1
|
Letafati A, Fakhr SSH, Najafabadi AQ, Karami N, Karami H. Marburg Virus Disease: A Narrative Review. Health Sci Rep 2025; 8:e70669. [PMID: 40330770 PMCID: PMC12053247 DOI: 10.1002/hsr2.70669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/22/2025] [Accepted: 03/21/2025] [Indexed: 05/08/2025] Open
Abstract
Background and Aims Given the recent deadly outbreaks of the Marburg virus (MARV), in early 2023 in Tanzania and Equatorial Guinea, and the most recent one in Rwanda in 2024, there has been renewed attention across Africa on the threat posed by the re-emergence of MARV as a growing concern for public health. Therefore, it needs to provide a comprehensive overview of the virus and its related infections, encompassing virus classification, historical outbreaks, transmission dynamics, the intricate interface between the virus and its hosts, the methods of diagnosis, core prevention strategies, and current therapeutic options, to better understand the virus and the disease characteristics in responding to future outbreaks. Methods For this review, four scientific online databases, including PubMed, Google Scholar, Scopus, and Web of Science were thoroughly searched for peer-reviewed journal papers (original, case reports/series, and review studies) published in English language using the following keywords: Filovirus, Marburg virus, Marburg Haemorrhagic Fever, Marburg virus disease, and Marburg virus outbreak. Results MARV shares similarities with its close cousin -the Ebola virus [EBOV]-in terms of viral characteristics and most clinical features. These two viruses are of animal origin and primarily spread to humans through infected bats (both direct and indirect close contact), which serve as the common natural host reservoirs. The potential for interhuman transmission, coupled with the ability to cross borders of endemic regions combined with the absence of a licensed vaccine and effective treatment, have made MARV a significant threat to human health. This virus is clinically characterized by a range of symptoms and organ dysfunctions. The disease is often fatal in a significant proportion of infected individuals. This viral infection is diagnosed by various diagnostic tools, prevented mainly through personal protective measures, and treated usually with clinical management and supportive care. Conclusion The outbreaks of MARV are continuously threaten public health; therefore, the world must be alert and well-prepared. For MVD, taking precautions along with investing in research and preparedness at regional, national, and global levels is of crucial importance and should be prioritized.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, Faculty of Public HealthTehran University of Medical Sciences (TUMS)TehranIran
| | | | - Ali Qaraee Najafabadi
- Department of Biochemistry, Faculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Negin Karami
- Department of Nursing, Faculty of NursingAlborz University of Medical SciencesKarajIran
| | - Hassan Karami
- Department of Virology, Faculty of Public HealthTehran University of Medical Sciences (TUMS)TehranIran
| |
Collapse
|
2
|
Prasad AN, Woolsey C, Borisevich V, Agans KN, Deer DJ, Geisbert JB, Harrison MB, Dobias NS, Fenton KA, Cross RW, Geisbert TW. Remdesivir, mAb114, REGN-EB3, and ZMapp partially rescue nonhuman primates infected with a low passage Kikwit variant of Ebola virus. Nat Commun 2025; 16:3824. [PMID: 40268932 PMCID: PMC12019533 DOI: 10.1038/s41467-025-59168-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/12/2025] [Indexed: 04/25/2025] Open
Abstract
In 2018, a clinical trial of four investigational therapies for Ebola virus disease (EVD), known as the PALM trial, was conducted in the Democratic Republic of Congo. All patients received either the antiviral remdesivir (RDV) or a monoclonal antibody product: ZMapp, mAb114 (Ebanga), or REGN-EB3 (Inmazeb). The study concluded that both mAb114 and REGN-EB3 were superior to ZMapp and RDV in reducing mortality from EVD. However, the data suggested that some patients in the RDV and ZMapp groups might have been sicker at the time of treatment initiation. Here, we assessed the efficacy of each of these therapies in a uniformly lethal rhesus monkey model of EVD when treatment was initiated 5 days after Ebola exposure. Treatment with RDV, mAb114, REGN-EB3, and ZMapp each resulted in similar survival (approximately 40%). Survival was associated with circulating viral load at treatment initiation. A trend of more escape mutants in the GP1 and GP2 domains was observed for the mAb114 group. Our data show similar suboptimal efficacy of individual therapeutics in the uniformly lethal NHP model of EVD, supporting further clinical investigation of therapeutic combinations to maximize the overall therapeutic effect and improve patient outcomes, particularly for the treatment of advanced stage EVD.
Collapse
MESH Headings
- Animals
- Hemorrhagic Fever, Ebola/drug therapy
- Hemorrhagic Fever, Ebola/virology
- Hemorrhagic Fever, Ebola/mortality
- Ebolavirus/drug effects
- Ebolavirus/genetics
- Ebolavirus/immunology
- Adenosine Monophosphate/analogs & derivatives
- Adenosine Monophosphate/therapeutic use
- Adenosine Monophosphate/pharmacology
- Macaca mulatta
- Alanine/analogs & derivatives
- Alanine/therapeutic use
- Alanine/pharmacology
- Antiviral Agents/therapeutic use
- Antiviral Agents/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/pharmacology
- Disease Models, Animal
- Humans
- Female
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Male
Collapse
Affiliation(s)
- Abhishek N Prasad
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Courtney Woolsey
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Daniel J Deer
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Joan B Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mack B Harrison
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Natalie S Dobias
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Karla A Fenton
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
3
|
Taha MS, Akram A, Abdelbary GA. Unlocking the potential of remdesivir: innovative approaches to drug delivery. Drug Deliv Transl Res 2025:10.1007/s13346-025-01843-7. [PMID: 40244526 DOI: 10.1007/s13346-025-01843-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/19/2025] [Indexed: 04/18/2025]
Abstract
Given the recurrent waves of COVID-19 and the emergence of new viral infections, optimizing the potential of remdesivir as an antiviral agent is critical. While several reviews have explored the efficacy of remdesivir, few have comprehensively addressed its challenges, such as the necessity for intravenous infusion, suboptimal lung accumulation, and safety concerns related to its formulation. This review critically examines these challenges while proposing innovative solutions and effective combinations with other antiviral agents and repurposed drugs. By highlighting the role of complex generics, we aim to enhance therapeutic efficacy in ways not previously discussed in existing literature. Furthermore, we address the development of novel drug delivery systems which specifically aim to improve remdesivir's pharmacological profile. By analyzing recent findings, we assess both the successes and limitations of current approaches, providing insights into ongoing challenges and strategies for further optimization. This review uniquely focuses on targeted drug delivery systems and innovative formulations, thereby maximizing remdesivir's therapeutic benefits and broadening its application in combating emerging viral threats. In doing so, we fill a critical gap in literature, offering a comprehensive overview that informs future research and clinical strategies.
Collapse
Affiliation(s)
- Maie S Taha
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Alaa Akram
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ghada A Abdelbary
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
4
|
Gerresheim GK, Kraft F, Werner AD, Biedenkopf N. Semi-automated diagnostic RT-PCR as a screening assay for antiviral compounds in a 96-well format against highly pathogenic RNA viruses. Adv Virus Res 2025; 121:101-122. [PMID: 40379381 DOI: 10.1016/bs.aivir.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2025]
Abstract
In response to outbreaks of (re)emerging highly pathogenic RNA viruses, simple and scalable antiviral screening methods are urgently needed. Using established and validated diagnostic methods like RT-PCR for antiviral screening offers a rapid readout of viral replication. This becomes particular important when other traditional viral replication readouts, such as TCID50 or plaque assays cannot be used due to the absence of cytopathic effects, lack of reporter gene-containing recombinant viruses or unavailability of appropriate antibodies - the latter two common challenges when so far unknown viruses emerge. This study evaluated semi-automated diagnostic RT-PCR in a 96-well approach for antiviral compound screening using Marburg virus serving as a case study. Remdesivir, a prodrug that exhibits antiviral activities against multiple RNA viruses, was used as positive control inhibiting replication of filoviruses. Applicability of the protocol to other members of the filovirus family was feasible using the same settings, while for other viruses like Middle East respiratory syndrome coronavirus (MERS-CoV) or Crimean-Congo hemorrhagic fever virus (CCHFV) adaptations to optimal infection settings were necessary. Our results demonstrate a high reproducibility and highlight the rapid adaptability of semi-automated RT-PCR assays as an accelerated antiviral screening assay with high scalability against a wide range of newly or (re)emerging RNA viruses. This is critical especially during outbreak situations where timely antiviral assessments are urgently needed.
Collapse
Affiliation(s)
- Gesche K Gerresheim
- Institute of Virology, Philipps University Marburg, Marburg, Germany; German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, Marburg, Germany
| | - Franziska Kraft
- Institute of Virology, Philipps University Marburg, Marburg, Germany; German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, Marburg, Germany
| | | | - Nadine Biedenkopf
- Institute of Virology, Philipps University Marburg, Marburg, Germany; German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, Marburg, Germany.
| |
Collapse
|
5
|
Rahim FO, Fallah M, Muvunyi CM, Abraham L, Nsanzimana S, Kaseya J. Rwanda's science-led response to Marburg virus holds lessons for other outbreaks in Africa. Nat Med 2025; 31:1055-1056. [PMID: 40140621 DOI: 10.1038/s41591-025-03551-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Affiliation(s)
| | - Mosoka Fallah
- Africa Centres for Disease Control Prevention, Addis Ababa, Ethiopia
| | | | | | | | - Jean Kaseya
- Africa Centres for Disease Control Prevention, Addis Ababa, Ethiopia.
| |
Collapse
|
6
|
Cross RW, Woolsey C, Prasad AN, Borisevich V, Agans KN, Deer DJ, Harrison MB, Dobias NS, Fenton KA, Cihlar T, Nguyen AQ, Babusis D, Bannister R, Vermillion MS, Chu VC, Geisbert TW. Oral obeldesivir provides postexposure protection against Marburg virus in nonhuman primates. Nat Med 2025; 31:1303-1311. [PMID: 39805309 PMCID: PMC12003170 DOI: 10.1038/s41591-025-03496-y] [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: 11/07/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
The recent outbreak of Marburg virus (MARV) in Rwanda underscores the need for effective countermeasures against this highly fatal pathogen, with case fatality rates reaching 90%. Currently, no vaccines or approved treatments exist for MARV infection, distinguishing it from related viruses such as Ebola. Our study demonstrates that the oral drug obeldesivir (ODV), a nucleoside analog prodrug, shows promising antiviral activity against filoviruses in vitro and offers significant protection in animal models. Here with cynomolgus macaques (n = 6), a 10 day regimen of once-daily ODV, initiated 24 h after exposure, provided 80% protection against a thousandfold lethal MARV challenge, delaying viral replication and disease onset. Transcriptome analysis revealed that early adaptive responses correlated with successful outcomes. Compared with intravenous options, oral antivirals such as ODV offer logistical advantages in outbreak settings, enabling easier administration and broader contact coverage. Our findings support the potential of ODV as a broad-spectrum, oral postexposure prophylaxis for filoviruses.
Collapse
Affiliation(s)
- Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Courtney Woolsey
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Abhishek N Prasad
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Daniel J Deer
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mack B Harrison
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Natalie S Dobias
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Karla A Fenton
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | | | | | | | | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
7
|
Musafiri S, Siddig EE, Nkuranga JB, Rukundo A, Mpunga T, Sendegeya A, Twagirumugabe T, Ahmed A, Muvunyi CM. Emerging Strategies and Progress in the Medical Management of Marburg Virus Disease. Pathogens 2025; 14:322. [PMID: 40333077 PMCID: PMC12030108 DOI: 10.3390/pathogens14040322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 05/09/2025] Open
Abstract
During the current outbreak of Marburg virus disease (MVD) in Rwanda, we synthesized evidence from the literature to improve case management. Accordingly, experimental treatment was offered to patients under close follow-up. Remdesivir alone or in combination with monoclonal antibody treatment (MBP091) complemented with supportive care has improved the clinical outcomes of patients. Additionally, we have identified several experimental therapies currently under investigation, including antiviral drugs such as favipiravir, galidesivir, obeldesivir, and remdesivir, along with monoclonal and polyclonal antibodies (e.g., polyclonal IgG, monoclonal antibody MR-78-N; MR82-N; MR191-N; monoclonal antibodies MR186-YTE and MBP091). Furthermore, substantial progress is being made in vaccine development, with promising candidates including adenovirus-vectored vaccines, DNA vaccines, and the recombinant vesicular stomatitis virus (rVSV) vaccine. Moreover, innovative preventive and treatment strategies-such as synthetic hormones like estradiol benzoate, small interfering RNA (siRNA), interferon-β therapy, and phosphorodiamidate morpholino oligomers-are emerging as potential options for MVD management. Further investment is needed to accelerate research and optimize these therapeutics and preventive modalities. Additional epidemiological, preclinical, and clinical studies are warranted to generate the evidence required to inform policymaking, resource mobilization, and the implementation of cost-effective interventions for the prevention, control, and treatment of MVD.
Collapse
Affiliation(s)
- Sanctus Musafiri
- University Teaching Hospital of Kigali (CHUK), Kigali KN 4 Ave, Rwanda
- School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Kigali 3900, Rwanda
| | | | | | - Athanase Rukundo
- Department of Clinical Service, Ministry of Health, Kigali 84, Rwanda
| | - Tharcisse Mpunga
- University Teaching Hospital of Kigali (CHUK), Kigali KN 4 Ave, Rwanda
| | | | | | - Ayman Ahmed
- Rwanda Biomedical Centre, Kigali 7162, Rwanda
- Pan-Africa One Health Institute (PAOHI), Kigali 11KG ST203, Rwanda
| | | |
Collapse
|
8
|
Rojek A, Fieggen J, Apiyo P, Caluwaerts S, Fowler RA, Kaleebu P, Kojan R, Lado M, Lambe T, Dunning J, Horby P. Ebola disease: bridging scientific discoveries and clinical application. THE LANCET. INFECTIOUS DISEASES 2025; 25:e165-e176. [PMID: 39675368 DOI: 10.1016/s1473-3099(24)00673-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 12/17/2024]
Abstract
The west Africa Ebola disease epidemic (2014-16) marked a historic change of course for patient care during emerging infectious disease outbreaks. The epidemic response was a failure in many ways-a slow, cumbersome, and disjointed effort by a global architecture that was not fit for purpose for a rapidly spreading outbreak. In the most affected countries, health-care workers and other responders felt helpless-dealing with an overwhelming number of patients but with few, if any, tools at their disposal to provide high-quality care. These inadequacies, however, led to attention and innovation. The decade since then has seen remarkable achievements in clinical care for Ebola disease, including the approval of the first vaccines and treatments. In this paper, the first in a two-part Series, we reflect on this progress and provide expert summary of the modern landscape of Ebola disease, highlighting the priorities and ongoing activities aimed at further improving patient survival and wellbeing in the years ahead.
Collapse
Affiliation(s)
| | | | - Paska Apiyo
- Gulu Regional Referral Hospital Ministry of Health, Pece Laroo Division, Gulu City, Uganda
| | - Séverine Caluwaerts
- Medical Department, Médecins Sans Frontières, Brussels, Belgium; Institute of Tropical Medicine, Antwerp, Belgium
| | - Robert A Fowler
- Sunnybrook Health Sciences Centre, Sunnybrook Hospital, Toronto, ON, Canada
| | - Pontiano Kaleebu
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda; Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Marta Lado
- Partners In Health, Freetown, Sierra Leone
| | - Teresa Lambe
- Pandemic Sciences Institute, Oxford, UK; Oxford Vaccine Group, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | | | | |
Collapse
|
9
|
Martins KA, Wolfe DN. Marburg Virus Medical Countermeasures. Methods Mol Biol 2025; 2877:25-43. [PMID: 39585611 DOI: 10.1007/978-1-0716-4256-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Among the Filoviridae, Marburg virus (MARV) is a biological threat for which no licensed vaccines or therapeutics are currently available. In contrast, we have licensed products for Ebola virus (EBOV), another member of the Filoviridae family. The availability of licensed medical countermeasures (MCMs) for EBOV provides an opportunity to test a key objective of many pandemic preparedness plans, which is to apply some of the same technological approaches demonstrated to be successful for one agent, EBOV, to a second agent, MARV. This chapter will discuss (1) lessons learned from EBOV outbreak responses and MCM development that are applicable to MARV MCM development, (2) the likely concept of operations for using vaccines and therapeutics against MARV, and (3) target product profiles based on the concept of operations. Finally, we will discuss the current status of the MCM pipeline for MARV and next steps to advance these countermeasures to licensure.
Collapse
Affiliation(s)
- Karen A Martins
- Biomedical Advanced Research and Development Authority (BARDA), Administration for Strategic Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington, DC, USA.
| | - Daniel N Wolfe
- Biomedical Advanced Research and Development Authority (BARDA), Administration for Strategic Preparedness and Response (ASPR), U.S. Department of Health and Human Services (HHS), Washington, DC, USA
| |
Collapse
|
10
|
Tamilarasi W, Balamurugan BJ. New reverse sum Revan indices for physicochemical and pharmacokinetic properties of anti-filovirus drugs. Front Chem 2024; 12:1486933. [PMID: 39749221 PMCID: PMC11693449 DOI: 10.3389/fchem.2024.1486933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/21/2024] [Indexed: 01/04/2025] Open
Abstract
Ebola and Marburg viruses, biosafety level 4 pathogens, cause severe hemorrhaging and organ failure with high mortality. Although some FDA-approved vaccines or therapeutics like Ervebo for Zaire Ebola virus exist, still there is a lack of effective therapeutics that cover all filoviruses, including both Ebola and Marburg viruses. Therefore, some anti-filovirus drugs such as Pinocembrin, Favipiravir, Remdesivir and others are used to manage infections. In theoretical chemistry, a chemical molecule is converted into an isomorphic molecular graph, G ( V , E ) by considering atom set V as vertices and bond set E as edges. A topological index is a molecular descriptor derived from the molecular graph of a chemical compound that characterizes its topology. The relationship between a compound's chemical structure and its properties is investigated through the quantitative structure-property relationship (QSPR). This article introduces new reverse sum Revan degree based indices to explore the physicochemical and pharmacokinetic properties of anti-filovirus drugs via multilinear regression. The findings reveal a strong correlation between these proposed indices and the properties of anti-filovirus drugs when compared to reverse and Revan degree-based indices. Thus, reverse sum Revan indices offer valuable insights for analyzing the drugs properties used to treat Ebola and Marburg virus infections. Moreover, the multilinear regression (MLR) results through reverse sum Revan indices are compared with Artificial Neural Network (ANN) modelling technique and it provides the better prediction of the physicochemical and pharmacokinetic properties of anti-filovirus drugs.
Collapse
Affiliation(s)
| | - B. J. Balamurugan
- Department of Mathematics, School of Advanced Sciences, Vellore Institute of Technology, Chennai, Tamil Nadu, India
| |
Collapse
|
11
|
Zumbrun EE, Garvey CB, Wells JB, Lynn GC, Van Tongeren SA, Steffens JT, Wetzel KS, Wetzel DL, Esham HL, Garza NL, Lee ED, Scruggs JL, Rossi FD, Brown ES, Weidner JM, Gomba LM, O’Brien KA, Jay AN, Zeng X, Akers KS, Kallgren PA, Englund E, Meinig JM, Kugelman JR, Moore JL, Bloomfield HA, Norris SL, Bryan T, Scheuerell CH, Walters J, Mollova N, Blair C, Babusis D, Cihlar T, Porter DP, Singh B, Hedskog C, Bavari S, Warren TK, Bannister R. A Randomized, Blinded, Vehicle-Controlled Dose-Ranging Study to Evaluate and Characterize Remdesivir Efficacy Against Ebola Virus in Rhesus Macaques. Viruses 2024; 16:1934. [PMID: 39772240 PMCID: PMC11680158 DOI: 10.3390/v16121934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Ebola virus (EBOV) causes severe disease in humans, with mortality as high as 90%. The small-molecule antiviral drug remdesivir (RDV) has demonstrated a survival benefit in EBOV-exposed rhesus macaques. Here, we characterize the efficacy of multiple intravenous RDV dosing regimens on survival of rhesus macaques 42 days after intramuscular EBOV exposure. Thirty rhesus macaques underwent surgical implantation of telemetry devices for the fine-scale monitoring of body temperature and activity, as well as central venous catheters, to enable treatment administration and blood collection. Treatment, consisting of a loading dose of RDV followed by once-daily maintenance doses for 11 days, was initiated 4 days after virus exposure when all animals were exhibiting disease signs consistent with incipient EBOV disease as well as quantifiable levels of EBOV RNA in plasma. In the RDV treatment groups receiving loading/maintenance doses of 5/2.5 mg/kg, 10/5 mg/kg, and 20/10 mg/kg, a total of 6 of 8 (75%), 7 of 8 (87.5%), and 5 of 7 (71.4%) animals survived, respectively. In the vehicle control group, one of seven animals (14.3%) survived. The improved survival rate compared to the control group was statistically significant only for the 10/5 mg/kg RDV treatment group. This treatment regimen also resulted in a significantly lower systemic viral load compared to the vehicle control after a single RDV treatment. All three RDV regimens produced a significantly lower systemic viral load after two treatments. For most animals, RDV treatment, regardless of dose, resulted in the amelioration of many of the clinical-pathological changes associated with EBOV disease in this model.
Collapse
Affiliation(s)
- Elizabeth E. Zumbrun
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Carly B. Garvey
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Jay B. Wells
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Ginger C. Lynn
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Sean A. Van Tongeren
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Jesse T. Steffens
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Kelly S. Wetzel
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Darrell L. Wetzel
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Heather L. Esham
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Nicole L. Garza
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Eric D. Lee
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Jennifer L. Scruggs
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Franco D. Rossi
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Elizabeth S. Brown
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Jessica M. Weidner
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Laura M. Gomba
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Kristan A. O’Brien
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Alexandra N. Jay
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Kristen S. Akers
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Paul A. Kallgren
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Ethan Englund
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - J. Matthew Meinig
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Jeffrey R. Kugelman
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Joshua L. Moore
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Holly A. Bloomfield
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Sarah L. Norris
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Tameka Bryan
- PharPoint Research, Inc., Wilmington, NC 28401, USA;
| | | | - Jesse Walters
- Labcorp Early Development Laboratories, Madison, WI 53704, USA; (C.H.S.); (J.W.)
| | - Nevena Mollova
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Christiana Blair
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Darius Babusis
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Tomas Cihlar
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Danielle P. Porter
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Bali Singh
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Charlotte Hedskog
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
| | - Travis K. Warren
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.A.V.T.); (J.T.S.); (K.S.W.); (D.L.W.); (H.L.E.); (E.D.L.); (J.L.S.); (F.D.R.); (E.S.B.); (J.M.W.); (L.M.G.); (K.A.O.); (A.N.J.); (X.Z.); (K.S.A.); (P.A.K.); (E.E.); (J.M.M.); (J.R.K.); (J.L.M.); (H.A.B.); (S.L.N.); (S.B.); (T.K.W.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Roy Bannister
- Gilead Sciences, Inc., Foster City, CA 94404, USA; (N.M.); (C.B.); (D.B.); (T.C.); (D.P.P.); (B.S.); (C.H.); (R.B.)
| |
Collapse
|
12
|
Muvunyi CM, Ngabonziza JCS, Bigirimana N, Ndembi N, Siddig EE, Kaseya J, Ahmed A. Evidence-Based Guidance for One Health Preparedness, Prevention, and Response Strategies to Marburg Virus Disease Outbreaks. Diseases 2024; 12:309. [PMID: 39727639 PMCID: PMC11727285 DOI: 10.3390/diseases12120309] [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: 10/20/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/28/2024] Open
Abstract
OBJECTIVES Marburg virus disease (MVD) is on the WHO list for pandemic-prone pathogens. The current outbreak in Rwanda provides an opportunity to map outbreaks and generate information to inform policymaking, resource mobilization, and guide the implementation of cost-effective response strategies. METHODS We synthesized available information about MVD to build holistic, up-to-date evidence to inform policymakers, public health leaders, and healthcare and public health services providers in their development and implementation of cost-effective preparedness, prevention, and control measures. RESULTS We have identified 20 outbreaks of MVD that occurred in 14 countries between 1967 and 2024; these outbreaks led to 580 confirmed cases and 423 deaths in total. We summarize the available information about the main clinical signs, diagnostic tools, primary reservoir, transmission dynamics, and case management protocol. We also document the best practices in the prevention and control of MVD outbreaks, including the implementation of a multisectoral One Health strategy for preparedness, prevention, and response to MVD outbreaks that incorporates the strict implementation of WASH and infection prevention measures, contact tracing, and the isolation of infected and suspected humans and animals, and enhances the implementation of the International Health Regulations, particularly efficient cross-country coordination. CONCLUSIONS In the absence of a licensed treatment or vaccine for MVD, the response strategy to MVD should focus on preventive measures, including community engagement to promote the reduction in contact between humans and reservoirs, the supportive care and isolation of patients, and proper waste management. High risk populations such as frontline responders, including healthcare providers and community health workers, should be prioritized so that they can access all currently available protection measures.
Collapse
Affiliation(s)
| | - Jean Claude Semuto Ngabonziza
- Department of Clinical Biology, University of Rwanda, Kigali 3900, Rwanda
- Research, Innovation and Data Science Division, Rwanda Biomedical Centre, Kigali 7162, Rwanda
| | | | - Nicaise Ndembi
- The Africa Centres for Disease Control and Prevention (Africa CDC), Ring Road, 16/17, Haile Garment Lafto Square, Addis Ababa P.O. Box 3243, Ethiopia
| | - Emmanuel Edwar Siddig
- Unit of Applied Medical Sciences, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum 11111, Sudan
| | - Jean Kaseya
- The Africa Centres for Disease Control and Prevention (Africa CDC), Ring Road, 16/17, Haile Garment Lafto Square, Addis Ababa P.O. Box 3243, Ethiopia
| | - Ayman Ahmed
- Rwanda Biomedical Center (RBC), Kigali 7162, Rwanda (A.A.)
- Institute of Endemic Diseases, University of Khartoum, Khartoum 11111, Sudan
- Pan-Africa One Health Institute (PAOHI), Kigali 11KG ST203, Rwanda
| |
Collapse
|
13
|
Sidik S. Lethal Marburg virus is on the rise in Rwanda: why scientists are worried. Nature 2024; 634:522-523. [PMID: 39384913 DOI: 10.1038/d41586-024-03275-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2024]
|
14
|
de La Vega MA, XIII A, Massey CS, Spengler JR, Kobinger GP, Woolsey C. An update on nonhuman primate usage for drug and vaccine evaluation against filoviruses. Expert Opin Drug Discov 2024; 19:1185-1211. [PMID: 39090822 PMCID: PMC11466704 DOI: 10.1080/17460441.2024.2386100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Due to their faithful recapitulation of human disease, nonhuman primates (NHPs) are considered the gold standard for evaluating drugs against Ebolavirus and other filoviruses. The long-term goal is to reduce the reliance on NHPs with more ethical alternatives. In silico simulations and organoid models have the potential to revolutionize drug testing by providing accurate, human-based systems that mimic disease processes and drug responses without the ethical concerns associated with animal testing. However, as these emerging technologies are still in their developmental infancy, NHP models are presently needed for late-stage evaluation of filovirus vaccines and drugs, as they provide critical insights into the efficacy and safety of new medical countermeasures. AREAS COVERED In this review, the authors introduce available NHP models and examine the existing literature on drug discovery for all medically significant filoviruses in corresponding models. EXPERT OPINION A deliberate shift toward animal-free models is desired to align with the 3Rs of animal research. In the short term, the use of NHP models can be refined and reduced by enhancing replicability and publishing negative data. Replacement involves a gradual transition, beginning with the selection and optimization of better small animal models; advancing organoid systems, and using in silico models to accurately predict immunological outcomes.
Collapse
Affiliation(s)
- Marc-Antoine de La Vega
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Ara XIII
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Christopher S. Massey
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Jessica R. Spengler
- Viral Special Pathogens Branch and Infectious Diseases
Pathology Branch, Division of High Consequence Pathogens and Pathology, Centers for
Disease Control and Prevention, Atlanta, GA
| | - Gary P. Kobinger
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| | - Courtney Woolsey
- Galveston National Laboratory, Department of Microbiology
and Immunology, Institute for Human Infections and Immunity, University of Texas
Medical Branch, Galveston, TX, USA
| |
Collapse
|
15
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
16
|
Iketani S, Ho DD. SARS-CoV-2 resistance to monoclonal antibodies and small-molecule drugs. Cell Chem Biol 2024; 31:632-657. [PMID: 38640902 PMCID: PMC11084874 DOI: 10.1016/j.chembiol.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
Over four years have passed since the beginning of the COVID-19 pandemic. The scientific response has been rapid and effective, with many therapeutic monoclonal antibodies and small molecules developed for clinical use. However, given the ability for viruses to become resistant to antivirals, it is perhaps no surprise that the field has identified resistance to nearly all of these compounds. Here, we provide a comprehensive review of the resistance profile for each of these therapeutics. We hope that this resource provides an atlas for mutations to be aware of for each agent, particularly as a springboard for considerations for the next generation of antivirals. Finally, we discuss the outlook and thoughts for moving forward in how we continue to manage this, and the next, pandemic.
Collapse
Affiliation(s)
- Sho Iketani
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
17
|
Cross RW, Woolsey C, Chu VC, Babusis D, Bannister R, Vermillion MS, Geleziunas R, Barrett KT, Bunyan E, Nguyen AQ, Cihlar T, Porter DP, Prasad AN, Deer DJ, Borisevich V, Agans KN, Martinez J, Harrison MB, Dobias NS, Fenton KA, Bilello JP, Geisbert TW. Oral administration of obeldesivir protects nonhuman primates against Sudan ebolavirus. Science 2024; 383:eadk6176. [PMID: 38484056 DOI: 10.1126/science.adk6176] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/24/2024] [Indexed: 03/19/2024]
Abstract
Obeldesivir (ODV, GS-5245) is an orally administered prodrug of the parent nucleoside of remdesivir (RDV) and is presently in phase 3 trials for COVID-19 treatment. In this work, we show that ODV and its circulating parent nucleoside metabolite, GS-441524, have similar in vitro antiviral activity against filoviruses, including Marburg virus, Ebola virus, and Sudan virus (SUDV). We also report that once-daily oral ODV treatment of cynomolgus monkeys for 10 days beginning 24 hours after SUDV exposure confers 100% protection against lethal infection. Transcriptomics data show that ODV treatment delayed the onset of inflammation and correlated with antigen presentation and lymphocyte activation. Our results offer promise for the further development of ODV to control outbreaks of filovirus disease more rapidly.
Collapse
Affiliation(s)
- Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Courtney Woolsey
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | - Abhishek N Prasad
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Daniel J Deer
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jasmine Martinez
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Mack B Harrison
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Natalie S Dobias
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Karla A Fenton
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
18
|
Guito JC, Arnold CE, Schuh AJ, Amman BR, Sealy TK, Spengler JR, Harmon JR, Coleman-McCray JD, Sanchez-Lockhart M, Palacios GF, Towner JS, Prescott JB. Peripheral immune responses to filoviruses in a reservoir versus spillover hosts reveal transcriptional correlates of disease. Front Immunol 2024; 14:1306501. [PMID: 38259437 PMCID: PMC10800976 DOI: 10.3389/fimmu.2023.1306501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/27/2023] [Indexed: 01/24/2024] Open
Abstract
Several filoviruses, including Marburg virus (MARV), cause severe disease in humans and nonhuman primates (NHPs). However, the Egyptian rousette bat (ERB, Rousettus aegyptiacus), the only known MARV reservoir, shows no overt illness upon natural or experimental infection, which, like other bat hosts of zoonoses, is due to well-adapted, likely species-specific immune features. Despite advances in understanding reservoir immune responses to filoviruses, ERB peripheral blood responses to MARV and how they compare to those of diseased filovirus-infected spillover hosts remain ill-defined. We thus conducted a longitudinal analysis of ERB blood gene responses during acute MARV infection. These data were then contrasted with a compilation of published primate blood response studies to elucidate gene correlates of filovirus protection versus disease. Our work expands on previous findings in MARV-infected ERBs by supporting both host resistance and disease tolerance mechanisms, offers insight into the peripheral immunocellular repertoire during infection, and provides the most direct known cross-examination between reservoir and spillover hosts of the most prevalently-regulated response genes, pathways and activities associated with differences in filovirus pathogenesis and pathogenicity.
Collapse
Affiliation(s)
- Jonathan C. Guito
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Catherine E. Arnold
- Biological Defense Research Directorate, Naval Medical Research Center, Frederick, MD, United States
- RD-CBR, Research and Development Directorate, Chemical and Biological Technologies Directorate, Research Center of Excellence, Defense Threat Reduction Agency, Fort Belvoir, VA, United States
| | - Amy J. Schuh
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Brian R. Amman
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Tara K. Sealy
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jessica R. Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Jessica R. Harmon
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Joann D. Coleman-McCray
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Mariano Sanchez-Lockhart
- Center for Genome Sciences, Molecular Biology Division, U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, United States
| | - Gustavo F. Palacios
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jonathan S. Towner
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
| | - Joseph B. Prescott
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, United States
- Center for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
19
|
Alawam AS, Alawam HS, Alshahrani MM, Alwethaynani MS, Alneghery LM, Alamri MA. Structural and Dynamical Basis of VP35-RBD Inhibition by Marine Fungi Compounds to Combat Marburg Virus Infection. Mar Drugs 2024; 22:34. [PMID: 38248659 PMCID: PMC10820117 DOI: 10.3390/md22010034] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
The Marburg virus (MBV), a deadly pathogen, poses a serious threat to world health due to the lack of effective treatments, calling for an immediate search for targeted and efficient treatments. In this study, we focused on compounds originating from marine fungi in order to identify possible inhibitory compounds against the Marburg virus (MBV) VP35-RNA binding domain (VP35-RBD) using a computational approach. We started with a virtual screening procedure using the Lipinski filter as a guide. Based on their docking scores, 42 potential candidates were found. Four of these compounds-CMNPD17596, CMNPD22144, CMNPD25994, and CMNPD17598-as well as myricetin, the control compound, were chosen for re-docking analysis. Re-docking revealed that these particular compounds had a higher affinity for MBV VP35-RBD in comparison to the control. Analyzing the chemical interactions revealed unique binding properties for every compound, identified by a range of Pi-cation interactions and hydrogen bond types. We were able to learn more about the dynamic behaviors and stability of the protein-ligand complexes through a 200-nanosecond molecular dynamics simulation, as demonstrated by the compounds' consistent RMSD and RMSF values. The multidimensional nature of the data was clarified by the application of principal component analysis, which suggested stable conformations in the complexes with little modification. Further insight into the energy profiles and stability states of these complexes was also obtained by an examination of the free energy landscape. Our findings underscore the effectiveness of computational strategies in identifying and analyzing potential inhibitors for MBV VP35-RBD, offering promising paths for further experimental investigations and possible therapeutic development against the MBV.
Collapse
Affiliation(s)
- Abdullah S. Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | - Hadil Sultan Alawam
- College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammed Merae Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia;
| | - Maher S. Alwethaynani
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Shaqra University, Al-Quwayiyah 19257, Saudi Arabia
| | - Lina M. Alneghery
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia;
| | - Mubarak A. Alamri
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| |
Collapse
|
20
|
Zumbrun EE, Garvey CB, Wells JB, Lynn GC, Van Tongeren S, Steffens JT, Wetzel KS, Gomba LM, O’Brien KA, Rossi FD, Zeng X, Lee ED, Raymond JLW, Hoffman DA, Jay AN, Brown ES, Kallgren PA, Norris SL, Cantey-Kiser J, Kudiya H, Arthur C, Blair C, Babusis D, Chu VC, Singh B, Bannister R, Porter DP, Cihlar T, Dye JM. Characterization of the Cynomolgus Macaque Model of Marburg Virus Disease and Assessment of Timing for Therapeutic Treatment Testing. Viruses 2023; 15:2335. [PMID: 38140576 PMCID: PMC10748006 DOI: 10.3390/v15122335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2023] Open
Abstract
Marburg virus (MARV) causes severe disease and high mortality in humans. The objective of this study was to characterize disease manifestations and pathogenesis in cynomolgus macaques exposed to MARV. The results of this natural history study may be used to identify features of MARV disease useful in defining the ideal treatment initiation time for subsequent evaluations of investigational therapeutics using this model. Twelve cynomolgus macaques were exposed to a target dose of 1000 plaque-forming units MARV by the intramuscular route, and six control animals were mock-exposed. The primary endpoint of this study was survival to Day 28 post-inoculation (PI). Anesthesia events were minimized with the use of central venous catheters for periodic blood collection, and temperature and activity were continuously monitored by telemetry. All mock-exposed animals remained healthy for the duration of the study. All 12 MARV-exposed animals (100%) became infected, developed illness, and succumbed on Days 8-10 PI. On Day 4 PI, 11 of the 12 MARV-exposed animals had statistically significant temperature elevations over baseline. Clinically observable signs of MARV disease first appeared on Day 5 PI, when 6 of the 12 animals exhibited reduced responsiveness. Ultimately, systemic inflammation, coagulopathy, and direct cytopathic effects of MARV all contributed to multiorgan dysfunction, organ failure, and death or euthanasia of all MARV-exposed animals. Manifestations of MARV disease, including fever, systemic viremia, lymphocytolysis, coagulopathy, and hepatocellular damage, could be used as triggers for initiation of treatment in future therapeutic efficacy studies.
Collapse
Affiliation(s)
- Elizabeth E. Zumbrun
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
| | - Carly B. Garvey
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Jay B. Wells
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Ginger C. Lynn
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Sean Van Tongeren
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Jesse T. Steffens
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Kelly S. Wetzel
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Laura M. Gomba
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Kristan A. O’Brien
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Franco D. Rossi
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
| | - Eric D. Lee
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
| | - Jo Lynne W. Raymond
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
| | - Diana A. Hoffman
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
| | - Alexandra N. Jay
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
| | - Elizabeth S. Brown
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
- Geneva Foundation, Tacoma, WA 98402, USA
| | - Paul A. Kallgren
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
| | - Sarah L. Norris
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
| | | | - Humza Kudiya
- Gilead Sciences, Foster City, CA 94404, USA; (H.K.); (C.A.); (C.B.); (D.B.); (V.C.C.); (B.S.); (R.B.); (D.P.P.); (T.C.)
| | - Chris Arthur
- Gilead Sciences, Foster City, CA 94404, USA; (H.K.); (C.A.); (C.B.); (D.B.); (V.C.C.); (B.S.); (R.B.); (D.P.P.); (T.C.)
| | - Christiana Blair
- Gilead Sciences, Foster City, CA 94404, USA; (H.K.); (C.A.); (C.B.); (D.B.); (V.C.C.); (B.S.); (R.B.); (D.P.P.); (T.C.)
| | - Darius Babusis
- Gilead Sciences, Foster City, CA 94404, USA; (H.K.); (C.A.); (C.B.); (D.B.); (V.C.C.); (B.S.); (R.B.); (D.P.P.); (T.C.)
| | - Victor C. Chu
- Gilead Sciences, Foster City, CA 94404, USA; (H.K.); (C.A.); (C.B.); (D.B.); (V.C.C.); (B.S.); (R.B.); (D.P.P.); (T.C.)
| | - Bali Singh
- Gilead Sciences, Foster City, CA 94404, USA; (H.K.); (C.A.); (C.B.); (D.B.); (V.C.C.); (B.S.); (R.B.); (D.P.P.); (T.C.)
| | - Roy Bannister
- Gilead Sciences, Foster City, CA 94404, USA; (H.K.); (C.A.); (C.B.); (D.B.); (V.C.C.); (B.S.); (R.B.); (D.P.P.); (T.C.)
| | - Danielle P. Porter
- Gilead Sciences, Foster City, CA 94404, USA; (H.K.); (C.A.); (C.B.); (D.B.); (V.C.C.); (B.S.); (R.B.); (D.P.P.); (T.C.)
| | - Tomas Cihlar
- Gilead Sciences, Foster City, CA 94404, USA; (H.K.); (C.A.); (C.B.); (D.B.); (V.C.C.); (B.S.); (R.B.); (D.P.P.); (T.C.)
| | - John M. Dye
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD 21702, USA; (C.B.G.); (J.B.W.); (G.C.L.); (S.V.T.); (J.T.S.); (K.S.W.); (L.M.G.); (K.A.O.); (F.D.R.); (X.Z.); (E.D.L.); (J.L.W.R.); (D.A.H.); (A.N.J.); (E.S.B.); (P.A.K.); (S.L.N.); (J.M.D.)
| |
Collapse
|
21
|
Srivastava S, Sharma D, Kumar S, Sharma A, Rijal R, Asija A, Adhikari S, Rustagi S, Sah S, Al-qaim ZH, Bashyal P, Mohanty A, Barboza JJ, Rodriguez-Morales AJ, Sah R. Emergence of Marburg virus: a global perspective on fatal outbreaks and clinical challenges. Front Microbiol 2023; 14:1239079. [PMID: 37771708 PMCID: PMC10526840 DOI: 10.3389/fmicb.2023.1239079] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/25/2023] [Indexed: 09/30/2023] Open
Abstract
The Marburg virus (MV), identified in 1967, has caused deadly outbreaks worldwide, the mortality rate of Marburg virus disease (MVD) varies depending on the outbreak and virus strain, but the average case fatality rate is around 50%. However, case fatality rates have varied from 24 to 88% in past outbreaks depending on virus strain and case management. Designated a priority pathogen by the National Institute of Allergy and Infectious Diseases (NIAID), MV induces hemorrhagic fever, organ failure, and coagulation issues in both humans and non-human primates. This review presents an extensive exploration of MVD outbreak evolution, virus structure, and genome, as well as the sources and transmission routes of MV, including human-to-human spread and involvement of natural hosts such as the Egyptian fruit bat (Rousettus aegyptiacus) and other Chiroptera species. The disease progression involves early viral replication impacting immune cells like monocytes, macrophages, and dendritic cells, followed by damage to the spleen, liver, and secondary lymphoid organs. Subsequent spread occurs to hepatocytes, endothelial cells, fibroblasts, and epithelial cells. MV can evade host immune response by inhibiting interferon type I (IFN-1) synthesis. This comprehensive investigation aims to enhance understanding of pathophysiology, cellular tropism, and injury sites in the host, aiding insights into MVD causes. Clinical data and treatments are discussed, albeit current methods to halt MVD outbreaks remain elusive. By elucidating MV infection's history and mechanisms, this review seeks to advance MV disease treatment, drug development, and vaccine creation. The World Health Organization (WHO) considers MV a high-concern filovirus causing severe and fatal hemorrhagic fever, with a death rate ranging from 24 to 88%. The virus often spreads through contact with infected individuals, originating from animals. Visitors to bat habitats like caves or mines face higher risk. We tailored this search strategy for four databases: Scopus, Web of Science, Google Scholar, and PubMed. we primarily utilized search terms such as "Marburg virus," "Epidemiology," "Vaccine," "Outbreak," and "Transmission." To enhance comprehension of the virus and associated disease, this summary offers a comprehensive overview of MV outbreaks, pathophysiology, and management strategies. Continued research and learning hold promise for preventing and controlling future MVD outbreaks. GRAPHICAL ABSTRACT.
Collapse
Affiliation(s)
- Shriyansh Srivastava
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, India
| | - Deepika Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, India
| | - Sachin Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| | - Aditya Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, India
| | - Rishikesh Rijal
- Division of Infectious Diseases, University of Louisville, Louisville, KY, United States
| | - Ankush Asija
- WVU United Hospital Center, Bridgeport, WV, United States
| | | | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Sanjit Sah
- Global Consortium for Public Health and Research, Datta Meghe Institute of Higher Education and Research, Jawaharlal Nehru Medical College, Wardha, India
- Department of Anesthesia Techniques, SR Sanjeevani Hospital, Siraha, Nepal
| | | | - Prashant Bashyal
- Lumbini Medical College and Teaching Hospital, Kathmandu University Parvas, Palpa, Nepal
| | - Aroop Mohanty
- Department of Clinical Microbiology, All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| | | | - Alfonso J. Rodriguez-Morales
- Master Program on Clinical Epidemiology and Biostatistics, Universidad Científica del Sur, Lima, Peru
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Ranjit Sah
- Department of Microbiology, Tribhuvan University Teaching Spital, Institute of Medicine, Kathmandu, Nepal
- Department of Microbiology, Dr. D. Y. Patil Medical College, Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
- Department of Public Health Dentistry, Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra, India
| |
Collapse
|
22
|
von Creytz I, Gerresheim GK, Lier C, Schneider J, Schauflinger M, Benz M, Kämper L, Rohde C, Eickmann M, Biedenkopf N. Rescue and characterization of the first West African Marburg virus 2021 from Guinea. Heliyon 2023; 9:e19613. [PMID: 37810116 PMCID: PMC10558868 DOI: 10.1016/j.heliyon.2023.e19613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 10/10/2023] Open
Abstract
Marburg virus (MARV) is a causative agent of a severe hemorrhagic fever with high fatality rates endemic in central Africa. Current outbreaks of MARV in Equatorial Guinea and Tanzania underline the relevance of MARV as a public health emergency pathogen. In 2021, the first known human MARV case was confirmed in Guinea, West Africa. Since no infectious virus could be isolated from that fatal case in 2021, we generated recombinant (rec) MARV Guinea by reverse genetics in order to study and characterize this new MARV, which occurred in West Africa for the first time, in terms of its growth properties, detection by antibodies, and therapeutic potential compared to known MARV strains. Our results showed a solid viral replication of recMARV Guinea in human, bat, and monkey cell lines in comparison to other known MARV strains. We further demonstrated that replication of recMARV Guinea in cells can be inhibited by the nucleoside analogue remdesivir. Taken together, we could successfully reconstitute de novo the first West African MARV from Guinea showing similar replication kinetics in cells compared to other central African MARV strains. Our reverse genetics approach has proven successful in characterizing emerging viruses, especially when virus isolates are missing and viral genome sequences are incomplete.
Collapse
Affiliation(s)
- Isabel von Creytz
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany
| | | | - Clemens Lier
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany
| | - Jana Schneider
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany
| | | | - Marcel Benz
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany
| | - Lennart Kämper
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany
| | - Cornelius Rohde
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Markus Eickmann
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany
| | - Nadine Biedenkopf
- Institute of Virology, Philipps-University Marburg, 35043 Marburg, Germany
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| |
Collapse
|
23
|
Elsheikh R, Makram AM, Selim H, Nguyen D, Le TTT, Tran VP, Elaziz Khader SA, Huy NT. Reemergence of Marburgvirus disease: Update on current control and prevention measures and review of the literature. Rev Med Virol 2023; 33:e2461. [PMID: 37208958 DOI: 10.1002/rmv.2461] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 04/13/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023]
Abstract
In 1967, the very first case of the Marburgvirus disease (MVD) was detected in Germany and Serbia sequentially. Since then, MVD has been considered one of the most serious and deadly infectious diseases in the world with a case-fatality rate between 23% and 90% and a substantial number of recorded deaths. Marburgvirus belongs to the family of Filoviridae (filoviruses), which causes severe viral hemorrhagic fever (VHF). Some major risk factors for human infections are close contact with African fruit bats, MVD-infected non-human primates, and MVD-infected individuals. Currently, there is no vaccine or specific treatment for MVD, which emphasizes the seriousness of this disease. In July 2022, the World Health Organization reported outbreaks of MVD in Ghana after two suspected VHF cases were detected. This was followed in February and March 2023 with the emergence of the virus in two countries new to the virus: Equatorial Guinea and Tanzania, respectively. In this review, we aim to highlight the characteristics, etiology, epidemiology, and clinical symptoms of MVD, along with the current prevention measures and the possible treatments to control this virus.
Collapse
Affiliation(s)
- Randa Elsheikh
- Deanery of Biomedical Sciences, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- Online Research Club, Nagasaki, Japan
| | - Abdelrahman M Makram
- Online Research Club, Nagasaki, Japan
- School of Public Health, Imperial College London, London, UK
| | - Hager Selim
- Online Research Club, Nagasaki, Japan
- Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Dung Nguyen
- Online Research Club, Nagasaki, Japan
- Faculty of Health Sciences, University of the People, Pasadena, California, USA
| | - Thi Thu Thao Le
- Online Research Club, Nagasaki, Japan
- University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam
| | - Van-Phu Tran
- Online Research Club, Nagasaki, Japan
- Tra Vinh University, Tra Vinh City, Vietnam
| | - Sarah Abd Elaziz Khader
- Online Research Club, Nagasaki, Japan
- Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nguyen Tien Huy
- Online Research Club, Nagasaki, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
24
|
Sword J, Lee JH, Castro MA, Solomon J, Aiosa N, Reza SMS, Chu WT, Johnson JC, Bartos C, Cooper K, Jahrling PB, Johnson RF, Calcagno C, Crozier I, Kuhn JH, Hensley LE, Feuerstein IM, Mani V. Computed Tomography Imaging for Monitoring of Marburg Virus Disease: a Nonhuman Primate Proof-Of-Concept Study. Microbiol Spectr 2023; 11:e0349422. [PMID: 37036346 PMCID: PMC10269526 DOI: 10.1128/spectrum.03494-22] [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: 08/31/2022] [Accepted: 02/01/2023] [Indexed: 04/11/2023] Open
Abstract
Marburg virus (MARV) is a highly virulent zoonotic filovirid that causes Marburg virus disease (MVD) in humans. The pathogenesis of MVD remains poorly understood, partially due to the low number of cases that can be studied, the absence of state-of-the-art medical equipment in areas where cases are reported, and limitations on the number of animals that can be safely used in experimental studies under maximum containment animal biosafety level 4 conditions. Medical imaging modalities, such as whole-body computed tomography (CT), may help to describe disease progression in vivo, potentially replacing ethically contentious and logistically challenging serial euthanasia studies. Towards this vision, we performed a pilot study, during which we acquired whole-body CT images of 6 rhesus monkeys before and 7 to 9 days after intramuscular MARV exposure. We identified imaging abnormalities in the liver, spleen, and axillary lymph nodes that corresponded to clinical, virological, and gross pathological hallmarks of MVD in this animal model. Quantitative image analysis indicated hepatomegaly with a significant reduction in organ density (indicating fatty infiltration of the liver), splenomegaly, and edema that corresponded with gross pathological and histopathological findings. Our results indicated that CT imaging could be used to verify and quantify typical MVD pathogenesis versus altered, diminished, or absent disease severity or progression in the presence of candidate medical countermeasures, thus possibly reducing the number of animals needed and eliminating serial euthanasia. IMPORTANCE Marburg virus (MARV) is a highly virulent zoonotic filovirid that causes Marburg virus disease (MVD) in humans. Much is unknown about disease progression and, thus, prevention and treatment options are limited. Medical imaging modalities, such as whole-body computed tomography (CT), have the potential to improve understanding of MVD pathogenesis. Our study used CT to identify abnormalities in the liver, spleen, and axillary lymph nodes that corresponded to known clinical signs of MVD in this animal model. Our results indicated that CT imaging and analyses could be used to elucidate pathogenesis and possibly assess the efficacy of candidate treatments.
Collapse
Affiliation(s)
- Jennifer Sword
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Ji Hyun Lee
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Marcelo A. Castro
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Jeffrey Solomon
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Nina Aiosa
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Syed M. S. Reza
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Winston T. Chu
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Joshua C. Johnson
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Christopher Bartos
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Kurt Cooper
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Peter B. Jahrling
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Reed F. Johnson
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Claudia Calcagno
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Ian Crozier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Lisa E. Hensley
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Irwin M. Feuerstein
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| | - Venkatesh Mani
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, Fort Detrick, National Institutes of Health, Fort Detrick Frederick, Maryland, USA
| |
Collapse
|
25
|
Peart Akindele NA, Katamoni LD, Brockhurst J, Ghimire S, Suwanmanee S, Pieterse L, Metcalf Pate KA, Bunyan E, Bannister R, Cihlar T, Porter DP, Griffin DE. Effect of remdesivir post-exposure prophylaxis and treatment on pathogenesis of measles in rhesus macaques. Sci Rep 2023; 13:6463. [PMID: 37081035 PMCID: PMC10116456 DOI: 10.1038/s41598-023-33572-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/14/2023] [Indexed: 04/22/2023] Open
Abstract
Measles is a systemic disease initiated in the respiratory tract with widespread measles virus (MeV) infection of lymphoid tissue. Mortality can be substantial, but no licensed antiviral therapy is available. We evaluated both post-exposure prophylaxis and treatment with remdesivir, a broad-spectrum antiviral, using a well-characterized rhesus macaque model of measles. Animals were treated with intravenous remdesivir for 12 days beginning either 3 days after intratracheal infection (post-exposure prophylaxis, PEP) or 11 days after infection at the onset of disease (late treatment, LT). As PEP, remdesivir lowered levels of viral RNA in peripheral blood mononuclear cells, but RNA rebounded at the end of the treatment period and infectious virus was continuously recoverable. MeV RNA was cleared more rapidly from lymphoid tissue, was variably detected in the respiratory tract, and not detected in urine. PEP did not improve clinical disease nor lymphopenia and reduced the antibody response to infection. In contrast, LT had little effect on levels of viral RNA or the antibody response but also did not decrease clinical disease. Therefore, remdesivir transiently suppressed expression of viral RNA and limited dissemination when provided as PEP, but virus was not cleared and resumed replication without improvement in the clinical disease parameters evaluated.
Collapse
Affiliation(s)
- Nadine A Peart Akindele
- Division of Pediatric Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, 21218, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St, Rm E5636, Baltimore, MD, 21205, USA
- United States Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Laharika Dasharath Katamoni
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St, Rm E5636, Baltimore, MD, 21205, USA
- Zanvyl Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, 21205, USA
- BioCheck, Inc., South San Francisco, CA, 94080, USA
| | - Jacqueline Brockhurst
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St, Rm E5636, Baltimore, MD, 21205, USA
- Department of Molecular and Comparative Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21218, USA
| | - Shristi Ghimire
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St, Rm E5636, Baltimore, MD, 21205, USA
| | - San Suwanmanee
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St, Rm E5636, Baltimore, MD, 21205, USA
- Department of Epidemiology, Faculty of Public Health, Mahidol University, Bangkok, Thailand
| | - Lisa Pieterse
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St, Rm E5636, Baltimore, MD, 21205, USA
| | - Kelly A Metcalf Pate
- Department of Molecular and Comparative Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21218, USA
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | | | | | - Tomas Cihlar
- Gilead Sciences Inc., Foster City, CA, 94404, USA
| | | | - Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St, Rm E5636, Baltimore, MD, 21205, USA.
| |
Collapse
|
26
|
Liu K, Stern S, Heil EL, Li L, Khairi R, Heyward S, Wang H. Dexamethasone mitigates remdesivir-induced liver toxicity in human primary hepatocytes and COVID-19 patients. Hepatol Commun 2023; 7:e0034. [PMID: 36809346 PMCID: PMC9949788 DOI: 10.1097/hc9.0000000000000034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is a global pandemic that has caused more than 600 million cases and over six million deaths worldwide. Despite the availability of vaccination, COVID-19 cases continue to grow making pharmacological interventions essential. Remdesivir (RDV) is an FDA-approved antiviral drug for treatment of both hospitalized and non-hospitalized COVID-19 patients, albeit with potential for hepatotoxicity. This study characterizes the hepatotoxicity of RDV and its interaction with dexamethasone (DEX), a corticosteroid often co-administered with RDV for inpatient treatment of COVID-19. METHODS Human primary hepatocytes and HepG2 cells were used as in vitro models for toxicity and drug-drug interaction studies. Real-world data from hospitalized COVID-19 patients were analyzed for drug-induced elevation of serum ALT and AST. RESULTS In cultured hepatocytes, RDV markedly reduced the hepatocyte viability and albumin synthesis, while it increased the cleavage of caspase-8 and caspase-3, phosphorylation of histone H2AX, and release of ALT and AST in a concentration-dependent manner. Importantly, co-treatment with DEX partially reversed RDV-induced cytotoxic responses in human hepatocytes. Moreover, data from COVID-19 patients treated with RDV with and without DEX co-treatment suggested that among 1037 patients matched by propensity score, receiving the drug combination was less likely to result in elevation of serum AST and ALT levels (≥ 3 × ULN) compared to the RDV alone treated patients (OR = 0.44, 95% CI = 0.22-0.92, p = 0.03). CONCLUSION Our findings obtained from in vitro cell-based experiments and patient data analysis provide evidence suggesting combination of DEX and RDV holds the potential to reduce the likelihood of RDV-induced liver injury in hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Kaiyan Liu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Sydney Stern
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Emily L. Heil
- Department of Pharmacy Practice and Science, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Rula Khairi
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Scott Heyward
- BioIVT, 1450 S Rolling Rd, Halethorpe, Maryland, USA
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Expanded profiling of Remdesivir as a broad-spectrum antiviral and low potential for interaction with other medications in vitro. Sci Rep 2023; 13:3131. [PMID: 36823196 PMCID: PMC9950143 DOI: 10.1038/s41598-023-29517-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Remdesivir (GS-5734; VEKLURY) is a single diastereomer monophosphoramidate prodrug of an adenosine analog (GS-441524). Remdesivir is taken up by target cells and metabolized in multiple steps to form the active nucleoside triphosphate (GS-443902), which acts as a potent inhibitor of viral RNA-dependent RNA polymerases. Remdesivir and GS-441524 have antiviral activity against multiple RNA viruses. Here, we expand the evaluation of remdesivir's antiviral activity to members of the families Flaviviridae, Picornaviridae, Filoviridae, Orthomyxoviridae, and Hepadnaviridae. Using cell-based assays, we show that remdesivir can inhibit infection of flaviviruses (such as dengue 1-4, West Nile, yellow fever, Zika viruses), picornaviruses (such as enterovirus and rhinovirus), and filoviruses (such as various Ebola, Marburg, and Sudan virus isolates, including novel geographic isolates), but is ineffective or is significantly less effective against orthomyxoviruses (influenza A and B viruses), or hepadnaviruses B, D, and E. In addition, remdesivir shows no antagonistic effect when combined with favipiravir, another broadly acting antiviral nucleoside analog, and has minimal interaction with a panel of concomitant medications. Our data further support remdesivir as a broad-spectrum antiviral agent that has the potential to address multiple unmet medical needs, including those related to antiviral pandemic preparedness.
Collapse
|
28
|
Islam MA, Adeiza SS, Amin MR, Kaifa FH, Lorenzo JM, Bhattacharya P, Dhama K. A bibliometric study on Marburg virus research with prevention and control strategies. FRONTIERS IN TROPICAL DISEASES 2023. [DOI: 10.3389/fitd.2022.1068364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Marburg virus (MARV) is a pathogenic zoonotic RNA virus etiologic for Marburg virus disease (MVD), a severe hemorrhagic fever. This is a rare disease, with a high fatality rate, that spreads via infected blood or body fluids or indirectly via fomites (contaminated objects and substances such as clothed, beds, personal protective equipment, or medical equipments). A few vaccines to protect against MARV are undergoing clinical trials, but there is not yet an approved vaccine against this disease. Eventually, prevention and control guidelines should be adhered to rigorously to alleviate this infection. This bibliometric analysis aimed to harness narrative evaluation, emphasizing the significance of quantitative approaches and delineating the most thought-provoking concerns for researchers using VOSviewer software (Centre for Science and Technology Studies, Leiden University, the Netherlands). “Marburg Virus” OR “MARV” AND “Diseases” search criteria were used for the analysis of articles published between 1962 and 2022. Co-occurrence analysis was carried out, which characterized different thematic clusters. From this analysis, we found that 1688 published articles, and the number of publications increased across that period annually, with a growth rate of 8.78%. It is also conspicuous that the number of publications in the United States reached its acme during this period (i.e., 714 publications, accounting for 42.29% of the total), and the United States Army Medical Research Institute of Infectious Diseases published the most literature (i.e., 146 papers). Our study found that the three pre-eminent authors of Marburg virus papers were “FELDMANN, HEINZ“ of the National Institute of Allergy and Infectious Diseases, United States, “BECKER, STEPHAN” of the Philipps University of Marburg, Germany, and “GEISBERT, THOMAS W” of the University of Texas Medical Branch, United States. In this study we found that “JOURNAL OF VIROLOGY” has published the most pertinent literature, totaling 88 articles, followed by “The journal of Infectious Diseases”, which published 76 relevant papers, and “VIRUSES”, which published 52 corresponding papers. The most cited paper on the Marburg virus was published in Nature Medicine, with 522 total citations and 29 citations/year. Studies of the changing epidemiology and evolving nature of the virus and its ecological niche are required; breakthrough and implementation of the efficacious vaccine candidate(s), prophylaxis and therapeutic alternatives and supervision strategies, unveiling awareness-raising programs, and developing apposite and timely preparedness, prevention, and proactive control strategies are of utmost importance.
Collapse
|
29
|
Islam MR, Akash S, Rahman MM, Sharma R. Epidemiology, pathophysiology, transmission, genomic structure, treatment, and future perspectives of the novel Marburg virus outbreak. Int J Surg 2023; 109:36-38. [PMID: 36799786 PMCID: PMC10389455 DOI: 10.1097/js9.0000000000000096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/20/2022] [Indexed: 02/18/2023]
Affiliation(s)
- Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
30
|
Markin VA. Marburg virus and the disease it causes. JOURNAL OF MICROBIOLOGY, EPIDEMIOLOGY AND IMMUNOBIOLOGY 2022. [DOI: 10.36233/0372-9311-273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the 50 years since its discovery, many properties of the Marburg virus have been studied, but no reliable medical remedies of preventing and treating the infection it causes have been developed, although it can potentially cause large-scale epidemics.
Marburg fever is relevant due to the risk of importation to other countries. The source of infection in nature is bats (reservoir) and monkeys (intermediate host), and the routes of transmission are aerosol, contact and alimentary. The mortality rate in recent outbreaks has reached 90%. In convalescents the causative agent was identified in tears, semen, and liver biopsies weeks and months after recovery.
The lack of therapeutic and prophylactic antiviral drugs, high rates of mortality, infectivity, the ability of aerosol contamination, and a high epidemic potential all together define Marburg fever as a serious global threat to international health. The development of medical protection against this infection should be an urgent task of ensuring the biological safety of the population of the Russian Federation.
The most promising ways to develop vaccines against Marburg fever are the construction of recombinants based on adenovirus, vesicular stomatitis virus or alphavirus replicon, DNA vaccines. A reliable protective effect of the chemotherapy drug remdesivir in combination with human antibodies, as well as an etiotropic drug with an antisense mechanism of action and an interferon inducer has been shown. In model experiments with pseudovirus, fundamentally new ways of developing pathogen inhibitors were found preventing its exit from cells, as well as the construction of anti-gene-binding Fab fragments that inhibit the synthesis of viral RNA.
Collapse
|
31
|
Abir MH, Rahman T, Das A, Etu SN, Nafiz IH, Rakib A, Mitra S, Emran TB, Dhama K, Islam A, Siyadatpanah A, Mahmud S, Kim B, Hassan MM. Pathogenicity and virulence of Marburg virus. Virulence 2022; 13:609-633. [PMID: 35363588 PMCID: PMC8986239 DOI: 10.1080/21505594.2022.2054760] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/10/2022] [Accepted: 03/13/2022] [Indexed: 12/25/2022] Open
Abstract
Marburg virus (MARV) has been a major concern since 1967, with two major outbreaks occurring in 1998 and 2004. Infection from MARV results in severe hemorrhagic fever, causing organ dysfunction and death. Exposure to fruit bats in caves and mines, and human-to-human transmission had major roles in the amplification of MARV outbreaks in African countries. The high fatality rate of up to 90% demands the broad study of MARV diseases (MVD) that correspond with MARV infection. Since large outbreaks are rare for MARV, clinical investigations are often inadequate for providing the substantial data necessary to determine the treatment of MARV disease. Therefore, an overall review may contribute to minimizing the limitations associated with future medical research and improve the clinical management of MVD. In this review, we sought to analyze and amalgamate significant information regarding MARV disease epidemics, pathophysiology, and management approaches to provide a better understanding of this deadly virus and the associated infection.
Collapse
Affiliation(s)
- Mehedy Hasan Abir
- Faculty of Food Science and Technology, Chattogram Veterinary and Animal Sciences University, Chittagong, Bangladesh
| | - Tanjilur Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Ayan Das
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Silvia Naznin Etu
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Iqbal Hossain Nafiz
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Ahmed Rakib
- Department of Pharmacy, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Ariful Islam
- EcoHealth Alliance, New York, NY, USA
- Centre for Integrative Ecology, School of Life and Environmental Science, Deakin University, Victoria, Australia
| | - Abolghasem Siyadatpanah
- Ferdows School of Paramedical and Health, Birjand University of Medical Sciences, Birjand, Iran
| | - Shafi Mahmud
- Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Bonlgee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Korea
| | - Mohammad Mahmudul Hassan
- Queensland Alliance for One Health Sciences, School of Veterinary Sciences, The University of Queensland, Gatton, Australia
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
| |
Collapse
|
32
|
Inhibition of Viral RNA-Dependent RNA Polymerases by Nucleoside Inhibitors: An Illustration of the Unity and Diversity of Mechanisms. Int J Mol Sci 2022; 23:ijms232012649. [PMID: 36293509 PMCID: PMC9604226 DOI: 10.3390/ijms232012649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
RNA-dependent RNA polymerase (RdRP) is essential for the replication and expression of RNA viral genomes. This class of viruses comprise a large number of highly pathogenic agents that infect essentially all species of plants and animals including humans. Infections often lead to epidemics and pandemics that have remained largely out of control due to the lack of specific and reliable preventive and therapeutic regimens. This unmet medical need has led to the exploration of new antiviral targets, of which RdRP is a major one, due to the fact of its obligatory need in virus growth. Recent studies have demonstrated the ability of several synthetic nucleoside analogs to serve as mimics of the corresponding natural nucleosides. These mimics cause stalling/termination of RdRP, or misincorporation, preventing virus replication or promoting large-scale lethal mutations. Several such analogs have received clinical approval and are being routinely used in therapy. In parallel, the molecular structural basis of their inhibitory interactions with RdRP is being elucidated, revealing both traditional and novel mechanisms including a delayed chain termination effect. This review offers a molecular commentary on these mechanisms along with their clinical implications based on analyses of recent results, which should facilitate the rational design of structure-based antiviral drugs.
Collapse
|
33
|
Cross RW, Longini IM, Becker S, Bok K, Boucher D, Carroll MW, Díaz JV, Dowling WE, Draghia-Akli R, Duworko JT, Dye JM, Egan MA, Fast P, Finan A, Finch C, Fleming TR, Fusco J, Geisbert TW, Griffiths A, Günther S, Hensley LE, Honko A, Hunegnaw R, Jakubik J, Ledgerwood J, Luhn K, Matassov D, Meshulam J, Nelson EV, Parks CL, Rustomjee R, Safronetz D, Schwartz LM, Smith D, Smock P, Sow Y, Spiropoulou CF, Sullivan NJ, Warfield KL, Wolfe D, Woolsey C, Zahn R, Henao-Restrepo AM, Muñoz-Fontela C, Marzi A. An introduction to the Marburg virus vaccine consortium, MARVAC. PLoS Pathog 2022; 18:e1010805. [PMID: 36227853 PMCID: PMC9560149 DOI: 10.1371/journal.ppat.1010805] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The emergence of Marburg virus (MARV) in Guinea and Ghana triggered the assembly of the MARV vaccine "MARVAC" consortium representing leaders in the field of vaccine research and development aiming to facilitate a rapid response to this infectious disease threat. Here, we discuss current progress, challenges, and future directions for MARV vaccines.
Collapse
Affiliation(s)
- Robert W. Cross
- Galveston National Laboratory, and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Ira M. Longini
- Department of Biostatistics, University of Florida, Gainesville, Florida, United States of America
| | - Stephan Becker
- Institute for Virology, Philipps-Universität Marburg, Marburg, Germany
| | - Karin Bok
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - David Boucher
- U.S. COVID-19 Response at U.S. Department of Health and Human Services, Washington, DC, United States of America
| | - Miles W. Carroll
- Pandemic Sciences Institute, Nuffield Department of Medicine, Oxford University, United Kingdom
| | | | - William E. Dowling
- Coalition for Epidemic Preparedness Innovations (CEPI), Washington, Washington, DC, United States of America
| | - Ruxandra Draghia-Akli
- Johnson & Johnson—Global Public Health Research and Development, Spring House, Pennsylvania, United States of America
| | - James T. Duworko
- Partnership for Research on Infectious Diseases in Liberia, Monrovia, Liberia
| | - John M. Dye
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Michael A. Egan
- Auro Vaccines, Pearl River, New York, United States of America
| | | | - Amy Finan
- Sabin vaccine Institute, Washington, DC, United States of America
| | - Courtney Finch
- Sabin vaccine Institute, Washington, DC, United States of America
| | - Thomas R. Fleming
- University of Washington, Seattle, Washington, United States of America
| | - Joan Fusco
- Public Health Vaccines, Cambridge, Massachusetts, United States of America
| | - Thomas W. Geisbert
- Galveston National Laboratory, and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Anthony Griffiths
- National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Maryland, United States of America
| | - Stephan Günther
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Lisa E. Hensley
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, United States of America
| | - Anna Honko
- National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, Maryland, United States of America
| | - Ruth Hunegnaw
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jocelyn Jakubik
- Sabin vaccine Institute, Washington, DC, United States of America
| | - Julie Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kerstin Luhn
- Janssen Vaccines & Prevention, Leiden, the Netherlands
| | | | | | - Emily V. Nelson
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | - Roxana Rustomjee
- Sabin vaccine Institute, Washington, DC, United States of America
| | - David Safronetz
- Zoonotic Diseases and Special Pathogens Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | | | - Dean Smith
- Bacterial and Combination Vaccines, Public Health Agency of Canada, Ottawa, Ontario, Canada
| | - Paul Smock
- Sabin vaccine Institute, Washington, DC, United States of America
| | - Ydrissa Sow
- Collaborative Clinical Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Christina F. Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Nancy J. Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kelly L. Warfield
- Emergent BioSolutions, Gaithersburg, Maryland, United States of America
| | - Daniel Wolfe
- Bacterial and Combination Vaccines, Public Health Agency of Canada, Ottawa, Ontario, Canada
| | - Courtney Woolsey
- Galveston National Laboratory, and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Roland Zahn
- Janssen Vaccines & Prevention, Leiden, the Netherlands
| | | | | | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| |
Collapse
|
34
|
Ogunsakin RE, Ebenezer O, Jordaan MA, Shapi M, Ginindza TG. Mapping Scientific Productivity Trends and Hotspots in Remdesivir Research Publications: A Bibliometric Study from 2016 to 2021. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19148845. [PMID: 35886696 PMCID: PMC9318242 DOI: 10.3390/ijerph19148845] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 01/18/2023]
Abstract
In response to global efforts to control and exterminate infectious diseases, this study aims to provide insight into the productivity of remdesivir research and highlight future directions. To achieve this, there is a need to summarize and curate evidence from the literature. As a result, this study carried out comprehensive scientific research to detect trends in published articles related to remdesivir using a bibliometric analysis. Keywords associated with remdesivir were used to access pertinent published articles using the Scopus database. A total of 5321 research documents were retrieved, primarily as novel research articles (n = 2440; 46%). The number of publications increased exponentially from 2020 up to the present. The papers published by the top 12 institutions focusing on remdesivir accounted for 25.69% of the overall number of articles. The USA ranked as the most productive country, with 906 documents (37.1%), equivalent to one-third of the global publications in this field. The most productive institution was Icahn School of Medicine, Mount Sinai, in the USA (103 publications). The New England Journal of Medicine was the most cited, with an h-index of 13. The publication of research on remdesivir has gained momentum in the past year. The importance of remdesivir suggests that it needs continued research to help global health organizations detect areas requiring instant action to implement suitable measures. Furthermore, this study offers evolving hotspots and valuable insights into the scientific advances in this field and provides scaling-up analysis and evidence diffusion on remdesivir.
Collapse
Affiliation(s)
- Ropo E. Ogunsakin
- Discipline of Public Health Medicine, School of Nursing & Public Health, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa;
- Correspondence:
| | - Oluwakemi Ebenezer
- Department of Chemistry, Faculty of Natural Sciences, Mangosuthu University of Technology, Umlazi 4031, South Africa; (O.E.); (M.A.J.); (M.S.)
| | - Maryam A. Jordaan
- Department of Chemistry, Faculty of Natural Sciences, Mangosuthu University of Technology, Umlazi 4031, South Africa; (O.E.); (M.A.J.); (M.S.)
| | - Michael Shapi
- Department of Chemistry, Faculty of Natural Sciences, Mangosuthu University of Technology, Umlazi 4031, South Africa; (O.E.); (M.A.J.); (M.S.)
| | - Themba G. Ginindza
- Discipline of Public Health Medicine, School of Nursing & Public Health, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa;
- Cancer & Infectious Diseases Epidemiology Research Unit (CIDERU), College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa
| |
Collapse
|
35
|
Cross RW, Bornholdt ZA, Prasad AN, Woolsey C, Borisevich V, Agans KN, Deer DJ, Abelson DM, Kim DH, Shestowsky WS, Campbell LA, Bunyan E, Geisbert JB, Dobias NS, Fenton KA, Porter DP, Zeitlin L, Geisbert TW. Combination therapy with remdesivir and monoclonal antibodies protects nonhuman primates against advanced Sudan virus disease. JCI Insight 2022; 7:e159090. [PMID: 35413016 PMCID: PMC9220838 DOI: 10.1172/jci.insight.159090] [Citation(s) in RCA: 18] [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: 02/03/2022] [Accepted: 04/06/2022] [Indexed: 12/02/2022] Open
Abstract
A major challenge in managing acute viral infections is ameliorating disease when treatment is delayed. Previously, we reported the success of a 2-pronged mAb and antiviral remdesivir therapeutic approach to treat advanced illness in rhesus monkeys infected with Marburg virus (MARV). Here, we explored the benefit of a similar combination therapy for Sudan ebolavirus (Sudan virus; SUDV) infection. Importantly, no licensed anti-SUDV therapeutics currently exist, and infection of rhesus macaques with SUDV results in a rapid disease course similar to MARV with a mean time to death of 8.3 days. When initiation of therapy with either remdesivir or a pan-ebolavirus mAb cocktail (MBP431) was delayed until 6 days after inoculation, only 20% of macaques survived. In contrast, when remdesivir and MBP431 treatment were combined beginning 6 days after inoculation, significant protection (80%) was achieved. Our results suggest that combination therapy may be a viable treatment for patients with advanced filovirus disease that warrants further clinical testing in future outbreaks.
Collapse
Affiliation(s)
- Robert W. Cross
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - Abhishek N. Prasad
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Courtney Woolsey
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Krystle N. Agans
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Daniel J. Deer
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - Do H. Kim
- Mapp Biopharmaceutical, Inc., San Diego, California, USA
| | | | | | | | - Joan B. Geisbert
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Natalie S. Dobias
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Karla A. Fenton
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | | | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., San Diego, California, USA
| | - Thomas W. Geisbert
- Galveston National Laboratory and
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
36
|
Julander JG, Bunyan E, Jordan R, Porter DP. Remdesivir efficacy against yellow fever in a hamster model. Antiviral Res 2022; 203:105331. [DOI: 10.1016/j.antiviral.2022.105331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 01/01/2023]
|
37
|
García-Lledó A, Gómez-Pavón J, González Del Castillo J, Hernández-Sampelayo T, Martín-Delgado MC, Martín Sánchez FJ, Martínez-Sellés M, Molero García JM, Moreno Guillén S, Rodríguez-Artalejo FJ, Ruiz-Galiana J, Cantón R, De Lucas Ramos P, García-Botella A, Bouza E. Pharmacological treatment of COVID-19: an opinion paper. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2022; 35:115-130. [PMID: 34894208 PMCID: PMC8972693 DOI: 10.37201/req/158.2021] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The precocity and efficacy of the vaccines developed so far against COVID-19 has been the most significant and saving advance against the pandemic. The development of vaccines has not prevented, during the whole period of the pandemic, the constant search for therapeutic medicines, both among existing drugs with different indications and in the development of new drugs. The Scientific Committee of the COVID-19 of the Illustrious College of Physicians of Madrid wanted to offer an early, simplified and critical approach to these new drugs, to new developments in immunotherapy and to what has been learned from the immune response modulators already known and which have proven effective against the virus, in order to help understand the current situation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - E Bouza
- Servicio de Microbiología Clínica y Enfermedades Infecciosas del Hospital General Universitario Gregorio Marañón, Universidad Complutense. CIBERES. Ciber de Enfermedades Respiratorias. Madrid, Spain.
| |
Collapse
|
38
|
Hickman MR, Saunders DL, Bigger CA, Kane CD, Iversen PL. The development of broad-spectrum antiviral medical countermeasures to treat viral hemorrhagic fevers caused by natural or weaponized virus infections. PLoS Negl Trop Dis 2022; 16:e0010220. [PMID: 35259154 PMCID: PMC8903284 DOI: 10.1371/journal.pntd.0010220] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense (JPEO-CBRND) began development of a broad-spectrum antiviral countermeasure against deliberate use of high-consequence viral hemorrhagic fevers (VHFs) in 2016. The effort featured comprehensive preclinical research, including laboratory testing and rapid advancement of lead molecules into nonhuman primate (NHP) models of Ebola virus disease (EVD). Remdesivir (GS-5734, Veklury, Gilead Sciences) was the first small molecule therapeutic to successfully emerge from this effort. Remdesivir is an inhibitor of RNA-dependent RNA polymerase, a viral enzyme that is essential for viral replication. Its robust potency and broad-spectrum antiviral activity against certain RNA viruses including Ebola virus and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) led to its clinical evaluation in randomized, controlled trials (RCTs) in human patients during the 2018 EVD outbreak in the Democratic Republic of the Congo (DRC) and the ongoing Coronavirus Disease 2019 (COVID-19) pandemic today. Remdesivir was recently approved by the US Food and Drug Administration (FDA) for the treatment of COVID-19 requiring hospitalization. Substantial gaps remain in improving the outcomes of acute viral infections for patients afflicted with both EVD and COVID-19, including how to increase therapeutic breadth and strategies for the prevention and treatment of severe disease. Combination therapy that joins therapeutics with complimentary mechanisms of action appear promising, both preclinically and in RCTs. Importantly, significant programmatic challenges endure pertaining to a clear drug and biological product development pathway for therapeutics targeting biodefense and emerging pathogens when human efficacy studies are not ethical or feasible. For example, remdesivir's clinical development was facilitated by outbreaks of Ebola and SARS-CoV-2; as such, the development pathway employed for remdesivir is likely to be the exception rather than the rule. The current regulatory licensure pathway for therapeutics targeting rare, weaponizable VHF agents is likely to require use of FDA's established Animal Rule (21 CFR 314.600-650 for drugs; 21 CFR 601.90-95 for biologics). The FDA may grant marketing approval based on adequate and well-controlled animal efficacy studies when the results of those studies establish that the drug is safe and likely to produce clinical benefit in humans. In practical terms, this is anticipated to include a series of rigorous, well-documented, animal challenge studies, to include aerosol challenge, combined with human safety data. While small clinical studies against naturally occurring, high-consequence pathogens are typically performed where possible, approval for the therapeutics currently under development against biodefense pathogens will likely require the Animal Rule pathway utilizing studies in NHPs. We review the development of remdesivir as illustrative of the effort that will be needed to field future therapeutics against highly lethal, infectious agents.
Collapse
Affiliation(s)
- Mark R. Hickman
- Joint Project Manager for Chemical, Biological, Radiological, and Nuclear Medical (JPM CBRN Medical), Fort Detrick, Maryland, United States of America
| | - David L. Saunders
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - Catherine A. Bigger
- Logistics Management International Inc, Tysons Corner, Virginia, United States of America
| | | | | |
Collapse
|
39
|
Cihlar T, Mackman RL. Journey of remdesivir from the inhibition of hepatitis C virus to the treatment of COVID-19. Antivir Ther 2022; 27:13596535221082773. [DOI: 10.1177/13596535221082773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
If a planned path reaches a dead-end, one can simply stop. Or one can turn around, walk back to the last intersection and take another path, or one can consider taking few paths in parallel. The last scenario is reflective of the journey of remdesivir, the first antiviral for the treatment of COVID-19, that was approved by FDA less than 10 months after the isolation of SARS-CoV-2, the virus responsible for the COVID-19 pandemic. As of January 2022, 10 million COVID-19 patients have been treated with remdesivir worldwide, but the journey of this molecule started more than a decade earlier with the search for a cure of hepatitis C virus. The development path of remdesivir before the emergence of COVID-19 represents a valuable example of a preemptive pandemic preparedness, but the pursuit of this path would not have been possible without sustaining support of John C. Martin, whom we will sorely miss for his piercing vision, uncompromising leadership, and genuine compassion for patients suffering around the world.
Collapse
|
40
|
Affiliation(s)
- Fang Zhao
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and the Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yun He
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and the Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Hongzhou Lu
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and the Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
41
|
Bradfute SB. The discovery and development of novel treatment strategies for filoviruses. Expert Opin Drug Discov 2021; 17:139-149. [PMID: 34962451 DOI: 10.1080/17460441.2022.2013800] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Filoviruses are negative-stranded, enveloped RNA viruses that can cause hemorrhagic fever in humans and include Ebola and Marburg viruses. Lethality rates can reach 90% in isolated outbreaks. The 2013-2016 Ebola virus epidemic demonstrated the global threat of filoviruses and hastened development of vaccines and therapeutics. There are six known filoviruses that cause disease in humans, but still few therapeutics are available for treatment. AREAS COVERED This review summarizes identification, testing, and development of therapeutics based on the peer-reviewed scientific literature beginning with the discovery of filoviruses in 1967. Small molecules, antibodies, cytokines, antisense, post-exposure vaccination, and host-targeted therapeutic approaches are discussed. An emphasis is placed on therapeutics that have shown promise in in vivo studies. EXPERT OPINION Two monoclonal antibody regimens are approved for use in humans for one filovirus (Ebola virus), and preclinical nonhuman primate studies suggest that other monoclonal-based therapies are likely to be effective against other filoviruses. Significant progress has been made in small-molecule antivirals and host-targeted approaches. An important consideration is the necessity of pan-filovirus therapeutics via broadly effective small molecules, antibody cocktails, and cross-reactive antibodies. The use of filovirus therapeutics as prophylactic treatment or in chronically infected individuals should be considered.
Collapse
Affiliation(s)
- Steven B Bradfute
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, USA
| |
Collapse
|
42
|
Yan D, Ra OH, Yan B. The nucleoside antiviral prodrug remdesivir in treating COVID-19 and beyond with interspecies significance. ANIMAL DISEASES 2021; 1:15. [PMID: 34778881 PMCID: PMC8422062 DOI: 10.1186/s44149-021-00017-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/25/2021] [Indexed: 01/18/2023] Open
Abstract
Infectious pandemics result in hundreds and millions of deaths, notable examples of the Spanish Flu, the Black Death and smallpox. The current pandemic, caused by SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), is unprecedented even in the historical term of pandemics. The unprecedentedness is featured by multiple surges, rapid identification of therapeutic options and accelerated development of vaccines. Remdesivir, originally developed for Ebola viral disease, is the first treatment of COVID-19 (Coronavirus disease 2019) approved by the United States Food and Drug Administration. As demonstrated by in vitro and preclinical studies, this therapeutic agent is highly potent with a broad spectrum activity against viruses from as many as seven families even cross species. However, randomized controlled trials have failed to confirm the efficacy and safety. Remdesivir improves some clinical signs but not critical parameters such as mortality. This antiviral agent is an ester/phosphorylation prodrug and excessive hydrolysis which increases cellular toxicity. Remdesivir is given intravenously, leading to concentration spikes and likely increasing the potential of hydrolysis-based toxicity. This review has proposed a conceptual framework for improving its efficacy and minimizing toxicity not only for the COVID-19 pandemic but also for future ones caused by remdesivir-sensitive viruses.
Collapse
Affiliation(s)
- Daisy Yan
- Sidney Kimmel Medical College, Thomas Jefferson University, 1025 Walnut St, Philadelphia, PA 19107 USA
| | - One Hyuk Ra
- Department of Anesthesiology, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115 USA
| | - Bingfang Yan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229 USA
| |
Collapse
|
43
|
Abstract
Remdesivir (GS-5734, Veklury®) has remained the only antiviral drug formally approved by the US FDA for the treatment of Covid-19 (SARS-CoV-2 infection). Its key structural features are the fact that it is a C-nucleoside (adenosine) analogue, contains a 1'-cyano function, and could be considered as a ProTide based on the presence of a phosphoramidate group. Its antiviral spectrum and activity in animal models have been well established and so has been its molecular mode of action as a delayed chain terminator of the viral RdRp (RNA-dependent RNA polymerase). Its clinical efficacy has been evaluated, but needs to be optimized with regard to timing, dosage and duration of treatment, and route of administration. Safety, toxicity and pharmacokinetics need to be further addressed, and so are its potential combinations with other drugs such as corticosteroids (i.e. dexamethasone) and ribavirin.
Collapse
|
44
|
Remdesivir is efficacious in rhesus monkeys exposed to aerosolized Ebola virus. Sci Rep 2021; 11:19458. [PMID: 34593911 PMCID: PMC8484580 DOI: 10.1038/s41598-021-98971-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/02/2021] [Indexed: 11/30/2022] Open
Abstract
Efficacious therapeutics for Ebola virus disease are in great demand. Ebola virus infections mediated by mucosal exposure, and aerosolization in particular, present a novel challenge due to nontypical massive early infection of respiratory lymphoid tissues. We performed a randomized and blinded study to compare outcomes from vehicle-treated and remdesivir-treated rhesus monkeys in a lethal model of infection resulting from aerosolized Ebola virus exposure. Remdesivir treatment initiated 4 days after exposure was associated with a significant survival benefit, significant reduction in serum viral titer, and improvements in clinical pathology biomarker levels and lung histology compared to vehicle treatment. These observations indicate that remdesivir may have value in countering aerosol-induced Ebola virus disease.
Collapse
|
45
|
Remdesivir inhibits the polymerases of the novel filoviruses Lloviu and Bombali virus. Antiviral Res 2021; 192:105120. [PMID: 34126139 DOI: 10.1016/j.antiviral.2021.105120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/21/2021] [Accepted: 06/10/2021] [Indexed: 01/18/2023]
Abstract
In recent years, a number of novel filoviruses (e.g. Lloviu virus (LLOV) and Bombali virus (BOMV)) have been discovered. While antibody-based therapeutics have recently been approved for treatment of infections with the filovirus Ebola virus (EBOV), no treatment options for novel filoviruses currently exist. Further, the development of antivirals against them is complicated by the fact that only sequence information, but no actual virus isolates, are available. To address this issue, we developed a reverse genetics-based minigenome system for BOMV, which allows us to assess the activity of the BOMV polymerase. Together with similar systems that we have developed for other filoviruses in the past (i.e. LLOV and Reston virus (RESTV)), we then assessed the efficiency of remdesivir, a known inhibitor of the EBOV polymerase that has recently been tested in a clinical trial for efficacy against Ebola disease. We show that remdesivir is indeed also active against the polymerases of BOMV, LLOV, and RESTV, with comparable IC50 values to its activity against EBOV. This suggests that treatment with remdesivir might represent a viable option in case of infections with novel filoviruses.
Collapse
|
46
|
Vicenti I, Zazzi M, Saladini F. SARS-CoV-2 RNA-dependent RNA polymerase as a therapeutic target for COVID-19. Expert Opin Ther Pat 2021; 31:325-337. [PMID: 33475441 PMCID: PMC7938656 DOI: 10.1080/13543776.2021.1880568] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/20/2021] [Indexed: 02/08/2023]
Abstract
Introduction: The current SARS-CoV-2 pandemic urgently demands for both prevention and treatment strategies. RNA-dependent RNA-polymerase (RdRp), which has no counterpart in human cells, is an excellent target for drug development. Given the time-consuming process of drug development, repurposing drugs approved for other indications or at least successfully tested in terms of safety and tolerability, is an attractive strategy to rapidly provide an effective medication for severe COVID-19 cases.Areas covered: The currently available data and upcominSg studies on RdRp which can be repurposed to halt SARS-CoV-2 replication, are reviewed.Expert opinion: Drug repurposing and design of novel compounds are proceeding in parallel to provide a quick response and new specific drugs, respectively. Notably, the proofreading SARS-CoV-2 exonuclease activity could limit the potential for drugs designed as immediate chain terminators and favor the development of compounds acting through delayed termination. While vaccination is awaited to curb the SARS-CoV-2 epidemic, even partially effective drugs from repurposing strategies can be of help to treat severe cases of disease. Considering the high conservation of RdRp among coronaviruses, an improved knowledge of its activity in vitro can provide useful information for drug development or drug repurposing to combat SARS-CoV-2 as well as future pandemics.
Collapse
Affiliation(s)
- Ilaria Vicenti
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Maurizio Zazzi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesco Saladini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
47
|
Cross RW, Bornholdt ZA, Prasad AN, Borisevich V, Agans KN, Deer DJ, Abelson DM, Kim DH, Shestowsky WS, Campbell LA, Bunyan E, Geisbert JB, Fenton KA, Zeitlin L, Porter DP, Geisbert TW. Combination therapy protects macaques against advanced Marburg virus disease. Nat Commun 2021; 12:1891. [PMID: 33767178 PMCID: PMC7994808 DOI: 10.1038/s41467-021-22132-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/03/2021] [Indexed: 11/08/2022] Open
Abstract
Monoclonal antibodies (mAbs) and remdesivir, a small-molecule antiviral, are promising monotherapies for many viruses, including members of the genera Marburgvirus and Ebolavirus (family Filoviridae), and more recently, SARS-CoV-2. One of the major challenges of acute viral infections is the treatment of advanced disease. Thus, extending the window of therapeutic intervention is critical. Here, we explore the benefit of combination therapy with a mAb and remdesivir in a non-human primate model of Marburg virus (MARV) disease. While rhesus monkeys are protected against lethal infection when treatment with either a human mAb (MR186-YTE; 100%), or remdesivir (80%), is initiated 5 days post-inoculation (dpi) with MARV, no animals survive when either treatment is initiated alone beginning 6 dpi. However, by combining MR186-YTE with remdesivir beginning 6 dpi, significant protection (80%) is achieved, thereby extending the therapeutic window. These results suggest value in exploring combination therapy in patients presenting with advanced filovirus disease.
Collapse
Affiliation(s)
- Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | | | - Abhishek N Prasad
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Krystle N Agans
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Daniel J Deer
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Dafna M Abelson
- Mapp Biopharmaceutical, Inc., 6160 Lusk Blvd Ste C200, San Diego, CA, USA
| | - Do H Kim
- Mapp Biopharmaceutical, Inc., 6160 Lusk Blvd Ste C200, San Diego, CA, USA
| | | | | | - Elaine Bunyan
- Gilead Sciences, Inc., 333 Lakeside Dr, Foster City, CA, USA
| | - Joan B Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Karla A Fenton
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., 6160 Lusk Blvd Ste C200, San Diego, CA, USA.
| | | | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX, USA.
| |
Collapse
|
48
|
Off-Target In Vitro Profiling Demonstrates that Remdesivir Is a Highly Selective Antiviral Agent. Antimicrob Agents Chemother 2021; 65:AAC.02237-20. [PMID: 33229429 PMCID: PMC7849018 DOI: 10.1128/aac.02237-20] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022] Open
Abstract
Remdesivir (RDV, GS-5734), the first FDA-approved antiviral for the treatment of COVID-19, is a single diastereomer monophosphoramidate prodrug of an adenosine analogue. It is intracellularly metabolized into the active triphosphate form, which in turn acts as a potent and selective inhibitor of multiple viral RNA polymerases. Remdesivir (RDV, GS-5734), the first FDA-approved antiviral for the treatment of COVID-19, is a single diastereomer monophosphoramidate prodrug of an adenosine analogue. It is intracellularly metabolized into the active triphosphate form, which in turn acts as a potent and selective inhibitor of multiple viral RNA polymerases. RDV has broad-spectrum activity against members of the coronavirus family, such as SARS-CoV-2, SARS-CoV, and MERS-CoV, as well as filoviruses and paramyxoviruses. To assess the potential for off-target toxicity, RDV was evaluated in a set of cellular and biochemical assays. Cytotoxicity was evaluated in a set of relevant human cell lines and primary cells. In addition, RDV was evaluated for mitochondrial toxicity under aerobic and anaerobic metabolic conditions, and for the effects on mitochondrial DNA content, mitochondrial protein synthesis, cellular respiration, and induction of reactive oxygen species. Last, the active 5′-triphosphate metabolite of RDV, GS-443902, was evaluated for potential interaction with human DNA and RNA polymerases. Among all of the human cells tested under 5 to 14 days of continuous exposure, the 50% cytotoxic concentration (CC50) values of RDV ranged from 1.7 to >20 μM, resulting in selectivity indices (SI, CC50/EC50) from >170 to 20,000, with respect to RDV anti-SARS-CoV-2 activity (50% effective concentration [EC50] of 9.9 nM in human airway epithelial cells). Overall, the cellular and biochemical assays demonstrated a low potential for RDV to elicit off-target toxicity, including mitochondria-specific toxicity, consistent with the reported clinical safety profile.
Collapse
|
49
|
Kortepeter MG, Dierberg K, Shenoy ES, Cieslak TJ. Marburg virus disease: A summary for clinicians. Int J Infect Dis 2020; 99:233-242. [PMID: 32758690 PMCID: PMC7397931 DOI: 10.1016/j.ijid.2020.07.042] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/09/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES This article summarizes the countermeasures for Marburg virus disease, focusing on pathogenesis, clinical features and diagnostics. There is an emphasis on therapies and vaccines that have demonstrated, through their evaluation in nonhuman primates (NHPs) and/or in humans, potential for use in an emergency situation. METHODS A standardized literature review was conducted on vaccines and treatments for Marburg virus disease, with a focus on human and nonhuman primate data published in the last five years. More detail on the methods that were used is summarized in a companion methods paper. RESULTS The study identified six treatments and four vaccine platforms that have demonstrated, through their efficacy in NHPs, potential benefit for treating or preventing infection in humans. CONCLUSION Succinct summaries of Marburg countermeasures are provided to give the busy clinician a head start in reviewing the literature if faced with a patient with Marburg virus disease. Links to other authoritative sources of information are also provided.
Collapse
|
50
|
Abstract
We highlighted the discovery process, preparation techniques, broad-spectrum activities, antiviral mechanism, and future perspectives of GS-5734.
Collapse
Affiliation(s)
- Zhonglei Wang
- School of Chemistry and Chemical Engineering
- Qufu Normal University
- Qufu 273165
- China
- School of Pharmaceutical Sciences
| | - Liyan Yang
- School of Physics and Engineering
- Qufu Normal University
- Qufu 273165
- China
| |
Collapse
|