1
|
Saha O, Siddiquee NH, Akter R, Sarker N, Bristi UP, Sultana KF, Remon SMLR, Sultana A, Shishir TA, Rahaman MM, Ahmed F, Hossen F, Amin MR, Akter MS. Antiviral Activity, Pharmacoinformatics, Molecular Docking, and Dynamics Studies of Azadirachta indica Against Nipah Virus by Targeting Envelope Glycoprotein: Emerging Strategies for Developing Antiviral Treatment. Bioinform Biol Insights 2024; 18:11779322241264145. [PMID: 39072258 PMCID: PMC11283663 DOI: 10.1177/11779322241264145] [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: 11/28/2023] [Accepted: 06/06/2024] [Indexed: 07/30/2024] Open
Abstract
The Nipah virus (NiV) belongs to the Henipavirus genus is a serious public health concern causing numerous outbreaks with higher fatality rate. Unfortunately, there is no effective medication available for NiV. To investigate possible inhibitors of NiV infection, we used in silico techniques to discover treatment candidates in this work. As there are not any approved treatments for NiV infection, the NiV-enveloped attachment glycoprotein was set as target for our study, which is responsible for binding to and entering host cells. Our in silico drug design approach included molecular docking, post-docking molecular mechanism generalised born surface area (MM-GBSA), absorption, distribution, metabolism, excretion/toxicity (ADME/T), and molecular dynamics (MD) simulations. We retrieved 418 phytochemicals associated with the neem plant (Azadirachta indica) from the IMPPAT database, and molecular docking was used to ascertain the compounds' binding strength. The top 3 phytochemicals with binding affinities of -7.118, -7.074, and -6.894 kcal/mol for CIDs 5280343, 9064, and 5280863, respectively, were selected for additional study based on molecular docking. The post-docking MM-GBSA of those 3 compounds was -47.56, -47.3, and -43.15 kcal/mol, respectively. As evidence of their efficacy and safety, all the chosen drugs had favorable toxicological and pharmacokinetic (Pk) qualities. We also performed MD simulations to confirm the stability of the ligand-protein complex structures and determine whether the selected compounds are stable at the protein binding site. All 3 phytochemicals, Quercetin (CID: 5280343), Cianidanol (CID: 9064), and Kaempferol (CID: 5280863), appeared to have outstanding binding stability to the target protein than control ribavirin, according to the molecular docking, MM-GBSA, and MD simulation outcomes. Overall, this work offers a viable approach to developing novel medications for treating NiV infection.
Collapse
Affiliation(s)
- Otun Saha
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Noimul Hasan Siddiquee
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Rahima Akter
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Nikkon Sarker
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Uditi Paul Bristi
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | | | - SM Lutfor Rahman Remon
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Afroza Sultana
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Tushar Ahmed Shishir
- Department of Mathematics and Natural Sciences, BRAC University, Dhaka, Bangladesh
| | | | - Firoz Ahmed
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Foysal Hossen
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Mohammad Ruhul Amin
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Mir Salma Akter
- Department of Microbiology, Noakhali Science and Technology University, Noakhali, Bangladesh
- Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
2
|
Olasoju T, Olasoju M, Dagash B, Abaye B, Enumah C, Isah S, Bolori M, Adebowale O. LASSA FEVER IN INTERNALLY-DISPLACED PERSONS' CAMP: A CASE REPORT AT ZABARMARI, BORNO STATE, NIGERIA. Ann Ib Postgrad Med 2024; 22:94-99. [PMID: 38939877 PMCID: PMC11205715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/01/2024] [Indexed: 06/29/2024] Open
Abstract
Introdution Lassa fever is a viral hemorrhagic disease caused by the Lassa virus, a single stranded RNA virus of the Arenavirus family. It is a zoonotic illness spread by rats of the speciesMastomys natalensis . Between weeks 1 and 17, (2017), 242 suspected Lassa fever cases were reported in Nigeria, with 58 laboratory confirmed cases and 46 fatalities (CFR, 19.01%) from 50 Local Government Areas (LGAs) in 20 States. Methods We conducted an outbreak investigation and gathered a thorough clinical history of the index case as well as contacts, who were then followed up using the standard viral hemorrhagic fever contact monitoring form. Following that, blood samples were collected from this patient. A total of 54 contacts were tracked for 21 days and their temperatures were recorded using a clinical thermometer. Furthermore, an environmental evaluation of the Zabarmari community and the Madinatu Internally-displaced persons' (IDP) camp was carried out. Results The index case was a 32-year-old woman who was internallydisplaced in Zabarmari community. Her symptoms began with fever and vaginal bleeding and progressed to bleeding from the nose, mouth, and urethra. There was a history of rat exposure as well as inadequate environmental sanitation and hygiene. Real Time PCR detected Lassa fever in the blood sample. The Borno State Ministry of Environment, in partnership with the Ministry of Health, undertook public health education on Lassa fever prevention and implemented excellent sanitary measures. Conclusion Increased awareness creation on good infection prevention and control practices is crucial among internally-displaced person and health care providers to prevent occurrence and spread of the disease.
Collapse
Affiliation(s)
- T.I Olasoju
- Nigeria Field Epidemiology and Laboratory Training Program (NFELTP)
- Department of Veterinary and Pest Control Services, Federal Ministry of Agriculture and Rural Development, Garki, Abuja, FCT, Nigeria
| | - M.I. Olasoju
- Department of Veterinary Public Health and Preventive Medicine, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - B. Dagash
- Nigeria Field Epidemiology and Laboratory Training Program (NFELTP)
- Department of Medical Microbiology, University of Maiduguri Teaching Hospital, Maiduguri, Borno State, Nigeria
| | - B.B Abaye
- Nigeria Field Epidemiology and Laboratory Training Program (NFELTP)
| | - C. Enumah
- Nigeria Field Epidemiology and Laboratory Training Program (NFELTP)
| | - S. Isah
- Nigeria Field Epidemiology and Laboratory Training Program (NFELTP)
- Bauchi State Primary Health Care Development Agency, Bauchi, Bauchi State, Nigeria
| | - M.T Bolori
- Department of Community Medicine, University of Maiduguri, Maiduguri, Borno State, Nigeria
| | - O.O. Adebowale
- Department of Veterinary Public Health and Preventive Medicine, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| |
Collapse
|
3
|
Grant DS, Samuels RJ, Garry RF, Schieffelin JS. Lassa Fever Natural History and Clinical Management. Curr Top Microbiol Immunol 2023. [PMID: 37106159 DOI: 10.1007/82_2023_263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Lassa fever is caused by Lassa virus (LASV), an Old World Mammarenavirus that is carried by Mastomys natalensis and other rodents. It is endemic in Sierra Leone, Nigeria, and other countries in West Africa. The clinical presentation of LASV infection is heterogenous varying from an inapparent or mild illness to a fatal hemorrhagic fever. Exposure to LASV is usually through contact with rodent excreta. After an incubation period of 1-3 weeks, initial symptoms such as fever, headache, and fatigue develop that may progress to sore throat, retrosternal chest pain, conjunctival injection, vomiting, diarrhea, and abdominal pain. Severe illness, including hypotension, shock, and multiorgan failure, develops in a minority of patients. Patient demographics and case fatality rates are distinctly different in Sierra Leone and Nigeria. Laboratory diagnosis relies on the detection of LASV antigens or genomic RNA. LASV-specific immunoglobulin G and M assays can also contribute to clinical management. The mainstay of treatment for Lassa fever is supportive care. The nucleoside analog ribavirin is commonly used to treat acute Lassa fever but is considered useful only if treatment is begun early in the disease course. Drugs in development, including a monoclonal antibody cocktail, have the potential to impact the management of Lassa fever.
Collapse
Affiliation(s)
- Donald S Grant
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health, Kenema, Sierra Leone
- College of Medicine and Allied Health Sciences (COMAHS), University of Sierra Leone, Freetown, Sierra Leone
| | - Robert J Samuels
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health, Kenema, Sierra Leone
| | - Robert F Garry
- School of Medicine, Department of Microbiology and Immunology, Tulane University, New Orleans, LA, 70112, USA
- Zalgen Labs, Frederick, MD, 21703, USA
- Global Virus Network (GVN), Baltimore, MD, 21201, USA
| | - John S Schieffelin
- School of Medicine, Department of Pediatrics, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
4
|
Madueme PGU, Chirove F. Understanding the transmission pathways of Lassa fever: A mathematical modeling approach. Infect Dis Model 2023; 8:27-57. [PMID: 36582748 PMCID: PMC9762202 DOI: 10.1016/j.idm.2022.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
The spread of Lassa fever infection is increasing in West Africa over the last decade. The impact of this can better be understood when considering the various possible transmission routes. We designed a mathematical model for the epidemiology of Lassa Fever using a system of nonlinear ordinary differential equations to determine the effect of transmission pathways toward the infection progression in humans and rodents including those usually neglected such as the environmental surface and aerosol routes. We analyzed the model and carried out numerical simulations to determine the impact of each transmission routes. Our results showed that the burden of Lassa fever infection is increased when all the transmission routes are incorporated and most single transmission routes are less harmful, but when in combination with other transmission routes, they increase the Lassa fever burden. It is therefore important to consider multiple transmission routes to better estimate the Lassa fever burden optimally and in turn determine control strategies targeted at the transmission pathways.
Collapse
Affiliation(s)
- Praise-God Uchechukwu Madueme
- Department of Mathematics and Applied Mathematics, University of Johannesburg, Cnr Kingsway and University Road, Auckland Park, 2092, Johannesburg, South Africa
| | - Faraimunashe Chirove
- Department of Mathematics and Applied Mathematics, University of Johannesburg, Cnr Kingsway and University Road, Auckland Park, 2092, Johannesburg, South Africa
| |
Collapse
|
5
|
Kale A, Shelke V, Dagar N, Anders HJ, Gaikwad AB. How to use COVID-19 antiviral drugs in patients with chronic kidney disease. Front Pharmacol 2023; 14:1053814. [PMID: 36843922 PMCID: PMC9947246 DOI: 10.3389/fphar.2023.1053814] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
Antiviral drugs such as Remdesivir (Veklury), Nirmatrelvir with Ritonavir (Paxlovid), Azvudine, and Molnupiravir (Lagevrio) can reduce the risk for severe and fatal Coronavirus Disease (COVID)-19. Although chronic kidney disease is a highly prevalent risk factor for severe and fatal COVID-19, most clinical trials with these drugs excluded patients with impaired kidney function. Advanced CKD is associated with a state of secondary immunodeficiency (SIDKD), which increases the susceptibility to severe COVID-19, COVID-19 complications, and the risk of hospitalization and mortality among COVID-19 patients. The risk to develop COVID-19 related acute kidney injury is higher in patients with precedent CKD. Selecting appropriate therapies for COVID-19 patients with impaired kidney function is a challenge for healthcare professionals. Here, we discuss the pharmacokinetics and pharmacodynamics of COVID-19-related antiviral drugs with a focus on their potential use and dosing in COVID-19 patients with different stages of CKD. Additionally, we describe the adverse effects and precautions to be taken into account when using these antivirals in COVID-19 patients with CKD. Lastly, we also discuss about the use of monoclonal antibodies in COVID-19 patients with kidney disease and related complications.
Collapse
Affiliation(s)
- Ajinath Kale
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Vishwadeep Shelke
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Neha Dagar
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Internal Medicine IV, Hospital of the Ludwig Maximilians University Munich, Munich, Germany
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| |
Collapse
|
6
|
Grant DS, Engel EJ, Roberts Yerkes N, Kanneh L, Koninga J, Gbakie MA, Alhasan F, Kanneh FB, Kanneh IM, Kamara FK, Momoh M, Yillah MS, Foday M, Okoli A, Zeoli A, Weldon C, Bishop CM, Zheng C, Hartnett J, Chao K, Shore K, Melnik LI, Mucci M, Bond NG, Doyle P, Yenni R, Podgorski R, Ficenec SC, Moses L, Shaffer JG, Garry RF, Schieffelin JS. Seroprevalence of anti-Lassa Virus IgG antibodies in three districts of Sierra Leone: A cross-sectional, population-based study. PLoS Negl Trop Dis 2023; 17:e0010938. [PMID: 36758101 PMCID: PMC9946222 DOI: 10.1371/journal.pntd.0010938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 02/22/2023] [Accepted: 11/09/2022] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Lassa virus (LASV), the cause of the acute viral hemorrhagic illness Lassa fever (LF), is endemic in West Africa. Infections in humans occur mainly after exposure to infected excrement or urine of the rodent-host, Mastomys natalensis. The prevalence of exposure to LASV in Sierra Leone is crudely estimated and largely unknown. This cross-sectional study aimed to establish a baseline point seroprevalence of IgG antibodies to LASV in three administrative districts of Sierra Leone and identify potential risk factors for seropositivity and LASV exposure. METHODOLOGY AND PRINCIPAL FINDINGS Between 2015 and 2018, over 10,642 participants from Kenema, Tonkolili, and Port Loko Districts were enrolled in this cross-sectional study. Previous LASV and LF epidemiological studies support classification of these districts as "endemic," "emerging," and "non-endemic", respectively. Dried blood spot samples were tested for LASV antibodies by ELISA to determine the seropositivity of participants, indicating previous exposure to LASV. Surveys were administered to each participant to assess demographic and environmental factors associated with a higher risk of exposure to LASV. Overall seroprevalence for antibodies to LASV was 16.0%. In Kenema, Port Loko, and Tonkolili Districts, seroprevalences were 20.1%, 14.1%, and 10.6%, respectively. In a multivariate analysis, individuals were more likely to be LASV seropositive if they were living in Kenema District, regardless of sex, age, or occupation. Environmental factors contributed to an increased risk of LASV exposure, including poor housing construction and proximity to bushland, forested areas, and refuse. CONCLUSIONS AND SIGNIFICANCE In this study we determine a baseline LASV seroprevalence in three districts which will inform future epidemiological, ecological, and clinical studies on LF and the LASV in Sierra Leone. The heterogeneity of the distribution of LASV and LF over both space, and time, can make the design of efficacy trials and intervention programs difficult. Having more studies on the prevalence of LASV and identifying potential hyper-endemic areas will greatly increase the awareness of LF and improve targeted control programs related to LASV.
Collapse
Affiliation(s)
- Donald S. Grant
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health and Sanitation, Kenema, Sierra Leone
- College of Medicine and Allied Health Sciences, University of Sierra Leone, Freetown, Sierra Leone
| | - Emily J. Engel
- Department of Pediatrics, Sections of Pediatric Infectious Diseases, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Nicole Roberts Yerkes
- Department of Pediatrics, Sections of Pediatric Infectious Diseases, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Lansana Kanneh
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health and Sanitation, Kenema, Sierra Leone
| | - James Koninga
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health and Sanitation, Kenema, Sierra Leone
| | - Michael A. Gbakie
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health and Sanitation, Kenema, Sierra Leone
| | - Foday Alhasan
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health and Sanitation, Kenema, Sierra Leone
| | - Franklyn B. Kanneh
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health and Sanitation, Kenema, Sierra Leone
| | - Ibrahim Mustapha Kanneh
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health and Sanitation, Kenema, Sierra Leone
| | - Fatima K. Kamara
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health and Sanitation, Kenema, Sierra Leone
| | - Mambu Momoh
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health and Sanitation, Kenema, Sierra Leone
- Eastern Technical University of Sierra Leone, Kenema, Sierra Leone
| | - Mohamed S. Yillah
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health and Sanitation, Kenema, Sierra Leone
| | - Momoh Foday
- Lassa Fever Program, Kenema Government Hospital, Ministry of Health and Sanitation, Kenema, Sierra Leone
| | - Adaora Okoli
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Ashley Zeoli
- Department of Pediatrics, Sections of Pediatric Infectious Diseases, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Caroline Weldon
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Christopher M. Bishop
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Crystal Zheng
- Department of Internal Medicine, Section of Infectious Diseases, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Jessica Hartnett
- Department of Pediatrics, Sections of Pediatric Infectious Diseases, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Karissa Chao
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Kayla Shore
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Lilia I. Melnik
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Mallory Mucci
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Nell G. Bond
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Philip Doyle
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Rachael Yenni
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Rachel Podgorski
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Samuel C. Ficenec
- Department of Internal Medicine, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Lina Moses
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Jeffrey G. Shaffer
- Department of Biostatistics and Data Science, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Robert F. Garry
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - John S. Schieffelin
- Department of Pediatrics, Sections of Pediatric Infectious Diseases, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| |
Collapse
|
7
|
Aloke C, Obasi NA, Aja PM, Emelike CU, Egwu CO, Jeje O, Edeogu CO, Onisuru OO, Orji OU, Achilonu I. Combating Lassa Fever in West African Sub-Region: Progress, Challenges, and Future Perspectives. Viruses 2023; 15:146. [PMID: 36680186 PMCID: PMC9864412 DOI: 10.3390/v15010146] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
Lassa fever (LF) is a rodent-borne disease that threatens human health in the sub-region of West Africa where the zoonotic host of Lassa virus (LASV) is predominant. Currently, treatment options for LF are limited and since no preventive vaccine is approved for its infectivity, there is a high mortality rate in endemic areas. This narrative review explores the transmission, pathogenicity of LASV, advances, and challenges of different treatment options. Our findings indicate that genetic diversity among the different strains of LASV and their ability to circumvent the immune system poses a critical challenge to the development of LASV vaccines/therapeutics. Thus, understanding the biochemistry, physiology and genetic polymorphism of LASV, mechanism of evading host immunity are essential for development of effective LASV vaccines/therapeutics to combat this lethal viral disease. The LASV nucleoprotein (NP) is a novel target for therapeutics as it functions significantly in several aspects of the viral life cycle. Consequently, LASV NP inhibitors could be employed as effective therapeutics as they will potentially inhibit LASV replication. Effective preventive control measures, vaccine development, target validation, and repurposing of existing drugs, such as ribavirin, using activity or in silico-based and computational bioinformatics, would aid in the development of novel drugs for LF management.
Collapse
Affiliation(s)
- Chinyere Aloke
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg 2050, South Africa
- Department of Medical Biochemistry, Alex Ekwueme Federal University Ndufu-Alike, Abakaliki PMB 1010, Ebonyi State, Nigeria
| | - Nwogo Ajuka Obasi
- Department of Medical Biochemistry, Alex Ekwueme Federal University Ndufu-Alike, Abakaliki PMB 1010, Ebonyi State, Nigeria
| | - Patrick Maduabuchi Aja
- Department of Biochemistry, Faculty of Biological Sciences, Ebonyi State University, Abakaliki PMB 053, Ebonyi State, Nigeria
- Department of Biochemistry, Faculty of Medicine, Mbarara University of Science and Technology (MUST), Mbarara P.O. Box 1410, Uganda
- Department of Medical Biochemistry, Kampala International University, Bushenyi, Ishaka P.O. Box 71, Uganda
| | - Chinedum Uche Emelike
- Department of Physiology, Alex Ekwueme Federal University Ndufu-Alike, Abakaliki PMB 1010, Ebonyi State, Nigeria
| | - Chinedu Ogbonnia Egwu
- Department of Medical Biochemistry, Alex Ekwueme Federal University Ndufu-Alike, Abakaliki PMB 1010, Ebonyi State, Nigeria
| | - Olamide Jeje
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg 2050, South Africa
| | - Chuks Oswald Edeogu
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, Ebonyi State University, Abakaliki PMB 053, Ebonyi State, Nigeria
| | - Olalekan Olugbenga Onisuru
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg 2050, South Africa
| | - Obasi Uche Orji
- Department of Biochemistry, Faculty of Biological Sciences, Ebonyi State University, Abakaliki PMB 053, Ebonyi State, Nigeria
| | - Ikechukwu Achilonu
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg 2050, South Africa
| |
Collapse
|
8
|
The Pan-ErbB tyrosine kinase inhibitor afatinib inhibits multiple steps of the mammarenavirus life cycle. Virology 2022; 576:83-95. [PMID: 36183499 DOI: 10.1016/j.virol.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/02/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022]
Abstract
The mammarenavirus Lassa virus (LASV) causes a life-threatening acute febrile disease, Lassa fever (LF). To date, no US Food and Drug Administration (FDA)-licensed medical countermeasures against LASV are available. This underscores the need for the development of novel anti-LASV drugs. Here, we screen an FDA-approved drug library to identify novel anti-LASV drug candidates using an infectious-free cell line expressing a functional LASV ribonucleoprotein (vRNP), where levels of vRNP-directed reporter gene expression serve as a surrogate for vRNP activity. Our screen identified the pan-ErbB tyrosine kinase inhibitor afatinib as a potent inhibitor of LASV vRNP activity. Afatinib inhibited multiplication of lymphocytic choriomeningitis virus (LCMV) a mammarenavirus closely related to LASV. Cell-based assays revealed that afatinib inhibited multiple steps of the LASV and LCMV life cycles. Afatinib also inhibited multiplication of Junín virus vaccine strain Candid#1, indicating that afatinib can have antiviral activity against a broad range of human pathogenic mammarenaviruses.
Collapse
|
9
|
Cheng HY, French CE, Salam AP, Dawson S, McAleenan A, McGuinness LA, Savović J, Horby PW, Sterne JAC. Lack of Evidence for Ribavirin Treatment of Lassa Fever in Systematic Review of Published and Unpublished Studies 1. Emerg Infect Dis 2022; 28:1559-1568. [PMID: 35876478 PMCID: PMC9328902 DOI: 10.3201/eid2808.211787] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Ribavirin has been used widely to treat Lassa fever in West Africa since the 1980s. However, few studies have systematically appraised the evidence for its use. We conducted a systematic review of published and unpublished literature retrieved from electronic databases and gray literature from inception to March 8, 2022. We identified 13 studies of the comparative effectiveness of ribavirin versus no ribavirin treatment on mortality outcomes, including unpublished data from a study in Sierra Leone provided through a US Freedom of Information Act request. Although ribavirin was associated with decreased mortality rates, results of these studies were at critical or serious risk for bias when appraised using the ROBINS-I tool. Important risks for bias related to lack of control for confounders, immortal time bias, and missing outcome data. Robust evidence supporting the use of ribavirin in Lassa fever is lacking. Well-conducted clinical trials to elucidate the effectiveness of ribavirin for Lassa fever are needed.
Collapse
|
10
|
Raabe V, Mehta AK, Evans JD. Lassa Virus Infection: a Summary for Clinicians. Int J Infect Dis 2022; 119:187-200. [PMID: 35395384 DOI: 10.1016/j.ijid.2022.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES This summary on Lassa virus (LASV) infection and Lassa fever disease (LF) was developed from a clinical perspective to provide clinicians a condensed, accessible understanding of the current literature. The information provided highlights pathogenesis, clinical features, and diagnostics with an emphasis on therapies and vaccines that have demonstrated potential value for use in clinical or research environments. METHODS An integrative literature review was conducted on the clinical and pathological features, vaccines, and treatments for LASV infection, with a focus on recent studies and in vivo evidence from humans and/or non-human primates (NHPs), when available. RESULTS Two antiviral medications with potential benefit for the treatment of LASV infection and one for post-exposure prophylaxis were identified, although a larger number of potential candidates are currently being evaluated. Multiple vaccine platforms are in pre-clinical development for LASV prevention, but data from human clinical trials are not yet available. CONCLUSION We provide succinct summaries of medical countermeasures against LASV to give the busy clinician a rapid reference. Although there are no approved drugs or vaccines for LF, we provide condensed information from a literature review for measures that can be taken when faced with a suspected infection, including investigational treatment options and hospital engineering controls.
Collapse
Affiliation(s)
- Vanessa Raabe
- New York University Grossman School of Medicine, New York, NY.
| | | | - Jared D Evans
- Johns Hopkins Applied Physics Laboratory, Laurel, MD.
| |
Collapse
|
11
|
Joseph AA, Fasipe OJ, Joseph OA, Olatunji OA. Contemporary and emerging pharmacotherapeutic agents for the treatment of Lassa viral haemorrhagic fever disease. J Antimicrob Chemother 2022; 77:1525-1531. [PMID: 35296886 DOI: 10.1093/jac/dkac064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This review was designed to discuss the emerging and current pharmacotherapeutic agents for the treatment of Lassa viral haemorrhagic fever disease (LVHFD), also known as Lassa fever (LF). Original peer-reviewed articles that investigated LF were identified using the Medline Entrez-PubMed search. Information was also sourced from printed textbooks and reports by recognized health professional bodies such as the WHO, CDC, the Nigerian Federal Ministry of Health and the United Nations Children's Fund (UNICEF). A total of 103 articles were reviewed and 78 were found to contain information relevant to the study. LF remains an endemic disease of public health concern in the West Africa region, and in the rest of the world as cases have been imported into non-endemic regions as well. Currently, there are no approved vaccines or therapeutics for the treatment of Lassa mammarenavirus (LASV) infection. There are, however, off-label therapeutics being used (ribavirin and convalescent plasma) whose efficacy is suboptimal. Research is still ongoing on possible therapeutic options and drug repurposing of therapeutic agents currently in use for other clinical conditions. Considered therapeutic options include favipiravir, taribavirin, Arevirumab-3 and experimental drugs such as losmapimod, adamantyl diphenyl piperazine 3.3, Arbidol (umifenovir) and decanoyl-RRLL-chloromethyl ketone (dec-RRLL-CMK). Current treatments for LF are limited, hence the institution of mitigating measures to prevent infection is of utmost importance and should be prioritized, especially in endemic regions. Heightened searches for other therapeutic options with greater efficacy and lower toxicity are still ongoing, as well as for vaccines as the absence of these classifies the disease as a priority disease of high public health impact.
Collapse
Affiliation(s)
| | - Olumuyiwa John Fasipe
- Department of Pharmacology and Therapeutics, University of Medical Sciences, Ondo, Nigeria
| | | | - Olalekan Aliu Olatunji
- Department of Medical Microbiology and Parasitology, University College Hospital, Ibadan, Nigeria
| |
Collapse
|
12
|
Murphy HL, Ly H. Pathogenicity and virulence mechanisms of Lassa virus and its animal modeling, diagnostic, prophylactic, and therapeutic developments. Virulence 2021; 12:2989-3014. [PMID: 34747339 PMCID: PMC8923068 DOI: 10.1080/21505594.2021.2000290] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Lassa fever (LF) is a deadly viral hemorrhagic disease that is endemic to West Africa. The causative agent of LF is Lassa virus (LASV), which causes approximately 300,000 infections and 5,000 deaths annually. There are currently no approved therapeutics or FDA-approved vaccines against LASV. The high genetic variability between LASV strains and immune evasion mediated by the virus complicate the development of effective therapeutics and vaccines. Here, we aim to provide a comprehensive review of the basic biology of LASV and its mechanisms of disease pathogenesis and virulence in various animal models, as well as an update on prospective vaccines, therapeutics, and diagnostics for LF. Until effective vaccines and/or therapeutics are available for use to prevent or treat LF, a better level of understanding of the basic biology of LASV, its natural genetic variations and immune evasion mechanisms as potential pathogenicity factors, and of the rodent reservoir-vector populations and their geographical distributions, is necessary for the development of accurate diagnostics and effective therapeutics and vaccines against this deadly human viral pathogen.
Collapse
Affiliation(s)
- Hannah L Murphy
- Department of Veterinary & Biomedical Sciences, Comparative & Molecular Biosciences Graduate Program, College of Veterinary Medicine, University of Minnesota, Twin Cities
| | - Hinh Ly
- Department of Veterinary & Biomedical Sciences, Comparative & Molecular Biosciences Graduate Program, College of Veterinary Medicine, University of Minnesota, Twin Cities
| |
Collapse
|
13
|
Abstract
Over the past 20 years, Nipah virus (NiV) has emerged as a significant, highly pathogenic bat-borne paramyxovirus causing severe respiratory disease and encephalitis in humans, and human-to-human transmission has been demonstrated in multiple outbreaks. In addition to causing serious illness in humans, NiV is a zoonotic pathogen capable of infecting a wide range of other mammalian species, including pigs and horses. While NiV has caused less than 700 human cases since its discovery in 1998/1999, the involvement of intermediate agricultural hosts can result in significant economic consequences. Owing to the severity of disease, capacity for human-to-human transmission, zoonotic potential, and lack of available approved therapeutic treatment options, NiV has been listed by the World Health Organization in their Blueprint list of priority pathogens as one of the eight most dangerous pathogens to monitor and prepare countermeasures to prevent a pandemic. Here, we discuss progress towards the development of therapeutic measures for the treatment of NiV infection and disease.
Collapse
Affiliation(s)
- Kendra Johnson
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Michelle Vu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Alexander N Freiberg
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
14
|
Davis S, Milechin L, Patel T, Hernandez M, Ciccarelli G, Samsi S, Hensley L, Goff A, Trefry J, Johnston S, Purcell B, Cabrera C, Fleischman J, Reuther A, Claypool K, Rossi F, Honko A, Pratt W, Swiston A. Detecting Pathogen Exposure During the Non-symptomatic Incubation Period Using Physiological Data: Proof of Concept in Non-human Primates. Front Physiol 2021; 12:691074. [PMID: 34552498 PMCID: PMC8451540 DOI: 10.3389/fphys.2021.691074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/05/2021] [Indexed: 12/15/2022] Open
Abstract
Background and Objectives: Early warning of bacterial and viral infection, prior to the development of overt clinical symptoms, allows not only for improved patient care and outcomes but also enables faster implementation of public health measures (patient isolation and contact tracing). Our primary objectives in this effort are 3-fold. First, we seek to determine the upper limits of early warning detection through physiological measurements. Second, we investigate whether the detected physiological response is specific to the pathogen. Third, we explore the feasibility of extending early warning detection with wearable devices. Research Methods: For the first objective, we developed a supervised random forest algorithm to detect pathogen exposure in the asymptomatic period prior to overt symptoms (fever). We used high-resolution physiological telemetry data (aortic blood pressure, intrathoracic pressure, electrocardiograms, and core temperature) from non-human primate animal models exposed to two viral pathogens: Ebola and Marburg (N = 20). Second, to determine reusability across different pathogens, we evaluated our algorithm against three independent physiological datasets from non-human primate models (N = 13) exposed to three different pathogens: Lassa and Nipah viruses and Y. pestis. For the third objective, we evaluated performance degradation when the algorithm was restricted to features derived from electrocardiogram (ECG) waveforms to emulate data from a non-invasive wearable device. Results: First, our cross-validated random forest classifier provides a mean early warning of 51 ± 12 h, with an area under the receiver-operating characteristic curve (AUC) of 0.93 ± 0.01. Second, our algorithm achieved comparable performance when applied to datasets from different pathogen exposures – a mean early warning of 51 ± 14 h and AUC of 0.95 ± 0.01. Last, with a degraded feature set derived solely from ECG, we observed minimal degradation – a mean early warning of 46 ± 14 h and AUC of 0.91 ± 0.001. Conclusion: Under controlled experimental conditions, physiological measurements can provide over 2 days of early warning with high AUC. Deviations in physiological signals following exposure to a pathogen are due to the underlying host’s immunological response and are not specific to the pathogen. Pre-symptomatic detection is strong even when features are limited to ECG-derivatives, suggesting that this approach may translate to non-invasive wearable devices.
Collapse
Affiliation(s)
- Shakti Davis
- Lincoln Laboratory, Massachusetts Institute of Technology, Lexington, MA, United States
| | - Lauren Milechin
- Lincoln Laboratory, Massachusetts Institute of Technology, Lexington, MA, United States
| | - Tejash Patel
- Lincoln Laboratory, Massachusetts Institute of Technology, Lexington, MA, United States
| | - Mark Hernandez
- Lincoln Laboratory, Massachusetts Institute of Technology, Lexington, MA, United States
| | - Greg Ciccarelli
- Lincoln Laboratory, Massachusetts Institute of Technology, Lexington, MA, United States
| | - Siddharth Samsi
- Lincoln Laboratory, Massachusetts Institute of Technology, Lexington, MA, United States
| | - Lisa Hensley
- US Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, United States
| | - Arthur Goff
- US Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, United States
| | - John Trefry
- US Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, United States
| | - Sara Johnston
- US Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, United States
| | - Bret Purcell
- US Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, United States
| | - Catherine Cabrera
- Lincoln Laboratory, Massachusetts Institute of Technology, Lexington, MA, United States
| | - Jack Fleischman
- Lincoln Laboratory, Massachusetts Institute of Technology, Lexington, MA, United States
| | - Albert Reuther
- Lincoln Laboratory, Massachusetts Institute of Technology, Lexington, MA, United States
| | - Kajal Claypool
- Lincoln Laboratory, Massachusetts Institute of Technology, Lexington, MA, United States
| | - Franco Rossi
- US Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, United States
| | - Anna Honko
- US Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, United States
| | - William Pratt
- US Army Medical Research Institute of Infectious Diseases, Ft. Detrick, MD, United States
| | - Albert Swiston
- Lincoln Laboratory, Massachusetts Institute of Technology, Lexington, MA, United States
| |
Collapse
|
15
|
CP100356 Hydrochloride, a P-Glycoprotein Inhibitor, Inhibits Lassa Virus Entry: Implication of a Candidate Pan-Mammarenavirus Entry Inhibitor. Viruses 2021; 13:v13091763. [PMID: 34578344 PMCID: PMC8473031 DOI: 10.3390/v13091763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 11/20/2022] Open
Abstract
Lassa virus (LASV)—a member of the family Arenaviridae—causes Lassa fever in humans and is endemic in West Africa. Currently, no approved drugs are available. We screened 2480 small compounds for their potential antiviral activity using pseudotyped vesicular stomatitis virus harboring the LASV glycoprotein (VSV-LASVGP) and a related prototypic arenavirus, lymphocytic choriomeningitis virus (LCMV). Follow-up studies confirmed that CP100356 hydrochloride (CP100356), a specific P-glycoprotein (P-gp) inhibitor, suppressed VSV-LASVGP, LCMV, and LASV infection with half maximal inhibitory concentrations of 0.52, 0.54, and 0.062 μM, respectively, without significant cytotoxicity. Although CP100356 did not block receptor binding at the cell surface, it inhibited low-pH-dependent membrane fusion mediated by arenavirus glycoproteins. P-gp downregulation did not cause a significant reduction in either VSV-LASVGP or LCMV infection, suggesting that P-gp itself is unlikely to be involved in arenavirus entry. Finally, our data also indicate that CP100356 inhibits the infection by other mammarenaviruses. Thus, our findings suggest that CP100356 can be considered as an effective virus entry inhibitor for LASV and other highly pathogenic mammarenaviruses.
Collapse
|
16
|
Ayotunde OA, Gbenga J, Chuks A, Dare OJ, Temitope A. Lassa fever presenting as a cause of post-operative pyrexia following open femoral interlocking nailing. Trop Doct 2021; 51:608-610. [PMID: 34229506 DOI: 10.1177/00494755211026811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We report a case where Lassa fever was diagnosed in the search for post-operative pyrexia. Protective measures may have prevented any transmission to the attending medical staff.
Collapse
Affiliation(s)
- Olasinde A Ayotunde
- Consultant Orthopaedic Surgeon, Department of Orthopaedic Surgery, Federal Medical Centre, Owo, Ondo, Nigeria
| | - Jones Gbenga
- Consultant Orthopaedic Surgeon, Department of Orthopaedic Surgery, Federal Medical Centre, Owo, Ondo, Nigeria
| | - Abejegah Chuks
- Consultant Public Health Physician, Department of Community Health, Federal Medical Centre, Owo, Ondo, Nigeria
| | - Ogunlusi J Dare
- Consultant Public Health Physician, Department of Community Health, Federal Medical Centre, Owo, Ondo, Nigeria
| | - Akinleye Temitope
- House officer in Orthoapedic Surgery, Department of Surgery, Ekiti State University Teaching Hospital, Ado Ekiti, Nigeria
| |
Collapse
|
17
|
Abstract
Lassa fever (LF) is on the top-priority infections list of both the Food and Drug Administration (FDA) and World Health Organization (WHO). This review explores the different treatment approaches found in the literature within the last 20 years. Even though ribavirin stands out among medication options, only one clinical trial was done to assess its efficacy in humans, which necessitated that we look in-depth about the latest updates in managing LF infection. For that matter, we used a Medical Subject Headings (MeSH) search on PubMed. Inclusion criteria included papers written in the English language and human subjects. Intravenous (IV) ribavirin is the most effective treatment for an acute infection. Post-exposure prophylaxis with oral ribavirin is recommended. There is not sufficient evidence to recommended convalescent plasma for the treatment of Lassa fever. LF continues to be left in the shade from global and scientific attention despite experts expecting a rise in current and future infections due to the Lassa fever virus (LFV).
Collapse
Affiliation(s)
- Ammar Alli
- Medicine, Tishreen University Faculty of Medicine, Lattakia, SYR.,Internal Medicine, Universitat de Barcelona, Barcelona, ESP
| | - Juan Fernando Ortiz
- Neurology, Universidad San Francisco de Quito, Quito, ECU.,Neurology, Larkin Community Hospital, Miami, USA
| | - Stephanie P Fabara
- Internal Medicine, Universidad Católica de Santiago de Guayaquil, Guayaquil, ECU
| | - Amrapali Patel
- Public Health, George Washington University, Washington , USA
| | - Taras Halan
- General Medicine, Ternopil National Medical University, Ternopil, UKR
| |
Collapse
|
18
|
Xu J, Xue Y, Zhou R, Shi PY, Li H, Zhou J. Drug repurposing approach to combating coronavirus: Potential drugs and drug targets. Med Res Rev 2021; 41:1375-1426. [PMID: 33277927 PMCID: PMC8044022 DOI: 10.1002/med.21763] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/03/2020] [Accepted: 11/20/2020] [Indexed: 01/18/2023]
Abstract
In the past two decades, three highly pathogenic human coronaviruses severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus, and, recently, SARS-CoV-2, have caused pandemics of severe acute respiratory diseases with alarming morbidity and mortality. Due to the lack of specific anti-CoV therapies, the ongoing pandemic of coronavirus disease 2019 (COVID-19) poses a great challenge to clinical management and highlights an urgent need for effective interventions. Drug repurposing is a rapid and feasible strategy to identify effective drugs for combating this deadly infection. In this review, we summarize the therapeutic CoV targets, focus on the existing small molecule drugs that have the potential to be repurposed for existing and emerging CoV infections of the future, and discuss the clinical progress of developing small molecule drugs for COVID-19.
Collapse
Affiliation(s)
- Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yu Xue
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Richard Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Hongmin Li
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, New York, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
19
|
Yaro CA, Kogi E, Opara KN, Batiha GES, Baty RS, Albrakati A, Altalbawy FMA, Etuh IU, Oni JP. Infection pattern, case fatality rate and spread of Lassa virus in Nigeria. BMC Infect Dis 2021; 21:149. [PMID: 33546623 PMCID: PMC7863503 DOI: 10.1186/s12879-021-05837-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Lassa fever (LF) is a zoonotic infectious disease of public concern in Nigeria. The infection dynamics of the disease is not well elucidated in Nigeria. This study was carried out to describe the pattern of infection, case fatality rate and spread of lassa virus (LASV) from 2017 to 2020. METHODS Weekly epidemiological data on LF from December, 2016 to September, 2020 were obtained from Nigeria Centre for Disease Control. The number of confirmed cases and deaths were computed according to months and states. Descriptive statistics was performed and case fatality rate was calculated. Distribution and spread maps of LF over the four years period was performed on ArcMap 10.7. RESULTS A total of 2787 confirmed cases and 516 deaths were reported in Nigeria from December, 2016 to September, 2020. Increase in number of cases and deaths were observed with 298, 528, 796 and 1165 confirmed cases and 79, 125, 158 and 158 deaths in 2017, 2018, 2019 and 2020 respectively. Over 60% of the cases were reported in two states, Edo and Ondo states. The LF cases spread from 19 states in 2017 to 32 states and Federal Capital Territory (FCT) in 2020. Ondo state (25.39%) had the highest of deaths rate from LF over the four years. Case fatality rate (CFR) of LF was highest in 2017 (26.5%) with CFR of 23.7, 19.6 and 13.4% in 2018, 2019 and 2020 respectively. The peak of infection was in the month of February for the four years. Infections increases at the onset of dry season in November and decline till April when the wet season sets-in. CONCLUSION There is an annual increase in the number of LASV infection across the states in Nigeria. There is need to heighten control strategies through the use of integrated approach, ranging from vector control, health education and early diagnosis.
Collapse
Affiliation(s)
- Clement Ameh Yaro
- Department of Animal and Environmental Biology, University of Uyo, Uyo, Akwa Ibom Nigeria
- Department of Zoology, Ahmadu Bello University, Zaria, Nigeria
| | - Ezekiel Kogi
- Department of Zoology, Ahmadu Bello University, Zaria, Nigeria
| | - Kenneth Nnamdi Opara
- Department of Animal and Environmental Biology, University of Uyo, Uyo, Akwa Ibom Nigeria
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, AlBeheira 22511 Egypt
| | - Roua S. Baty
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif, 21944 Saudi Arabia
| | - Ashraf Albrakati
- Department of Human Anatomy, College of Medicine, Taif University, P.O. Box 11099, Taif, 21944 Saudi Arabia
| | - Farag M. A. Altalbawy
- National Institute of Laser Enhanced Sciences (NILES), Cairo University, Giza, 12613 Egypt
| | | | - James Paul Oni
- Department of Animal and Environmental Biology, Kogi State University, Anyigba, Nigeria
| |
Collapse
|
20
|
Lingas G, Rosenke K, Safronetz D, Guedj J. Lassa viral dynamics in non-human primates treated with favipiravir or ribavirin. PLoS Comput Biol 2021; 17:e1008535. [PMID: 33411731 PMCID: PMC7817048 DOI: 10.1371/journal.pcbi.1008535] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 01/20/2021] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
Lassa fever is an haemorrhagic fever caused by Lassa virus (LASV). There is no vaccine approved against LASV and the only recommended antiviral treatment relies on ribavirin, despite limited evidence of efficacy. Recently, the nucleotide analogue favipiravir showed a high antiviral efficacy, with 100% survival obtained in an otherwise fully lethal non-human primate (NHP) model of Lassa fever. However the mechanism of action of the drug is not known and the absence of pharmacokinetic data limits the translation of these results to the human setting. Here we aimed to better understand the antiviral effect of favipiravir by developping the first mathematical model recapitulating Lassa viral dynamics and treatment. We analyzed the viral dynamics in 24 NHPs left untreated or treated with ribavirin or favipiravir, and we put the results in perspective with those obtained with the same drugs in the context of Ebola infection. Our model estimates favipiravir EC50 in vivo to 2.89 μg.mL-1, which is much lower than what was found against Ebola virus. The main mechanism of action of favipiravir was to decrease virus infectivity, with an efficacy of 91% at the highest dose. Based on our knowledge acquired on the drug pharmacokinetics in humans, our model predicts that favipiravir doses larger than 1200 mg twice a day should have the capability to strongly reduce the production infectious virus and provide a milestone towards a future use in humans.
Collapse
Affiliation(s)
| | - Kyle Rosenke
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana, USA
| | - David Safronetz
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada.,Zoonotic Diseases and Special Pathogens, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | | |
Collapse
|
21
|
Moreno H, Rastrojo A, Pryce R, Fedeli C, Zimmer G, Bowden TA, Gerold G, Kunz S. A novel circulating tamiami mammarenavirus shows potential for zoonotic spillover. PLoS Negl Trop Dis 2020; 14:e0009004. [PMID: 33370288 PMCID: PMC7794035 DOI: 10.1371/journal.pntd.0009004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 01/08/2021] [Accepted: 11/23/2020] [Indexed: 11/18/2022] Open
Abstract
A detailed understanding of the mechanisms underlying the capacity of a virus to break the species barrier is crucial for pathogen surveillance and control. New World (NW) mammarenaviruses constitute a diverse group of rodent-borne pathogens that includes several causative agents of severe viral hemorrhagic fever in humans. The ability of the NW mammarenaviral attachment glycoprotein (GP) to utilize human transferrin receptor 1 (hTfR1) as a primary entry receptor plays a key role in dictating zoonotic potential. The recent isolation of Tacaribe and lymphocytic choriominingitis mammarenaviruses from host-seeking ticks provided evidence for the presence of mammarenaviruses in arthropods, which are established vectors for numerous other viral pathogens. Here, using next generation sequencing to search for other mammarenaviruses in ticks, we identified a novel replication-competent strain of the NW mammarenavirus Tamiami (TAMV-FL), which we found capable of utilizing hTfR1 to enter mammalian cells. During isolation through serial passaging in mammalian immunocompetent cells, the quasispecies of TAMV-FL acquired and enriched mutations leading to the amino acid changes N151K and D156N, within GP. Cell entry studies revealed that both substitutions, N151K and D156N, increased dependence of the virus on hTfR1 and binding to heparan sulfate proteoglycans. Moreover, we show that the substituted residues likely map to the sterically constrained trimeric axis of GP, and facilitate viral fusion at a lower pH, resulting in viral egress from later endosomal compartments. In summary, we identify and characterize a naturally occurring TAMV strain (TAMV-FL) within ticks that is able to utilize hTfR1. The TAMV-FL significantly diverged from previous TAMV isolates, demonstrating that TAMV quasispecies exhibit striking genetic plasticity that may facilitate zoonotic spillover and rapid adaptation to new hosts. Mammarenaviruses include emergent pathogens responsible of severe disease in humans in zoonotic events. The ability to use the human Transferrin receptor 1 (hTfR1) strongly correlates with their pathogenicity in humans. We isolated a new infectious Tamiami virus strain (TAMV-FL) from host-seeking ticks, which, contrary to the previous rodent-derived reference strain, can use hTfR1 to enter human cells. Moreover, serial passaging of TAMV-FL in human immunocompetent cells selected for two substitutions in the viral envelope glycoprotein: N151K and D156N. These substitutions increase the ability to highjack hTfR1 and the binding capacity to heparan sulfate proteoglycans and cause delayed endosomal escape. Our findings provide insight into the acquisition of novel traits by currently circulating TAMV that increase its potential to trespass the inter-species barrier.
Collapse
Affiliation(s)
- Hector Moreno
- Institute of Microbiology, Lausanne University Hospital (IMUL-CHUV), Lausanne, Switzerland
- * E-mail:
| | - Alberto Rastrojo
- Department of Virology and Microbiology, Centro de Biología Molecular Severo Ochoa (CBMSO-CSIC), Madrid, Spain
- Genetic Unit, Department of Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rhys Pryce
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
| | - Chiara Fedeli
- Institute of Microbiology, Lausanne University Hospital (IMUL-CHUV), Lausanne, Switzerland
| | - Gert Zimmer
- Institute of Virology and Immunology (IVI), Mittelhäusern, Switzerland
- Department of Infectious Diseases and Pathobiology (DIP), Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, United Kingdom
| | - Gisa Gerold
- TWINCORE -Center for Experimental and Clinical Infection Research, Institute for Experimental Virology, Hannover, Germany
- Department of Clinical Microbiology, Virology & Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover Germany
| | - Stefan Kunz
- Institute of Microbiology, Lausanne University Hospital (IMUL-CHUV), Lausanne, Switzerland
| |
Collapse
|
22
|
Overbosch F, de Boer M, Veldkamp KE, Ellerbroek P, Bleeker-Rovers CP, Goorhuis B, van Vugt M, van der Eijk A, Leenstra T, Khargi M, Ros J, Brandwagt D, Haverkate M, Swaan C, Reusken C, Timen A, Koopmans M, van Dissel J. Public health response to two imported, epidemiologically related cases of Lassa fever in the Netherlands (ex Sierra Leone), November 2019. ACTA ACUST UNITED AC 2020; 25. [PMID: 32317052 PMCID: PMC7175652 DOI: 10.2807/1560-7917.es.2020.25.15.2000265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
On 20 November 2019, Lassa fever was diagnosed in a physician repatriated from Sierra Leone to the Netherlands. A second physician with suspected Lassa fever, repatriated a few days later from the same healthcare facility, was confirmed infected with Lassa virus on 21 November. Comprehensive contact monitoring involving high- and low-risk contacts proved to be feasible and follow-up of the contacts did not reveal any case of secondary transmission in the Netherlands.
Collapse
Affiliation(s)
- Femke Overbosch
- Stichting BeroepsOpleiding Huisartsen (SBOH), Utrecht, the Netherlands
| | - Mark de Boer
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Karin Ellen Veldkamp
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Pauline Ellerbroek
- Department of Infectious Diseases, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Chantal P Bleeker-Rovers
- Department of Internal Medicine, Division of Infectious Diseases, Radboud university medical center, Nijmegen, the Netherlands
| | - Bram Goorhuis
- Centre of Tropical Medicine and Travel Medicine, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands
| | - Michele van Vugt
- Centre of Tropical Medicine and Travel Medicine, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands
| | - Annemiek van der Eijk
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tjalling Leenstra
- Department of Infectious Diseases, Public Health Service Amsterdam, Amsterdam, the Netherlands
| | - Martin Khargi
- Department of Infectious Diseases, Public Health Service Hollands Midden, Leiden, the Netherlands
| | - Jeanette Ros
- Department of Infectious Diseases, Public Health Service Kennemerland, Haarlem, the Netherlands
| | - Diederik Brandwagt
- Department of Infectious Diseases, Public Health Service region Utrecht, Zeist, the Netherlands
| | - Manon Haverkate
- National Institute for Public Health and the Environment (RIVM), Centre for Communicable Disease Control (CIb), Bilthoven, the Netherlands
| | - Corien Swaan
- National Institute for Public Health and the Environment (RIVM), Centre for Communicable Disease Control (CIb), Bilthoven, the Netherlands
| | - Chantal Reusken
- National Institute for Public Health and the Environment (RIVM), Centre for Communicable Disease Control (CIb), Bilthoven, the Netherlands.,Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Aura Timen
- Athena Institute, VU University Amsterdam, the Netherlands.,National Institute for Public Health and the Environment (RIVM), Centre for Communicable Disease Control (CIb), Bilthoven, the Netherlands
| | - Marion Koopmans
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jaap van Dissel
- National Institute for Public Health and the Environment (RIVM), Centre for Communicable Disease Control (CIb), Bilthoven, the Netherlands.,Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | -
- The members of the Lassa fever response team of the Netherlands have been listed at the end of this article
| |
Collapse
|
23
|
Balakrishnan A, Mun AB. Ribavirin inhibits Chandipura virus replication in Vero cells. J Med Virol 2020; 92:2969-2975. [PMID: 32543712 DOI: 10.1002/jmv.26184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/10/2020] [Indexed: 01/04/2023]
Abstract
Chandipura virus (CHPV) is an emerging tropical pathogen in India. The virus has been reported to be associated with an acute encephalitis syndrome in young children with a case fatality rate of 55% to 75%. Clinical management with symptomatic treatment is the only option available to treat infected patients. No vaccines are available for prophylaxis. In light of the prophylactic limitations, antiviral therapy would play an important role in control of CHPV infection. In the present study, ribavirin (RBV), an antiviral drug widely accepted for human use and having an antiviral effect on many RNA and DNA viruses, was tested against the CHPV. A screening assay that scores for the virus-mediated plaque formation in the cultured Vero cells was used. RBV exhibited 50% inhibitory concentration (IC50 ) at 89.84 ± 1.8 µM. The drug was very effective when the cells were treated either within an hour postinfection or 4 to 6 hours before infection. The plaque morphology was different in RBV treated cells; the plaques were smaller in size as compared with the plaques in the virus infected cells. The study reports the antiviral activity of RBV against CHPV, and hence, suggests the possible utility of RBV in CHPV infected patients to mitigate the disease. A further clinical trial is needed before introducing the drug for human use against CHPV infection.
Collapse
Affiliation(s)
| | - Amol B Mun
- ICMR-National Institute of Research in Tribal Health, Jabalpur, Madhya Pradesh, India
| |
Collapse
|
24
|
Teimury A, Mahmoodi Khaledi E. Current Options in the Treatment of COVID-19: A Review. Risk Manag Healthc Policy 2020; 13:1999-2010. [PMID: 33116980 PMCID: PMC7549493 DOI: 10.2147/rmhp.s265030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022] Open
Abstract
Novel Coronavirus, also known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in December 2019 in China and spread rapidly all around the world infecting many people. To date, no specific vaccines and drugs have been developed for this disease. Also, due to the COVID-19 pandemic and high prevalence of the infected patients, the drugs and the therapies of other past viral epidemics have been used for this disease. Many studies have been performed on the specific treatments to find whether or not they are effective on COVID-19 patients. In this review, we collected information about the most widely used drugs to treat COVID-19 (coronavirus disease 2019) belonging to groups of antivirals, antibiotics, immune modulators, and anticoagulants. Some of these compounds and drugs were used directly by inpatients, so researchers have examined others in laboratory conditions. This study considered the pros and cons of using these treatments separately and together and compared their results. By studying this review, we hope to provide useful information for researchers.
Collapse
Affiliation(s)
- Azadeh Teimury
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Elahe Mahmoodi Khaledi
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran
| |
Collapse
|
25
|
Affiliation(s)
- S. A. El‐Sebaey
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Girls) Al-Azhar University Youssef Abbas street, Nasr City Cairo Egypt
| |
Collapse
|
26
|
Iversen PL, Kane CD, Zeng X, Panchal RG, Warren TK, Radoshitzky SR, Kuhn JH, Mudhasani RR, Cooper CL, Shurtleff AC, Nasar F, Sunay MM, Duplantier AJ, Eaton BP, Zumbrun EE, Bixler SL, Martin S, Meinig JM, Chiang CY, Sanchez-Lockhart M, Palacios GF, Kugelman JR, Martins KA, Pitt ML, Crozier I, Saunders DL. Recent successes in therapeutics for Ebola virus disease: no time for complacency. THE LANCET. INFECTIOUS DISEASES 2020; 20:e231-e237. [PMID: 32563280 PMCID: PMC7302789 DOI: 10.1016/s1473-3099(20)30282-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/20/2022]
Abstract
The PALM trial in the Democratic Republic of the Congo identified a statistically significant survival benefit for two monoclonal antibody-based therapeutics in the treatment of acute Ebola virus disease; however, substantial gaps remain in improving the outcomes of acute Ebola virus disease and for the survivors. Ongoing efforts are needed to develop more effective strategies, particularly for individuals with severe disease, for prevention and treatment of viral persistence in immune-privileged sites, for optimisation of post-exposure prophylaxis, and to increase therapeutic breadth. As antibody-based approaches are identified and advanced, promising small-molecule antivirals currently in clinical stage development should continue to be evaluated for filovirus diseases, with consideration of their added value in combination approaches with bundled supportive care, their penetration in tissues of interest, the absence of interaction with glycoprotein-based vaccines, and filoviral breadth.
Collapse
Affiliation(s)
- Patrick L Iversen
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Christopher D Kane
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Rekha G Panchal
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Travis K Warren
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Sheli R Radoshitzky
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Rajini R Mudhasani
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Christopher L Cooper
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Amy C Shurtleff
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Farooq Nasar
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Melek Me Sunay
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Allen J Duplantier
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Brett P Eaton
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Elizabeth E Zumbrun
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Sandra L Bixler
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Shannon Martin
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - J Matthew Meinig
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Chih-Yuan Chiang
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Mariano Sanchez-Lockhart
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Gustavo F Palacios
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Jeffrey R Kugelman
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Karen A Martins
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Margaret L Pitt
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Ian Crozier
- Integrated Research Facility at Fort Detrick, Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - David L Saunders
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA.
| |
Collapse
|
27
|
Tewogbola P, Aung N. Lassa fever: History, causes, effects, and reduction strategies. INTERNATIONAL JOURNAL OF ONE HEALTH 2020. [DOI: 10.14202/ijoh.2020.95-98] [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] Open
Abstract
Lassa fever is a disease that is not well-known worldwide, particularly due to the inability of the multimammate rat, the primary vector of the Lassa virus, to breed in temperate regions. The aim of this review is to provide an overview of the disease and its modus operandi while also providing information about trends in the past decade, as well as proven strategies that have been used to manage its spread.
Collapse
Affiliation(s)
- Promise Tewogbola
- School of Psychological and Behavioral Sciences, Southern Illinois University, Carbondale, 62901, USA
| | - Norah Aung
- Department of Health Sciences and Social Work, Western Illinois University, Macomb, 61455, USA
| |
Collapse
|
28
|
Wolf T, Ellwanger R, Goetsch U, Wetzstein N, Gottschalk R. Fifty years of imported Lassa fever: a systematic review of primary and secondary cases. J Travel Med 2020; 27:5808990. [PMID: 32219400 DOI: 10.1093/jtm/taaa035] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 03/08/2020] [Indexed: 01/15/2023]
Abstract
RATIONALE FOR SYSTEMATIC REVIEW Lassa fever is the most common cause of imported haemorrhagic fevers cases in non-endemic countries. As a disease with a high case fatality rate that has regularly caused clusters of nosocomial transmission in endemic areas, prompt diagnosis is vital. We conducted a systematic review of imported cases of the last 50 years with the aim of defining the clinical and epidemiological characteristics that will enhance early diagnosis, prompt initiation of treatment and an appropriate public health response to Lassa fever cases. METHODS We performed a retrospective, systematic review of 36 primary and two secondary cases of Lassa fever in non-endemic countries outside West Africa by searching the PubMed database. This yielded 56 relevant publications that were included in our analysis. RESULTS The case fatality rate of 35.1% for imported cases was higher than that reported for endemic countries. The majority of patients showed clinical features consistent with Lassa fever and had a typical exposure. There was a considerable delay in diagnosis in imported cases with high associated numbers of contacts. Ribavirin was rarely used for post-exposure prophylaxis. Only two secondary transmissions occurred. Thirty-one percent of patients received Lassa fever-specific treatment and five required intensive care. CONCLUSIONS Although importation of Lassa fever to non-endemic countries is a rare event, it has repeatedly happened over five decades. Suspicion of Lassa fever should be based on careful consideration of clinical features and exposure history in order to assist early diagnosis in returning travellers from West Africa.
Collapse
Affiliation(s)
- Timo Wolf
- Department of Internal Medicine II-Infectious Diseases, University Hospital Frankfurtxs, Frankfurt, Germany
| | - Regina Ellwanger
- Department of Internal Medicine II-Infectious Diseases, University Hospital Frankfurtxs, Frankfurt, Germany.,Health Protection Authority, Municipality of Frankfurt am Main, Frankfurt, Germany
| | - Udo Goetsch
- Health Protection Authority, Municipality of Frankfurt am Main, Frankfurt, Germany
| | - Nils Wetzstein
- Department of Internal Medicine II-Infectious Diseases, University Hospital Frankfurtxs, Frankfurt, Germany
| | - Rene Gottschalk
- Health Protection Authority, Municipality of Frankfurt am Main, Frankfurt, Germany.,Institute of Medical Virology, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
29
|
Usifoh SF, Odigie AE, Ighedosa SU, Uwagie-Ero EA, Aighewi IT. Lassa Fever-associated Stigmatization among Staff and Students of the University of Benin, Nigeria. J Epidemiol Glob Health 2020; 9:107-115. [PMID: 31241868 PMCID: PMC7310750 DOI: 10.2991/jegh.k.190514.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/21/2019] [Indexed: 11/10/2022] Open
Abstract
Lassa Fever (LF) remains a health burden in several endemic areas of Nigeria, and its toll remains unabated over several decades. Although most studies have focused on virological and clinical considerations, few studies have attempted to address the perceived psychosocial component of LF disease in Nigeria. Evaluation of stigmatization and discrimination faced by LF survivors is an important step in improving individual health and protecting public health. This study aimed to assess LF-associated stigmatization associated among staff and students of the University of Benin. Descriptive analyses of 600 consenting respondents (300 staff and 300 students) sampled using pretested questionnaires was conducted, and the Chi-square test was used to test for significant association between perceived LF stigmatization and predefined variables. LF was a potential cause of stigmatization in a higher proportion of student (n = 162, 57.9%) than staff (n = 112, 39.9%). LF-associated stigmatization among students was significantly associated with sex (p = 0.012) and poor knowledge (p = 0.013) of LF transmission and prevention. A greater tendency for stigmatization was observed among females than males. A comprehensive emergency response plan incorporating accurate knowledge dissemination about the disease may be a first step toward tackling perceived LF stigmatization.
Collapse
Affiliation(s)
- Stella Folajole Usifoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, University of Benin, Benin City, Nigeria
| | - Amienwanlen Eugene Odigie
- Department of Public Health and Preventive Medicine, Faculty of Veterinary Medicine, University of Benin, Benin City, Nigeria
| | | | | | - Isoken Tito Aighewi
- Department of Environmental Management and Toxicology, Faculty of Life Sciences, University of Benin, Benin City, Nigeria
| |
Collapse
|
30
|
Chen H, Lao Z, Xu J, Li Z, Long H, Li D, Lin L, Liu X, Yu L, Liu W, Li G, Wu J. Antiviral activity of lycorine against Zika virus in vivo and in vitro. Virology 2020; 546:88-97. [PMID: 32452420 PMCID: PMC7194111 DOI: 10.1016/j.virol.2020.04.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/09/2022]
Abstract
The emergence and re-emergence of Zika virus (ZIKV), is a cause for international concern. These highly pathogenic arboviruses represent a serious health burden in tropical and subtropical areas worldwide. Despite these burdens, antiviral therapies do not exist, and inhibitors of ZIKV are therefore urgently needed. To elucidate the anti-ZIKV effect of lycorine, we used reverse transcription-quantitative real-time PCR (qRT-PCR), immunofluorescence, Westernwestern blot, and plaque forming assay to analyse viral RNA (vRNA), viral protein, progeny virus counts, and validated inhibitors in vitro using a variety of cell lines. Additionally, we found that lycorine acts post-infection according to time-of-addition assay, and inhibits RdRp activity. Lycorine protected AG6 mice against ZIKV-induced lethality by decreasing the viral load in the blood. Due to its potency and ability to target ZIKV infection in vivo and in vitro, lycorine might offer promising therapeutic possibilities for combatting ZIKV infections in the future.
Collapse
Affiliation(s)
- Huini Chen
- Guangdong Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, 510632, China
| | - Zizhao Lao
- Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jiangtao Xu
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhaoxin Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Haishan Long
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Detang Li
- Department of Pharmacy, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Luping Lin
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510060, China
| | - Xiaohong Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Liangwen Yu
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Weiyong Liu
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Geng Li
- Guangdong Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, 510632, China; Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Jianguo Wu
- Guangdong Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
31
|
Cai Y, Iwasaki M, Motooka D, Liu DX, Yu S, Cooper K, Hart R, Adams R, Burdette T, Postnikova EN, Kurtz J, St Claire M, Ye C, Kuhn JH, Martínez-Sobrido L, de la Torre JC. A Lassa Virus Live-Attenuated Vaccine Candidate Based on Rearrangement of the Intergenic Region. mBio 2020; 11:e00186-20. [PMID: 32209677 PMCID: PMC7157513 DOI: 10.1128/mbio.00186-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 02/21/2020] [Indexed: 01/31/2023] Open
Abstract
Lassa virus (LASV) poses a significant public health problem within the regions of Lassa fever endemicity in Western Africa. LASV infects several hundred thousand individuals yearly, and a considerable number of Lassa fever cases are associated with high morbidity and lethality. No approved LASV vaccine is available, and current therapy is limited to an off-label usage of ribavirin that is only partially effective and associated with significant side effects. The impact of Lassa fever on human health, together with the limited existing countermeasures, highlights the importance of developing effective vaccines against LASV. Here, we present the development and characterization of a recombinant LASV (rLASV) vaccine candidate [rLASV(IGR/S-S)], which is based on the presence of the noncoding intergenic region (IGR) of the small (S) genome segment (S-IGR) in both large (L) and S LASV segments. In cultured cells, rLASV(IGR/S-S) was modestly less fit than wild-type rLASV (rLASV-WT). rLASV(IGR/S-S) was highly attenuated in guinea pigs, and a single subcutaneous low dose of the virus completely protected against otherwise lethal infection with LASV-WT. Moreover, rLASV(IGR/S-S) was genetically stable during serial passages in cultured cells. These findings indicate that rLASV(IGR/S-S) can be developed into a LASV live-attenuated vaccine (LAV) that has the same antigenic composition as LASV-WT and a well-defined mechanism of attenuation that overcomes concerns about increased virulence that could be caused by genetic changes in the LAV during multiple rounds of multiplication.IMPORTANCE Lassa virus (LASV), the causative agent of Lassa fever, infects several hundred thousand people in Western Africa, resulting in many lethal Lassa fever cases. No U.S. Food and Drug Administration-licensed countermeasures are available to prevent or treat LASV infection. We describe the generation of a novel LASV live-attenuated vaccine candidate rLASV(IGR/S-S), which is based on the replacement of the large genomic segment noncoding intergenic region (IGR) with that of the small genome segment. rLASV(IGR/S-S) is less fit in cell culture than wild-type virus and does not cause clinical signs in inoculated guinea pigs. Importantly, rLASV(IGR/S-S) protects immunized guinea pigs against an otherwise lethal exposure to LASV.
Collapse
Affiliation(s)
- Yingyun Cai
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Masaharu Iwasaki
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
- Laboratory of Emerging Viral Diseases, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Daisuke Motooka
- Laboratory of Pathogen Detection and Identification, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - David X Liu
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Shuiqing Yu
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Kurt Cooper
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Randy Hart
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Ricky Adams
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Tracey Burdette
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Elena N Postnikova
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Jonathan Kurtz
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Marisa St Claire
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Chengjin Ye
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, Maryland, USA
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Juan Carlos de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
32
|
Madelain V, Duthey A, Mentré F, Jacquot F, Solas C, Lacarelle B, Vallvé A, Barron S, Barrot L, Mundweiler S, Thomas D, Carbonnelle C, Raoul H, de Lamballerie X, Guedj J. Ribavirin does not potentiate favipiravir antiviral activity against Ebola virus in non-human primates. Antiviral Res 2020; 177:104758. [PMID: 32135218 DOI: 10.1016/j.antiviral.2020.104758] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 01/26/2023]
Abstract
BACKGROUND In spite of recurrent and dramatic outbreaks, there are no therapeutics approved against Ebola virus disease. Favipiravir, a RNA polymerase inhibitor active against several RNA viruses, recently demonstrated significant but not complete protection in a non-human primate model of Ebola virus disease. In this study, we assessed the benefit of the combination of favipiravir and ribavirin, another broad spectrum antiviral agent, in the same model. METHODS 15 female cynomolgus macaques were challenged intramuscularly with 1,000 FFU of Ebola virus Gabon 2001 strain and followed for 21 days. All animals received favipiravir 180 mg/kg twice a day (BID), either as monotherapy (n = 5) or in combination with ribavirin (n = 10). Ribavirin was given either at the dose 10 mg/kg BID (n = 5) or 5 mg/kg BID (n = 5). Favipiravir and ribavirin were initiated two and one days before viral challenge respectively and treatment were continued for 14 days. Treatment effects on viral and hematological markers were assessed using a mathematical model. Survival rate of 0% and 20% were obtained in macaques receiving favipiravir plus ribavirin 10 and 5 mg/kg BID, respectively, compared to 40% in the favipiravir monotherapy group (P = 0.061 when comparing monotherapy and bitherapy, log rank). Viral dynamic modeling analysis did not identify an association between plasma concentrations of ribavirin and viral load levels. Using a model of erythropoiesis, plasma concentrations of ribavirin were strongly associated with a hemoglobin drop (p = 0.0015). CONCLUSION Ribavirin plus favipiravir did not extend survival rates and did not lower viral replication rate compared to favipiravir monotherapy in this animal model. Patients receiving this combination in other indications, such as Lassa fever, should be closely monitored to prevent potential toxicity associated with anemia.
Collapse
Affiliation(s)
| | - Aurélie Duthey
- Laboratoire P4 Inserm-Jean Mérieux, US003 Inserm, 69365, Lyon, France
| | - France Mentré
- Université de Paris, IAME, INSERM, F-75018, Paris, France
| | - Frédéric Jacquot
- Laboratoire P4 Inserm-Jean Mérieux, US003 Inserm, 69365, Lyon, France
| | - Caroline Solas
- Aix-Marseille Univ, APHM, UMR "Emergence des Pathologies Virales" IRD190-Inserm1207-EHESP, Laboratoire Pharmacocinétique-Toxicologie, Hôpital La Timone, 13005, Marseille, France
| | - Bruno Lacarelle
- Aix-Marseille Univ, APHM, UMR "Emergence des Pathologies Virales" IRD190-Inserm1207-EHESP, Laboratoire Pharmacocinétique-Toxicologie, Hôpital La Timone, 13005, Marseille, France
| | - Audrey Vallvé
- Laboratoire P4 Inserm-Jean Mérieux, US003 Inserm, 69365, Lyon, France
| | - Stéphane Barron
- Laboratoire P4 Inserm-Jean Mérieux, US003 Inserm, 69365, Lyon, France
| | - Laura Barrot
- Laboratoire P4 Inserm-Jean Mérieux, US003 Inserm, 69365, Lyon, France
| | | | - Damien Thomas
- Laboratoire P4 Inserm-Jean Mérieux, US003 Inserm, 69365, Lyon, France
| | | | - Hervé Raoul
- Laboratoire P4 Inserm-Jean Mérieux, US003 Inserm, 69365, Lyon, France
| | - Xavier de Lamballerie
- UMR "Emergence des Pathologies Virales" (EPV: Aix-Marseille University - IRD 190 - Inserm 1207 - EHESP), Institut Hospitalo-Universitaire Méditerranée Infection, F-13385, Marseille, France
| | - Jérémie Guedj
- Université de Paris, IAME, INSERM, F-75018, Paris, France.
| |
Collapse
|
33
|
Cai Y, Ye C, Cheng B, Nogales A, Iwasaki M, Yu S, Cooper K, Liu DX, Hart R, Adams R, Brady T, Postnikova EN, Kurtz J, St Claire M, Kuhn JH, de la Torre JC, Martínez-Sobrido L. A Lassa Fever Live-Attenuated Vaccine Based on Codon Deoptimization of the Viral Glycoprotein Gene. mBio 2020; 11:e00039-20. [PMID: 32098811 PMCID: PMC7042690 DOI: 10.1128/mbio.00039-20] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Lassa virus (LASV) is endemic in Western Africa and is estimated to infect hundreds of thousands of individuals annually. A considerable number of these infections result in Lassa fever (LF), which is associated with significant morbidity and a case-fatality rate as high as 69% among hospitalized confirmed patients. U.S. Food and Drug Administration-approved LF vaccines are not available. Current antiviral treatment is limited to off-label use of a nucleoside analogue, ribavirin, that is only partially effective and associated with significant side effects. We generated and characterized a recombinant LASV expressing a codon-deoptimized (CD) glycoprotein precursor gene (GPC), rLASV-GPC/CD. Comparison of growth kinetics and peak titers showed that rLASV-GPC/CD is slightly attenuated in cell culture compared to wild-type (WT) recombinant LASV (rLASV-WT). However, rLASV-GPC/CD is highly attenuated in strain 13 and Hartley guinea pigs, as reflected by the absence of detectable clinical signs in animals inoculated with rLASV-GPC/CD. Importantly, a single subcutaneous dose of rLASV-GPC/CD provides complete protection against an otherwise lethal exposure to LASV. Our results demonstrate the feasibility of implementing a CD approach for developing a safe and effective LASV live-attenuated vaccine candidate. Moreover, rLASV-GPC/CD might provide investigators with a tool to safely study LASV outside maximum (biosafety level 4) containment, which could accelerate the elucidation of basic aspects of the molecular and cell biology of LASV and the development of novel LASV medical countermeasures.IMPORTANCE Lassa virus (LASV) infects several hundred thousand people in Western Africa, resulting in many lethal Lassa fever (LF) cases. Licensed LF vaccines are not available, and anti-LF therapy is limited to off-label use of the nucleoside analog ribavirin with uncertain efficacy. We describe the generation of a novel live-attenuated LASV vaccine candidate. This vaccine candidate is based on mutating wild-type (WT) LASV in a key region of the viral genome, the glycoprotein precursor (GPC) gene. These mutations do not change the encoded GPC but interfere with its production in host cells. This mutated LASV (rLASV-GPC/CD) behaves like WT LASV (rLASV-WT) in cell culture, but in contrast to rLASV-WT, does not cause disease in inoculated guinea pigs. Guinea pigs immunized with rLASV-GPC/CD were protected against an otherwise lethal exposure to WT LASV. Our results support the testing of this candidate vaccine in nonhuman primate models ofLF.
Collapse
Affiliation(s)
- Yingyun Cai
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - Chengjin Ye
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Benson Cheng
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Aitor Nogales
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| | - Masaharu Iwasaki
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Shuiqing Yu
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - Kurt Cooper
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - David X Liu
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - Randy Hart
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - Ricky Adams
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - Tyler Brady
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - Elena N Postnikova
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - Jonathan Kurtz
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - Marisa St Claire
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - Juan Carlos de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, USA
| |
Collapse
|
34
|
Javelle E, Lesueur A, Pommier de Santi V, de Laval F, Lefebvre T, Holweck G, Durand GA, Leparc-Goffart I, Texier G, Simon F. The challenging management of Rift Valley Fever in humans: literature review of the clinical disease and algorithm proposal. Ann Clin Microbiol Antimicrob 2020; 19:4. [PMID: 31969141 PMCID: PMC6977312 DOI: 10.1186/s12941-020-0346-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 01/12/2020] [Indexed: 01/01/2023] Open
Abstract
Rift Valley Fever (RVF) is an emerging zoonotic arbovirus with a complex cycle of transmission that makes difficult the prediction of its expansion. Recent outbreaks outside Africa have led to rediscover the human disease but it remains poorly known. The wide spectrum of acute and delayed manifestations with potential unfavorable outcome much complicate the management of suspected cases and prediction of morbidity and mortality during an outbreak. We reviewed literature data on bio-clinical characteristics and treatments of RVF human illness. We identified gaps in the field and provided a practical algorithm to assist clinicians in the cases assessment, determination of setting of care and prolonged follow-up.
Collapse
Affiliation(s)
- Emilie Javelle
- Laveran Military Teaching Hospital, CS500413384, Marseille Cedex 13, France. .,IRD, AP-HM, SSA, VITROME, IHU-Méditerranée Infection, Aix Marseille Univ, Marseille, France.
| | - Alexandre Lesueur
- Laveran Military Teaching Hospital, CS500413384, Marseille Cedex 13, France
| | - Vincent Pommier de Santi
- IRD, AP-HM, SSA, VITROME, IHU-Méditerranée Infection, Aix Marseille Univ, Marseille, France.,French Armed Forces Centre for Epidemiology and Public Health (CESPA), Marseille, France
| | - Franck de Laval
- French Armed Forces Centre for Epidemiology and Public Health (CESPA), Marseille, France.,INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de la Santé & Traitement de l'Information Médicale, Aix Marseille Univ, Marseille, France
| | - Thibault Lefebvre
- French Military Health Service, RSMA Medical Unit, Paris, Mayotte, France
| | - Guillaume Holweck
- Laveran Military Teaching Hospital, CS500413384, Marseille Cedex 13, France
| | - Guillaume André Durand
- French Armed Forces Biomedical Research Institute (IRBA)-CNR des arbovirus-IHU Méditerranée Infection, Marseille, France.,IRD 190, Inserm 1207, IHU Méditerranée Infection, AP-HM, UVE, Aix-Marseille Univ, Marseille, France
| | - Isabelle Leparc-Goffart
- French Armed Forces Biomedical Research Institute (IRBA)-CNR des arbovirus-IHU Méditerranée Infection, Marseille, France.,IRD 190, Inserm 1207, IHU Méditerranée Infection, AP-HM, UVE, Aix-Marseille Univ, Marseille, France
| | - Gaëtan Texier
- IRD, AP-HM, SSA, VITROME, IHU-Méditerranée Infection, Aix Marseille Univ, Marseille, France.,French Armed Forces Centre for Epidemiology and Public Health (CESPA), Marseille, France
| | - Fabrice Simon
- Laveran Military Teaching Hospital, CS500413384, Marseille Cedex 13, France.,IRD 190, Inserm 1207, IHU Méditerranée Infection, AP-HM, UVE, Aix-Marseille Univ, Marseille, France
| |
Collapse
|
35
|
Duvignaud A, Jaspard M, Etafo IC, Serra B, Abejegah C, Gabillard D, Doutchi M, Alabi JF, Adedokun MA, Akinpelu AO, Oyegunle OO, Etafo J, Dede AO, Onyechi MN, Ireneh MU, Gbenga-Ayeni O, Fadiminiyi KG, Ehigbor PI, Ouattara E, Levy-Marchal C, Karcher S, N'guessan-Koffi L, Ahyi I, Amani E, Diabaté M, Siloué B, Schaeffer J, Augier A, Ogbaini-Emovon E, Salam AP, Horby P, Ahmed LA, Günther S, Adedosu AN, Anglaret X, Ayodeji OO, Malvy D. Lassa fever clinical course and setting a standard of care for future randomized trials: A protocol for a cohort study of Lassa-infected patients in Nigeria (LASCOPE). Travel Med Infect Dis 2020; 36:101557. [PMID: 31978611 DOI: 10.1016/j.tmaid.2020.101557] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/28/2019] [Accepted: 01/16/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Lassa Fever (LF), is a severe viral disease prevalent in Western Africa. It is classified as a priority disease by the World Health Organization (WHO). Ribavirin is the recommended therapy despite weak evidence of its efficacy. Promising therapeutic agents are becoming available for evaluation in human. Before launching therapeutic trials, we need data on the evolution of the disease under the best possible conditions of care. METHODS We have initiated a prospective study in Nigeria to better understand the clinical course and prognostic factors of LF while implementing high quality standardized care. Inclusion criteria are: suspected or confirmed LF and informed consent. Participants are followed 60 days from admission and receive free of charge standardized supportive care and biological monitoring, as well as intravenous ribavirin for those with confirmed LF. Data are collected using standardized case report forms (CRF). Primary and secondary outcomes are fatality and severe morbidity, with special focus on acute kidney dysfunction and pregnancy complications. Factors associated with outcomes will be investigated. RESULTS The cohort is planned for 3 years. Inclusions started in April 2018 at the Federal Medical Center Owo in Ondo State. A second site will open in Nigeria in 2020 and discussions are underway to open a site in Benin. 150 to 200 new participants are expected per year. CONCLUSIONS This cohort will: provide evidence to standardize LF case management; provide key inputs to design future clinical trials of novel therapeutics; and establish clinical research teams capable of conducting such trials in LF-endemic areas. STUDY REGISTRATION The LASCOPE study was registered on ClinicalTrial.gov (NCT03655561).
Collapse
Affiliation(s)
- Alexandre Duvignaud
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Department of Infectious Diseases and Tropical Medicine, Division of Tropical Medicine and Clinical International Health, CHU de Bordeaux, Hôpital Pellegrin, Place Amélie Raba Léon, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire.
| | - Marie Jaspard
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire; The Alliance for International Medical Action, Route de l'Aéroport, Rue NG 96 BP: 15530, Dakar, Senegal.
| | - Ijeoma Chukwudumebi Etafo
- Lassa Fever Response Team, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Béatrice Serra
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire; The Alliance for International Medical Action, Route de l'Aéroport, Rue NG 96 BP: 15530, Dakar, Senegal.
| | - Chukwuyem Abejegah
- Lassa Fever Response Team, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Delphine Gabillard
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire.
| | - Mahamadou Doutchi
- The Alliance for International Medical Action, Route de l'Aéroport, Rue NG 96 BP: 15530, Dakar, Senegal; Department of Infectious Diseases, Centre Hospitalier National de Zinder, Zinder, Niger.
| | - Josephine Funmilola Alabi
- Lassa Fever Response Team, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Moses Adeniyi Adedokun
- Lassa Fever Response Team, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Adewale Oladayo Akinpelu
- Lassa Fever Response Team, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Oyebimpe Ope Oyegunle
- Lassa Fever Response Team, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Johnson Etafo
- Viral Hemorrhagic Fever Laboratory, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Ayoleyi Omowunmi Dede
- Viral Hemorrhagic Fever Laboratory, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Macdonald Nonso Onyechi
- Viral Hemorrhagic Fever Laboratory, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Moronke Uzuajemeh Ireneh
- Viral Hemorrhagic Fever Laboratory, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Olufunke Gbenga-Ayeni
- Viral Hemorrhagic Fever Laboratory, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Kehinde Gbemisola Fadiminiyi
- Viral Hemorrhagic Fever Laboratory, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Patience Iziegbe Ehigbor
- Viral Hemorrhagic Fever Laboratory, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Eric Ouattara
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire.
| | - Claire Levy-Marchal
- The Alliance for International Medical Action, Route de l'Aéroport, Rue NG 96 BP: 15530, Dakar, Senegal.
| | - Sophie Karcher
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire.
| | - Larissa N'guessan-Koffi
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire.
| | - Irmine Ahyi
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire.
| | - Elvis Amani
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire.
| | - Mamoudou Diabaté
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire.
| | - Bertine Siloué
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire.
| | - Justine Schaeffer
- The Alliance for International Medical Action, Route de l'Aéroport, Rue NG 96 BP: 15530, Dakar, Senegal.
| | - Augustin Augier
- The Alliance for International Medical Action, Route de l'Aéroport, Rue NG 96 BP: 15530, Dakar, Senegal.
| | - Ephraim Ogbaini-Emovon
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, KM 87 Benin Auchi Rd, Irrua, Edo State, Nigeria.
| | - Alex Paddy Salam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, Oxford, OX3 7FZ, United Kingdom.
| | - Peter Horby
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, Oxford, OX3 7FZ, United Kingdom.
| | - Liasu Adeagbo Ahmed
- Department of Family Medicine, Owo Federal Medical Centre, Michael Adekun Ajasin Road, PMB, 1053, Owo, Ondo State, Nigeria.
| | - Stephan Günther
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, Bernhard-Nocht-Straße 74, 20359, Hamburg, Germany.
| | - Akinola Nelson Adedosu
- Viral Hemorrhagic Fever Laboratory, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Xavier Anglaret
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire.
| | - Oladele Oluwafemi Ayodeji
- Lassa Fever Response Team, Infection Control and Research Centre, Federal Medical Centre Owo, Michael Adekun Ajasin Road, PMB 1053, Owo, Ondo State, Nigeria.
| | - Denis Malvy
- Inserm U1219, University of Bordeaux, 146 Rue Léo Saignat, 33076, Bordeaux, France; Department of Infectious Diseases and Tropical Medicine, Division of Tropical Medicine and Clinical International Health, CHU de Bordeaux, Hôpital Pellegrin, Place Amélie Raba Léon, 33076, Bordeaux, France; Programme PAC-CI/ANRS Research Site, CHU de Treichville, 18 BP 1954 Abidjan 18, Abidjan, Côte d'Ivoire.
| |
Collapse
|
36
|
Abstract
Increases in tick-borne disease prevalence and transmission are important public health issues. Efforts to control these emerging diseases are frustrated by the struggle to control tick populations and to detect and treat infections caused by the pathogens that they transmit. This review covers tick-borne infectious diseases of nonrickettsial bacterial, parasitic, and viral origins. While tick surveillance and tracking inform our understanding of the importance of the spread and ecology of ticks and help identify areas of risk for disease transmission, the vectors are not the focus of this document. Here, we emphasize the most significant pathogens that infect humans as well as the epidemiology, clinical features, diagnosis, and treatment of diseases that they cause. Although detection via molecular or immunological methods has improved, tick-borne diseases continue to remain underdiagnosed, making the scope of the problem difficult to assess. Our current understanding of the incidence of tick-borne diseases is discussed in this review. An awareness of the diseases that can be transmitted by ticks in specific locations is key to detection and selection of appropriate treatment. As tick-transmitted pathogens are discovered and emerge in new geographic regions, our ability to detect, describe, and understand the growing public health threat must also grow to meet the challenge.
Collapse
|
37
|
Cubitt B, Ortiz-Riano E, Cheng BY, Kim YJ, Yeh CD, Chen CZ, Southall NOE, Zheng W, Martinez-Sobrido L, de la Torre JC. A cell-based, infectious-free, platform to identify inhibitors of lassa virus ribonucleoprotein (vRNP) activity. Antiviral Res 2020; 173:104667. [PMID: 31786250 PMCID: PMC6954049 DOI: 10.1016/j.antiviral.2019.104667] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022]
Abstract
The mammarenavirus Lassa (LASV) is highly prevalent in West Africa where it infects several hundred thousand individuals annually resulting in a high number of Lassa fever (LF) cases, a febrile disease associated with high morbidity and significant mortality. Mounting evidence indicates that the worldwide-distributed prototypic mammarenavirus lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogen of clinical significance. There are not Food and Drug Administration (FDA) licensed vaccines and current anti-mammarenavirus therapy is limited to an off-label use of ribavirin that is only partially effective and can cause significant side effects. Therefore, there is an unmet need for novel antiviral drugs to combat LASV. This task would be facilitated by the implementation of high throughput screens (HTS) to identify inhibitors of the activity of the virus ribonucleoprotein (vRNP) responsible for directing virus RNA genome replication and gene transcription. The use of live LASV for this purpose is jeopardized by the requirement of biosafety level 4 (BSL4) containment. We have developed a virus-free cell platform, where expression levels of reporter genes serve as accurate surrogates of vRNP activity, to develop cell-based assays compatible with HTS to identify inhibitors of LASV and LCMV mammarenavirus vRNP activities.
Collapse
Affiliation(s)
- Beatrice Cubitt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Emilio Ortiz-Riano
- Department of Microbiology and Immunology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Benson Yh Cheng
- Department of Microbiology and Immunology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Yu-Jin Kim
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Charles D Yeh
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Catherine Z Chen
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - N O E Southall
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Luis Martinez-Sobrido
- Department of Microbiology and Immunology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Juan C de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
38
|
Okogbenin S, Okoeguale J, Akpede G, Colubri A, Barnes KG, Mehta S, Eifediyi R, Okogbo F, Eigbefoh J, Momoh M, Rafiu M, Adomeh D, Odia I, Aire C, Atafo R, Okonofua M, Pahlman M, Becker-Ziaja B, Asogun D, Okokhere P, Happi C, Günther S, Sabeti PC, Ogbaini-Emovon E. Retrospective Cohort Study of Lassa Fever in Pregnancy, Southern Nigeria. Emerg Infect Dis 2019; 25. [PMID: 31310586 PMCID: PMC6649346 DOI: 10.3201/eid2508.181299] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
39
|
Babalola SO, Babatunde JA, Remilekun OM, Amaobichukwu AR, Abiodun AM, Jide I, Adeshina ASI, Chikwe I, Aremu OS. Lassa virus RNA detection from suspected cases in Nigeria, 2011-2017. Pan Afr Med J 2019; 34:76. [PMID: 31819792 PMCID: PMC6884721 DOI: 10.11604/pamj.2019.34.76.16425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/02/2019] [Indexed: 11/11/2022] Open
Abstract
Introduction The diagnosis of Lassa fever is crucial to confirm cases, as well as to control/prevent nosocomial and community-based transmission and initiation of treatment, which is still limited in the country. Thus, we aimed at providing some information on the laboratory detection of Lassa from suspected cases in Nigeria. Methods This was a retrospective study of seasonal Lassa fever outbreaks data from 1,263 samples analyzed using Reverse Transcription-Polymerase Chain Reaction (RT-PCR) at the Virology Research Laboratory, College of Medicine, University of Lagos/Lagos University Teaching Hospital between year 2011 and 2017. Data were analyzed using the 21st edition of SPSS statistical software (2015). Results The RT-PCR test confirmed the presence of Lassa in 112 (8.9%) comprising 61 (54.4%) males, 48 (42.9%) females and 3 (2.7%) individuals without gender information. Those aged between 18 and 49 years were mostly affected. There was a decline in the detection of Lassa from 4.7% in 2011/2012 to less than 1% by the 2014/2015. However, during the 2015/2016 and 2016/2017 seasons the detection rates increased to 10.4% and 15.1% respectively. The Northern region of Nigeria reported high confirmed cases of Lassa. The South Western region also witnessed an increased Lassa fever positivity rate of 13.4% of which Lagos and Ogun states being the focal state of Lassa activity in the region. Conclusion These established the need for heightening the continued surveillance for Lassa as well as the establishment of other testing facilities within these endemic regions for prompt diagnosis of Lassa fever.
Collapse
Affiliation(s)
- Salu Olumuyiwa Babalola
- Department of Medical Microbiology and Parasitology, College of Medicine, University of Lagos (CMUL), PM.B. 12003, Lagos, Nigeria.,Centre for Human and Zoonotic Virology (CHAZVY), Central Research Laboratory, College of Medicine, University of Lagos (CMUL)/Lagos University Teaching Hospital (LUTH), Lagos, Nigeria
| | - James Ayorinde Babatunde
- Centre for Human and Zoonotic Virology (CHAZVY), Central Research Laboratory, College of Medicine, University of Lagos (CMUL)/Lagos University Teaching Hospital (LUTH), Lagos, Nigeria.,Department of Biochemistry, College of Medicine, University of Lagos, PM.B. 12003, Lagos, Nigeria
| | - Orenolu Mercy Remilekun
- Centre for Human and Zoonotic Virology (CHAZVY), Central Research Laboratory, College of Medicine, University of Lagos (CMUL)/Lagos University Teaching Hospital (LUTH), Lagos, Nigeria
| | - Anyanwu Roosevelt Amaobichukwu
- Centre for Human and Zoonotic Virology (CHAZVY), Central Research Laboratory, College of Medicine, University of Lagos (CMUL)/Lagos University Teaching Hospital (LUTH), Lagos, Nigeria
| | - Abdullah Mariam Abiodun
- Centre for Human and Zoonotic Virology (CHAZVY), Central Research Laboratory, College of Medicine, University of Lagos (CMUL)/Lagos University Teaching Hospital (LUTH), Lagos, Nigeria
| | - Idris Jide
- Honourable Commissioner for Health, Lagos State Ministry of Health, Alausa, Ikeja, Lagos, Nigeria
| | - Abdus-Salam Ismail Adeshina
- Epidemiology Unit, Directorate of Disease Control, Lagos State Ministry of Health, Alausa, Ikeja, Lagos, Nigeria
| | - Ihekweazu Chikwe
- Nigerian Centre for Disease Control (NCDC), Plot 801, Ebitu Ukiwe Street, Jabi, Abuja, Nigeria
| | - Omilabu Sunday Aremu
- Department of Medical Microbiology and Parasitology, College of Medicine, University of Lagos (CMUL), PM.B. 12003, Lagos, Nigeria.,Centre for Human and Zoonotic Virology (CHAZVY), Central Research Laboratory, College of Medicine, University of Lagos (CMUL)/Lagos University Teaching Hospital (LUTH), Lagos, Nigeria
| |
Collapse
|
40
|
Mejer N, Galli A, Ramirez S, Fahnøe U, Benfield T, Bukh J. Ribavirin inhibition of cell-culture infectious hepatitis C genotype 1-3 viruses is strain-dependent. Virology 2019; 540:132-140. [PMID: 31778898 DOI: 10.1016/j.virol.2019.09.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/27/2019] [Accepted: 09/27/2019] [Indexed: 12/20/2022]
Abstract
Ribavirin remains relevant for successful treatment of chronic hepatitis C virus (HCV) infections in low-income settings, as well as for therapy of difficult-to-treat HCV patients. We studied the effect of ribavirin against cell-culture adapted HCV of genotypes 1, 2 and 3, representing ~80% of global infections. TNcc(1a) was the most sensitive to ribavirin, while J6/JFH1(2a) was the most resistant. EC50s ranged from 21 μM (95%CI: 20-22 μM) to 189 μM (95%CI: 173-207 μM). Substitutions at position 415 of NS5B resulted in little or no change to ribavirin sensitivity (0.7-0.9 fold) but conferred moderate drug resistance during extended treatment of genotype 1 (1.8-fold). NS5A and NS5B sequences could alter ribavirin sensitivity 2-4-fold, although their contribution was not simply additive. Finally, we detected limited accumulation of mutations associated with ribavirin treatment. Our findings show that the antiviral effect of ribavirin on HCV is strain-dependent and is influenced by the specific sequence of multiple HCV nonstructural proteins.
Collapse
Affiliation(s)
- Niels Mejer
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Hvidovre Hospital, Hvidovre, Denmark
| | - Andrea Galli
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Hvidovre Hospital, Hvidovre, Denmark
| | - Santseharay Ramirez
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Hvidovre Hospital, Hvidovre, Denmark
| | - Ulrik Fahnøe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Hvidovre Hospital, Hvidovre, Denmark
| | - Thomas Benfield
- Department of Infectious Diseases, Hvidovre Hospital, Hvidovre, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Hvidovre Hospital, Hvidovre, Denmark.
| |
Collapse
|
41
|
de la Torre JC. Extending the Antiviral Value of Favipiravir. J Infect Dis 2019; 218:509-511. [PMID: 29762707 DOI: 10.1093/infdis/jiy153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/11/2018] [Indexed: 02/01/2023] Open
Affiliation(s)
- Juan C de la Torre
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California
| |
Collapse
|
42
|
Kulkarni PA, Chew D, Youssef-Bessler M, Hamdi HA, Montoya LA, Cervantes KB, Mazur NL, Lucas D, Wells JW, Cennimo D, Sutherland A, Di Domenico LM, Miller LP, Pierre-Louis F, Rokosz G, Nazir A, de Perio MA, Lowe L, Manning C, Mead KR, Christensen BE, Albariño CG, Ströher U, Glover M, Lifshitz EI, Tan CG, Rollin PE, Semple S. Case Report: Imported Case of Lassa Fever - New Jersey, May 2015. Am J Trop Med Hyg 2019; 99:1062-1065. [PMID: 30062993 DOI: 10.4269/ajtmh.17-0316] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
We report a fatal case of Lassa fever diagnosed in the United States in a Liberian traveler. We describe infection control protocols and public health response. One contact at high risk became symptomatic, but her samples tested negative for Lassa virus; no secondary cases occurred among health care, family, and community contacts.
Collapse
Affiliation(s)
- Prathit A Kulkarni
- Centers for Disease Control and Prevention, Atlanta, Georgia.,Communicable Disease Service, New Jersey Department of Health, Trenton, New Jersey
| | - Debra Chew
- University Hospital, Newark, New Jersey.,Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | | | - Hanaa A Hamdi
- Newark Department of Health and Community Wellness, Newark, New Jersey
| | - Lindsay A Montoya
- Communicable Disease Service, New Jersey Department of Health, Trenton, New Jersey
| | - Kimberly B Cervantes
- Communicable Disease Service, New Jersey Department of Health, Trenton, New Jersey
| | - Nicole L Mazur
- Communicable Disease Service, New Jersey Department of Health, Trenton, New Jersey
| | - Diana Lucas
- Communicable Disease Service, New Jersey Department of Health, Trenton, New Jersey
| | - Julia W Wells
- Communicable Disease Service, New Jersey Department of Health, Trenton, New Jersey
| | - David Cennimo
- University Hospital, Newark, New Jersey.,Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Anne Sutherland
- University Hospital, Newark, New Jersey.,Department of Medicine, Rutgers New Jersey Medical School, Newark, New Jersey
| | | | | | | | | | - Atif Nazir
- Newark Department of Health and Community Wellness, Newark, New Jersey
| | | | - Luis Lowe
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Craig Manning
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Kenneth R Mead
- Centers for Disease Control and Prevention, Cincinnati, Ohio
| | | | | | - Ute Ströher
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Maleeka Glover
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Edward I Lifshitz
- Communicable Disease Service, New Jersey Department of Health, Trenton, New Jersey
| | - Christina G Tan
- Communicable Disease Service, New Jersey Department of Health, Trenton, New Jersey
| | - Pierre E Rollin
- Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Shereen Semple
- Communicable Disease Service, New Jersey Department of Health, Trenton, New Jersey
| |
Collapse
|
43
|
Garnett LE, Strong JE. Lassa fever: With 50 years of study, hundreds of thousands of patients and an extremely high disease burden, what have we learned? Curr Opin Virol 2019; 37:123-131. [PMID: 31479990 DOI: 10.1016/j.coviro.2019.07.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 07/23/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Lauren E Garnett
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Canada; Departments of Medical Microbiology and Infectious Diseases, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - James E Strong
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Canada; Departments of Medical Microbiology and Infectious Diseases, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; Departments of Paediatrics and Child Health, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
44
|
Tang-Huau TL, Feldmann H, Rosenke K. Animal models for Lassa virus infection. Curr Opin Virol 2019; 37:112-117. [PMID: 31442921 DOI: 10.1016/j.coviro.2019.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/20/2022]
Abstract
In humans, Lassa virus infection can result in disease with hemorrhagic manifestations and high fatality rates. There are no approved treatments or vaccines available and the inherent danger of studying Lassa virus means it can only be studied in high containment labs (BSL4). Under these conditions, mouse models are becoming an important instrument in the study of Lassa virus infection, disease and host responses. While guinea pigs and non-human primates are the critical components in assessing treatments and vaccines and have recently been used with great affect in this capacity.
Collapse
Affiliation(s)
- Tsing-Lee Tang-Huau
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - H Feldmann
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA; Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - K Rosenke
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA.
| |
Collapse
|
45
|
Assessment of antiviral therapeutics in animal models of Lassa fever. Curr Opin Virol 2019; 37:84-90. [PMID: 31357140 DOI: 10.1016/j.coviro.2019.06.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/24/2019] [Indexed: 12/28/2022]
Abstract
Lassa virus (LASV) is an emerging zoonotic virus endemic in West Africa that can cause severe haemorrhagic Lassa fever (LF) in humans. LF recently gained international attention as a prominent infectious disease, leading to increasingly severe outbreaks in Nigeria over the past three years. Morbidity and mortality associated with LF disease in Nigeria continue to rise with 106 deaths reported in 2016, 143 in 2017 and 562 in 2018. Despite the significant health impact LF imposes on West Africa there are currently no FDA-approved therapeutics or vaccines available for treatment and prevention. This review focuses on the assessment and current state of LF antiviral therapeutics in animal models and their potential role in reducing disease burden throughout West Africa.
Collapse
|
46
|
Wendt L, Bostedt L, Hoenen T, Groseth A. High-throughput screening for negative-stranded hemorrhagic fever viruses using reverse genetics. Antiviral Res 2019; 170:104569. [PMID: 31356830 DOI: 10.1016/j.antiviral.2019.104569] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/28/2019] [Accepted: 07/25/2019] [Indexed: 02/06/2023]
Abstract
Viral hemorrhagic fevers (VHFs) cause thousands of fatalities every year, but the treatment options for their management remain very limited. In particular, the development of therapeutic interventions is restricted by the lack of commercial viability of drugs targeting individual VHF agents. This makes approaches like drug repurposing and/or the identification of broad range therapies (i.e. those directed at host responses or common proviral factors) highly attractive. However, the identification of candidates for such antiviral repurposing or of host factors/pathways important for the virus life cycle is reliant on high-throughput screening (HTS). Recently, such screening work has been increasingly facilitated by the availability of reverse genetics-based approaches, including tools such as full-length clone (FLC) systems to generate reporter-expressing viruses or various life cycle modelling (LCM) systems, many of which have been developed and/or greatly improved during the last years. In particular, since LCM systems are capable of modelling specific steps in the life cycle, they are a valuable tool for both targeted screening (i.e. for inhibitors of a specific pathway) and mechanism of action studies. This review seeks to summarize the currently available reverse genetics systems for negative-sense VHF causing viruses (i.e. arenaviruses, bunyaviruses and filoviruses), and to highlight the recent advancements made in applying these systems for HTS to identify either antivirals or new virus-host interactions that might hold promise for the development of future treatments for the infections caused by these deadly but neglected virus groups.
Collapse
Affiliation(s)
- Lisa Wendt
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald, Insel Riems, Germany
| | - Linus Bostedt
- Junior Research Group - Arenavirus Biology, Friedrich-Loeffler-Institut, Greifswald, Insel Riems, Germany
| | - Thomas Hoenen
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald, Insel Riems, Germany.
| | - Allison Groseth
- Junior Research Group - Arenavirus Biology, Friedrich-Loeffler-Institut, Greifswald, Insel Riems, Germany.
| |
Collapse
|
47
|
Eberhardt KA, Mischlinger J, Jordan S, Groger M, Günther S, Ramharter M. Ribavirin for the treatment of Lassa fever: A systematic review and meta-analysis. Int J Infect Dis 2019; 87:15-20. [PMID: 31357056 DOI: 10.1016/j.ijid.2019.07.015] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/14/2019] [Accepted: 07/22/2019] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVES Lassa fever (LF) causes annual outbreaks in endemic regions with high mortality of symptomatic patients. Ribavirin is recommended as standard treatment for LF in national and international guidelines but the evidence base for this recommendation has been questioned recently. METHODS We conducted a systematic review and included 6 studies providing efficacy data of ribavirin treatment for LF (PROSPERO protocol CRD42018103994). RESULTS Besides retrospective case series, the evidence mostly relies on a single prospective clinical trial with critical risk of bias. In this trial, LF associated mortality is reduced for patients with elevated aspartate aminotransferase (AST) when treated with ribavirin (OR 0.41, 95% CI 0.23-0.73), while mortality is higher for patients without elevated AST (OR 2.37, 95% CI 1.07-5.25). CONCLUSIONS Based on the available data, current treatment guidelines may therefore put patients with mild LF at increased risk of death. The role of ribavirin in the treatment of LF requires urgent reassessment.
Collapse
Affiliation(s)
- Kirsten Alexandra Eberhardt
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Mischlinger
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Jordan
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mirjam Groger
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Günther
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Michael Ramharter
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine and I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
48
|
Takah NF, Brangel P, Shrestha P, Peeling R. Sensitivity and specificity of diagnostic tests for Lassa fever: a systematic review. BMC Infect Dis 2019; 19:647. [PMID: 31324229 PMCID: PMC6642489 DOI: 10.1186/s12879-019-4242-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/30/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Lassa fever virus has been enlisted as a priority pathogen of epidemic potential by the World Health organization Research and Development (WHO R & D) Blueprint. Diagnostics play a crucial role in epidemic preparedness. This systematic review was conducted to determine the sensitivity and specificity of Lassa fever diagnostic tests for humans. METHODS We searched OVID Medline, OVID Embase, Scopus and Web of Science for laboratory based and field studies that reported the performance of diagnostic tests for Lassa fever in humans from 1 January 1990 to 25 January 2019. Two reviewers independently screened all the studies and included only studies that involved the evaluation of a Lassa fever diagnostic test and provided data on the sensitivity and specificity. The quality of the studies was assessed using the QUADAS-2 criteria. Data on the study location, study design, type of sample, index test, reference tests and diagnostic performance were extracted from the studies. RESULTS Out of a total of 1947 records identified, 1245 non-duplicate citations were obtained. Twenty-six (26) full-text articles examined which identified 08 studies meeting pre-defined criteria. Only one study was a field evaluation study. The sensitivity and specificity of the point of care (RDT) against the Nikisins qPCR were 91.2%(95% CI:75.2-97.7) and 86%(95% CI: 71.4-94.2) at temperatures 18-30 °C, while the sensitivity and specificity of the single IgM ELISA assay against standard RT-PCR were 31.1%(95%CI: 25.6-37) and 95.7%(95%CI:92.8-97.7). The sensitivity of the combined ELISA Antigen/IgM assay(against virus isolation), the recombinant IgM/IgG ELISA(against standard RT-PCR), and the IgM/IgG immunoblot(against IFA) were 88%(95%CI:77-95), 25.9%(95%CI:20.8-31.6), and 90.7%(95%CI:84.13-97.27) respectively. The specificity of the combined ELISA Antigen/IgM assay(against virus isolation), the recombinant IgM/IgG ELISA(against standard RT-PCR), and the IgM/IgG immunoblot(against IFA) were 90%(95%CI:88-91), 100%(95%CI:98.2-100), and 96.3%(95%CI:92.2-100) respectively. CONCLUSION Lassa fever has assays for antigenaemia, IgM, IgG and PCR detection. The RDT reportedly performed well but more data are needed from other countries and at temperatures above 30 °C. Most combined immunoassays perform better than the single IgM. Multiplex and pan-Lassa assays are needed. More well conducted field studies are needed. TRIAL REGISTRATION Prospero registration number: CRD42018091585 .
Collapse
Affiliation(s)
- Noah Fongwen Takah
- International Diagnostics Centre Africa, Addis Ababa, Ethiopia. .,International Diagnostics Centre, Clinical Research Department, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.
| | - Polina Brangel
- London Centre for Nanotechnology, University College London, London, UK
| | - Priyanka Shrestha
- International Diagnostics Centre, Clinical Research Department, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Rosanna Peeling
- International Diagnostics Centre, Clinical Research Department, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| |
Collapse
|
49
|
Ergönül Ö, Keske Ş, Çeldir MG, Kara İA, Pshenichnaya N, Abuova G, Blumberg L, Gönen M. Systematic Review and Meta-analysis of Postexposure Prophylaxis for Crimean-Congo Hemorrhagic Fever Virus among Healthcare Workers. Emerg Infect Dis 2019; 24:1642-1648. [PMID: 30124196 PMCID: PMC6106438 DOI: 10.3201/eid2409.171709] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We performed a systematic review and meta-analysis on the effectiveness of ribavirin use for the prevention of infection and death of healthcare workers exposed to patients with Crimean-Congo hemorrhagic fever virus (CCHFV) infection. Splashes with blood or bodily fluids (odds ratio [OR] 4.2), being a nurse or physician (OR 2.1), and treating patients who died from CCHFV infection (OR 3.8) were associated with healthcare workers acquiring CCHFV infection; 7% of the workers who received postexposure prophylaxis (PEP) with ribavirin and 89% of those who did not became infected. PEP with ribavirin reduced the odds of infection (OR 0.01, 95% CI 0–0.03), and ribavirin use <48 hours after symptom onset reduced the odds of death (OR 0.03, 95% CI 0–0.58). The odds of death increased 2.4-fold every day without ribavirin treatment. Ribavirin should be recommended as PEP and early treatment for workers at medium-to-high risk for CCHFV infection.
Collapse
|
50
|
Arbidol and Other Low-Molecular-Weight Drugs That Inhibit Lassa and Ebola Viruses. J Virol 2019; 93:JVI.02185-18. [PMID: 30700611 DOI: 10.1128/jvi.02185-18] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 01/22/2019] [Indexed: 02/08/2023] Open
Abstract
Antiviral therapies that impede virus entry are attractive because they act on the first phase of the infectious cycle. Drugs that target pathways common to multiple viruses are particularly desirable when laboratory-based viral identification may be challenging, e.g., in an outbreak setting. We are interested in identifying drugs that block both Ebola virus (EBOV) and Lassa virus (LASV), two unrelated but highly pathogenic hemorrhagic fever viruses that have caused outbreaks in similar regions in Africa and share features of virus entry: use of cell surface attachment factors, macropinocytosis, endosomal receptors, and low pH to trigger fusion in late endosomes. Toward this goal, we directly compared the potency of eight drugs known to block EBOV entry with their potency as inhibitors of LASV entry. Five drugs (amodiaquine, apilimod, arbidol, niclosamide, and zoniporide) showed roughly equivalent degrees of inhibition of LASV and EBOV glycoprotein (GP)-bearing pseudoviruses; three (clomiphene, sertraline, and toremifene) were more potent against EBOV. We then focused on arbidol, which is licensed abroad as an anti-influenza drug and exhibits activity against a diverse array of clinically relevant viruses. We found that arbidol inhibits infection by authentic LASV, inhibits LASV GP-mediated cell-cell fusion and virus-cell fusion, and, reminiscent of its activity on influenza virus hemagglutinin, stabilizes LASV GP to low-pH exposure. Our findings suggest that arbidol inhibits LASV fusion, which may partly involve blocking conformational changes in LASV GP. We discuss our findings in terms of the potential to develop a drug cocktail that could inhibit both LASV and EBOV.IMPORTANCE Lassa and Ebola viruses continue to cause severe outbreaks in humans, yet there are only limited therapeutic options to treat the deadly hemorrhagic fever diseases they cause. Because of overlapping geographic occurrences and similarities in mode of entry into cells, we seek a practical drug or drug cocktail that could be used to treat infections by both viruses. Toward this goal, we directly compared eight drugs, approved or in clinical testing, for the ability to block entry mediated by the glycoproteins of both viruses. We identified five drugs with approximately equal potencies against both. Among these, we investigated the modes of action of arbidol, a drug licensed abroad to treat influenza infections. We found, as shown for influenza virus, that arbidol blocks fusion mediated by the Lassa virus glycoprotein. Our findings encourage the development of a combination of approved drugs to treat both Lassa and Ebola virus diseases.
Collapse
|