals have to be euthanized due to rapid clinical deterioration, suggesting a potential role of favipiravir in disease worsening in SARS-CoV-2 infected animals. To summarize, favipiravir has an antiviral activity against Zika virus but not against SARS-CoV-2 infection in the cynomolgus macaque model. Our results support the clinical evaluation of favipiravir against Zika virus but they advocate against its use against SARS-CoV-2 infection. Repurposed antiviral drugs present as a valuable resource in the defence during outbreaks, with rigorous evaluation in large animal models keys for translation to clinical implementation. Here, the authors explore the antiviral activity of favipiravir against Zika virus and SARS-CoV-2 in cynomolgus macaques, in order to support future clinical investigations into this RNA polymerase inhibitor.
Collapse
Affiliation(s)
- Romain Marlin
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Delphine Desjardins
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Vanessa Contreras
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | | | - Caroline Solas
- Aix-Marseille Univ, APHM, Unité des Virus Emergents (UVE) IRD 190, INSERM 1207, Laboratoire de Pharmacocinétique et Toxicologie, Hôpital La Timone, 13005, Marseille, France
| | - Pierre Roques
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France.,Virology Unit, Institut Pasteur de Guinée, Conakry, Guinée
| | - Thibaut Naninck
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Quentin Pascal
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Sylvie Behillil
- Unité de Génétique Moléculaire des Virus à ARN, GMVR, Institut Pasteur, UMR CNRS 3569, Université de Paris, Paris, France.,Centre National de Référence des Virus des infections respiratoires (dont la grippe), Institut Pasteur, Paris, France
| | - Pauline Maisonnasse
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Julien Lemaitre
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Nidhal Kahlaoui
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Benoit Delache
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Andrés Pizzorno
- CIRI, Centre International de Recherche en Infectiologie, (Team VirPath), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Antoine Nougairede
- Unité des Virus Emergents, UVE: Aix Marseille Univ, IRD 190, INSERM 1207, IHU Méditerranée Infection, 13005, Marseille, France
| | - Camille Ludot
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Olivier Terrier
- CIRI, Centre International de Recherche en Infectiologie, (Team VirPath), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Francis Relouzat
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Catherine Chapon
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Raphael Ho Tsong Fang
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Sylvie van der Werf
- Unité de Génétique Moléculaire des Virus à ARN, GMVR, Institut Pasteur, UMR CNRS 3569, Université de Paris, Paris, France.,Centre National de Référence des Virus des infections respiratoires (dont la grippe), Institut Pasteur, Paris, France
| | - Manuel Rosa Calatrava
- CIRI, Centre International de Recherche en Infectiologie, (Team VirPath), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France.,VirNext, Université Claude Bernard Lyon 1, Faculté de Médecine Laennec, Lyon, France
| | - Denis Malvy
- Department of infectious ad tropical diseases, University hopsital, Bordeaux & Inserm 1219/IRD, Bordeaux University, Bordeaux, France
| | - Xavier de Lamballerie
- Unité des Virus Emergents, UVE: Aix Marseille Univ, IRD 190, INSERM 1207, IHU Méditerranée Infection, 13005, Marseille, France
| | - Jeremie Guedj
- Université de Paris, INSERM, IAME, F-75018, Paris, France.
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases » (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France.
| |
Collapse
|
13
|
Zadeh VR, Afowowe TO, Abe H, Urata S, Yasuda J. Potential and action mechanism of favipiravir as an antiviral against Junin virus. PLoS Pathog 2022; 18:e1010689. [PMID: 35816544 PMCID: PMC9302769 DOI: 10.1371/journal.ppat.1010689] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 07/21/2022] [Accepted: 06/19/2022] [Indexed: 12/26/2022] Open
Abstract
Favipiravir is a nucleoside analogue that inhibits the replication and transcription of a broad spectrum of RNA viruses, including pathogenic arenaviruses. In this study, we isolated a favipiravir-resistant mutant of Junin virus (JUNV), which is the causative agent of Argentine hemorrhagic fever, and analyzed the antiviral mechanism of favipiravir against JUNV. Two amino acid substitutions, N462D in the RNA-dependent RNA polymerase (RdRp) and A168T in the glycoprotein precursor GPC, were identified in the mutant. GPC-A168T substitution enhanced the efficiency of JUNV internalization, which explains the robust replication kinetics of the mutant in the virus growth analysis. Although RdRp-N462D substitution did not affect polymerase activity levels in a minigenome system, comparisons of RdRp error frequencies showed that the virus with RdRp-D462 possessed a significantly higher fidelity. Our next generation sequence (NGS) analysis showed a gradual accumulation of both mutations as we passaged the virus in presence of favipiravir. We also provided experimental evidence for the first time that favipiravir inhibited JUNV through the accumulation of transition mutations, confirming its role as a purine analogue against arenaviruses. Moreover, we showed that treatment with a combination of favipiravir and either ribavirin or remdesivir inhibited JUNV replication in a synergistic manner, blocking the generation of the drug-resistant mutant. Our findings provide new insights for the clinical management and treatment of Argentine hemorrhagic fever. Development of antivirals requires cautious and extensive assessment of action mechanism as well as potential for emergence of resistant phenotype of the virus. In recent years, favipiravir has been put forward as a promising candidate for the treatment of Argentine hemorrhagic fever (AHF) caused by Junin virus (JUNV). We, therefore, aimed to provide experimental evidence on action mechanism of favipiravir to help guide its clinical use. Here we show that favipiravir causes lethal mutation that impairs virus infectivity. More importantly, we demonstrate that the virus has the capability to escape favipiravir selective pressure by acquiring two amino acid substitutions on glycoprotein precursor and polymerase proteins. This observation raises concern over the use of only favipiravir in therapeutic regimens. To overcome this risk, we show that combination of favipiravir with other nucleoside analogues demonstrates a synergistic effect and suppresses the ability of JUNV to escape drug pressure. Favipiravir, ribavirin, and remdesivir have a broad spectrum of antiviral activity. Therefore, combination therapies of these drugs would be expected to have potential therapeutic effects for not only AHF but also the diseases caused by a variety of viruses, including emerging RNA viruses.
Collapse
Affiliation(s)
- Vahid Rajabali Zadeh
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Tosin Oladipo Afowowe
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Haruka Abe
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Shuzo Urata
- Department of Emerging Infectious Diseases, National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, Nagasaki, Japan
| | - Jiro Yasuda
- Department of Emerging Infectious Diseases, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
- Program for Nurturing Global Leaders in Tropical and Emerging Communicable Diseases, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Emerging Infectious Diseases, National Research Center for the Control and Prevention of Infectious Diseases (CCPID), Nagasaki University, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
14
|
Recent Trends in Protective Textiles against Biological Threats: A Focus on Biological Warfare Agents. Polymers (Basel) 2022; 14:polym14081599. [PMID: 35458353 PMCID: PMC9026340 DOI: 10.3390/polym14081599] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 02/07/2023] Open
Abstract
The rising threats to worldwide security (affecting the military, first responders, and civilians) urge us to develop efficient and versatile technological solutions to protect human beings. Soldiers, medical personnel, firefighters, and law enforcement officers should be adequately protected, so that their exposure to biological warfare agents (BWAs) is minimized, and infectious microorganisms cannot be spread so easily. Current bioprotective military garments include multilayered fabrics integrating activated carbon as a sorptive agent and a separate filtrating layer for passive protection. However, secondary contaminants emerge following their accumulation within the carbon filler. The clothing becomes too heavy and warm to wear, not breathable even, preventing the wearer from working for extended hours. Hence, a strong need exists to select and/or create selectively permeable layered fibrous structures with bioactive agents that offer an efficient filtering capability and biocidal skills, ensuring lightweightness, comfort, and multifunctionality. This review aims to showcase the main possibilities and trends of bioprotective textiles, focusing on metal-organic frameworks (MOFs), inorganic nanoparticles (e.g., ZnO-based), and organic players such as chitosan (CS)-based small-scale particles and plant-derived compounds as bioactive agents. The textile itself should be further evaluated as the foundation for the barrier effect and in terms of comfort. The outputs of a thorough, standardized characterization should dictate the best elements for each approach.
Collapse
|
15
|
Raabe V, Mehta AK, Evans JD. Lassa Virus Infection: a Summary for Clinicians. Int J Infect Dis 2022; 119:187-200. [PMID: 35395384 DOI: 10.1016/j.ijid.2022.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES This summary on Lassa virus (LASV) infection and Lassa fever disease (LF) was developed from a clinical perspective to provide clinicians a condensed, accessible understanding of the current literature. The information provided highlights pathogenesis, clinical features, and diagnostics with an emphasis on therapies and vaccines that have demonstrated potential value for use in clinical or research environments. METHODS An integrative literature review was conducted on the clinical and pathological features, vaccines, and treatments for LASV infection, with a focus on recent studies and in vivo evidence from humans and/or non-human primates (NHPs), when available. RESULTS Two antiviral medications with potential benefit for the treatment of LASV infection and one for post-exposure prophylaxis were identified, although a larger number of potential candidates are currently being evaluated. Multiple vaccine platforms are in pre-clinical development for LASV prevention, but data from human clinical trials are not yet available. CONCLUSION We provide succinct summaries of medical countermeasures against LASV to give the busy clinician a rapid reference. Although there are no approved drugs or vaccines for LF, we provide condensed information from a literature review for measures that can be taken when faced with a suspected infection, including investigational treatment options and hospital engineering controls.
Collapse
Affiliation(s)
- Vanessa Raabe
- New York University Grossman School of Medicine, New York, NY.
| | | | - Jared D Evans
- Johns Hopkins Applied Physics Laboratory, Laurel, MD.
| |
Collapse
|
16
|
Joseph AA, Fasipe OJ, Joseph OA, Olatunji OA. Contemporary and emerging pharmacotherapeutic agents for the treatment of Lassa viral haemorrhagic fever disease. J Antimicrob Chemother 2022; 77:1525-1531. [PMID: 35296886 DOI: 10.1093/jac/dkac064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This review was designed to discuss the emerging and current pharmacotherapeutic agents for the treatment of Lassa viral haemorrhagic fever disease (LVHFD), also known as Lassa fever (LF). Original peer-reviewed articles that investigated LF were identified using the Medline Entrez-PubMed search. Information was also sourced from printed textbooks and reports by recognized health professional bodies such as the WHO, CDC, the Nigerian Federal Ministry of Health and the United Nations Children's Fund (UNICEF). A total of 103 articles were reviewed and 78 were found to contain information relevant to the study. LF remains an endemic disease of public health concern in the West Africa region, and in the rest of the world as cases have been imported into non-endemic regions as well. Currently, there are no approved vaccines or therapeutics for the treatment of Lassa mammarenavirus (LASV) infection. There are, however, off-label therapeutics being used (ribavirin and convalescent plasma) whose efficacy is suboptimal. Research is still ongoing on possible therapeutic options and drug repurposing of therapeutic agents currently in use for other clinical conditions. Considered therapeutic options include favipiravir, taribavirin, Arevirumab-3 and experimental drugs such as losmapimod, adamantyl diphenyl piperazine 3.3, Arbidol (umifenovir) and decanoyl-RRLL-chloromethyl ketone (dec-RRLL-CMK). Current treatments for LF are limited, hence the institution of mitigating measures to prevent infection is of utmost importance and should be prioritized, especially in endemic regions. Heightened searches for other therapeutic options with greater efficacy and lower toxicity are still ongoing, as well as for vaccines as the absence of these classifies the disease as a priority disease of high public health impact.
Collapse
Affiliation(s)
| | - Olumuyiwa John Fasipe
- Department of Pharmacology and Therapeutics, University of Medical Sciences, Ondo, Nigeria
| | | | - Olalekan Aliu Olatunji
- Department of Medical Microbiology and Parasitology, University College Hospital, Ibadan, Nigeria
| |
Collapse
|
17
|
Salam AP, Duvignaud A, Jaspard M, Malvy D, Carroll M, Tarning J, Olliaro PL, Horby PW. Ribavirin for treating Lassa fever: A systematic review of pre-clinical studies and implications for human dosing. PLoS Negl Trop Dis 2022; 16:e0010289. [PMID: 35353804 PMCID: PMC9000057 DOI: 10.1371/journal.pntd.0010289] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 04/11/2022] [Accepted: 02/28/2022] [Indexed: 12/24/2022] Open
Abstract
Ribavirin is currently the standard of care for treating Lassa fever. However, the human clinical trial data supporting its use suffer from several serious flaws that render the results and conclusions unreliable. We performed a systematic review of available pre-clinical data and human pharmacokinetic data on ribavirin in Lassa. In in-vitro studies, the EC50 of ribavirin ranged from 0.6 μg/ml to 21.72 μg/ml and the EC90 ranged from 1.5 μg/ml to 29 μg/ml. The mean EC50 was 7 μg/ml and the mean EC90 was 15 μg/ml. Human PK data in patients with Lassa fever was sparse and did not allow for estimation of concentration profiles or pharmacokinetic parameters. Pharmacokinetic modelling based on healthy human data suggests that the concentration profiles of current ribavirin regimes only exceed the mean EC50 for less than 20% of the time and the mean EC90 for less than 10% of the time, raising the possibility that the current ribavirin regimens in clinical use are unlikely to reliably achieve serum concentrations required to inhibit Lassa virus replication. The results of this review highlight serious issues with the evidence, which, by today standards, would be unlikely to support the transition of ribavirin from pre-clinical studies to human clinical trials. Additional pre-clinical studies are needed before embarking on expensive and challenging clinical trials of ribavirin in Lassa fever.
Collapse
Affiliation(s)
- Alex P. Salam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- United Kingdom Public Health Rapid Support Team, London, United Kingdom
| | - Alexandre Duvignaud
- Department of Infectious Diseases and Tropical Medicine, Division of Tropical Medicine and Clinical International Health, CHU de Bordeaux, Bordeaux, France
- UMR1219, INSERM, French National Research Institute for Sustainable Development (IRD), and University of Bordeaux, Bordeaux, France
- Programme PAC-CI/ANRS Research Center, CHU de Treichville, Abidjan, Côte d’Ivoire
| | - Marie Jaspard
- UMR1219, INSERM, French National Research Institute for Sustainable Development (IRD), and University of Bordeaux, Bordeaux, France
- Programme PAC-CI/ANRS Research Center, CHU de Treichville, Abidjan, Côte d’Ivoire
- Alliance for International Medical Action, Dakar, Senegal
| | - Denis Malvy
- Department of Infectious Diseases and Tropical Medicine, Division of Tropical Medicine and Clinical International Health, CHU de Bordeaux, Bordeaux, France
- UMR1219, INSERM, French National Research Institute for Sustainable Development (IRD), and University of Bordeaux, Bordeaux, France
- Programme PAC-CI/ANRS Research Center, CHU de Treichville, Abidjan, Côte d’Ivoire
| | - Miles Carroll
- Wellcome Center for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Joel Tarning
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Piero L. Olliaro
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Peter W. Horby
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
18
|
Thielebein A, Ighodalo Y, Taju A, Olokor T, Omiunu R, Esumeh R, Ebhodaghe P, Ekanem A, Igenegbale G, Giwa R, Renevey A, Hinzmann J, Müller J, Pallasch E, Pahlmann M, Guedj J, Nwatuzor J, Femi Babatunde O, Adomeh DI, Asogun D, Akpede N, Okogbenin S, Günther S, Oestereich L, Duraffour S, Ogbaini-Emovon E. Virus persistence after recovery from acute Lassa fever in Nigeria: a 2-year interim analysis of a prospective longitudinal cohort study. THE LANCET MICROBE 2022; 3:e32-e40. [DOI: 10.1016/s2666-5247(21)00178-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/22/2021] [Accepted: 06/30/2021] [Indexed: 12/13/2022] Open
|
19
|
Breuil L, Debache A, Giraud-Sauveur F, Glascott-Jones E. [A further step against Lassa fever?]. Med Sci (Paris) 2021; 37:1076-1079. [PMID: 34851291 DOI: 10.1051/medsci/2021169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Lucas Breuil
- École normale supérieure de Lyon, Département de biologie, Master biologie, Lyon, France
| | - Auriane Debache
- École normale supérieure de Lyon, Département de biologie, Master biologie, Lyon, France
| | - Félicie Giraud-Sauveur
- École normale supérieure de Lyon, Département de biologie, Master biologie, Lyon, France
| | - Eleanor Glascott-Jones
- École normale supérieure de Lyon, Département de biologie, Master biologie, Lyon, France
| |
Collapse
|
20
|
Murphy HL, Ly H. Pathogenicity and virulence mechanisms of Lassa virus and its animal modeling, diagnostic, prophylactic, and therapeutic developments. Virulence 2021; 12:2989-3014. [PMID: 34747339 PMCID: PMC8923068 DOI: 10.1080/21505594.2021.2000290] [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] [Indexed: 11/24/2022] Open
Abstract
Lassa fever (LF) is a deadly viral hemorrhagic disease that is endemic to West Africa. The causative agent of LF is Lassa virus (LASV), which causes approximately 300,000 infections and 5,000 deaths annually. There are currently no approved therapeutics or FDA-approved vaccines against LASV. The high genetic variability between LASV strains and immune evasion mediated by the virus complicate the development of effective therapeutics and vaccines. Here, we aim to provide a comprehensive review of the basic biology of LASV and its mechanisms of disease pathogenesis and virulence in various animal models, as well as an update on prospective vaccines, therapeutics, and diagnostics for LF. Until effective vaccines and/or therapeutics are available for use to prevent or treat LF, a better level of understanding of the basic biology of LASV, its natural genetic variations and immune evasion mechanisms as potential pathogenicity factors, and of the rodent reservoir-vector populations and their geographical distributions, is necessary for the development of accurate diagnostics and effective therapeutics and vaccines against this deadly human viral pathogen.
Collapse
Affiliation(s)
- Hannah L Murphy
- Department of Veterinary & Biomedical Sciences, Comparative & Molecular Biosciences Graduate Program, College of Veterinary Medicine, University of Minnesota, Twin Cities
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, Comparative & Molecular Biosciences Graduate Program, College of Veterinary Medicine, University of Minnesota, Twin Cities
| |
Collapse
|
21
|
Hansen F, Jarvis MA, Feldmann H, Rosenke K. Lassa Virus Treatment Options. Microorganisms 2021; 9:microorganisms9040772. [PMID: 33917071 PMCID: PMC8067676 DOI: 10.3390/microorganisms9040772] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 12/27/2022] Open
Abstract
Lassa fever causes an approximate 5000 to 10,000 deaths annually in West Africa and cases have been imported into Europe and the Americas, challenging public health. Although Lassa virus was first described over 5 decades ago in 1969, no treatments or vaccines have been approved to treat or prevent infection. In this review, we discuss current therapeutics in the development pipeline for the treatment of Lassa fever, focusing on those that have been evaluated in humans or animal models. Several treatments, including the antiviral favipiravir and a human monoclonal antibody cocktail, have shown efficacy in preclinical rodent and non-human primate animal models and have potential for use in clinical settings. Movement of the promising preclinical treatment options for Lassa fever into clinical trials is critical to continue addressing this neglected tropical disease.
Collapse
Affiliation(s)
- Frederick Hansen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Michael A Jarvis
- The Vaccine Group Ltd., University of Plymouth, Plymouth PL4 8AA, UK
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Kyle Rosenke
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| |
Collapse
|