1
|
Emperador DM, Kelly-Cirino C, Bausch DG, Eckerle I. Systematic review and meta-analysis of antigen rapid diagnostic tests to detect Zaire ebolavirus. Diagn Microbiol Infect Dis 2024; 111:116568. [PMID: 39467415 DOI: 10.1016/j.diagmicrobio.2024.116568] [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: 07/02/2024] [Revised: 09/30/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024]
Abstract
We conducted a systematic review and meta-analysis of studies and reports comparing the performance of antigen rapid diagnostic tests (Ag RDT) for diagnosing Ebola disease (EVD). We searched PubMed, EMBASE, and Web of Science for diagnostic studies published between 1976 and 2023, evaluating them with QUADAS-2. Using a bivariate random-effects model, we estimated the pooled sensitivity and specificity of Ag RDTs. Of 64 eligible full studies and reports, 16 met the inclusion criteria. Pooled sensitivity and specificity were 82.1% (95%CI: 75.2 - 88.0) and 97.0% (95%CI: 95.1-98.2), respectively. We conducted subgroup analysis on 4 Ag RDTs, 3 RT-PCR tests, and 4 sample types, showing varied performance. The high specificity and positive predictive value of Ag RDTs support their use to "rule-in" patients with EVD. However, high-sensitivity RDTs suitable for field settings and capable of detecting multiple ebolavirus species are needed.
Collapse
Affiliation(s)
- Devy M Emperador
- Institute of Global Health, Université de Genève, Geneva, Switzerland; FIND, Geneva, Switzerland.
| | | | - Daniel G Bausch
- FIND, Geneva, Switzerland; London School of Tropical Medicine and Hygiene, London, United Kingdom
| | - Isabella Eckerle
- Center for Emerging Viral Diseases, Hôpitaux Universitaires de Genève, Geneva, Switzerland; Department of Medicine, Université de Genève, Geneva, Switzerland
| |
Collapse
|
2
|
Schäfer A, Marzi A, Furuyama W, Catanzaro NJ, Nguyen C, Haddock E, Feldmann F, Meade-White K, Thomas T, Hubbard ML, Gully KL, Leist SR, Hock P, Bell TA, De la Cruz GE, Midkiff BR, Martinez DR, Shaw GD, Miller DR, Vernon MJ, Graham RL, Cowley DO, Montgomery SA, Schughart K, de Villena FPM, Wilkerson GK, Ferris MT, Feldmann H, Baric RS. Mapping of susceptibility loci for Ebola virus pathogenesis in mice. Cell Rep 2024; 43:114127. [PMID: 38652660 PMCID: PMC11348656 DOI: 10.1016/j.celrep.2024.114127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 03/11/2024] [Accepted: 04/03/2024] [Indexed: 04/25/2024] Open
Abstract
Ebola virus (EBOV), a major global health concern, causes severe, often fatal EBOV disease (EVD) in humans. Host genetic variation plays a critical role, yet the identity of host susceptibility loci in mammals remains unknown. Using genetic reference populations, we generate an F2 mapping cohort to identify host susceptibility loci that regulate EVD. While disease-resistant mice display minimal pathogenesis, susceptible mice display severe liver pathology consistent with EVD-like disease and transcriptional signatures associated with inflammatory and liver metabolic processes. A significant quantitative trait locus (QTL) for virus RNA load in blood is identified in chromosome (chr)8, and a severe clinical disease and mortality QTL is mapped to chr7, which includes the Trim5 locus. Using knockout mice, we validate the Trim5 locus as one potential driver of liver failure and mortality after infection. The identification of susceptibility loci provides insight into molecular genetic mechanisms regulating EVD progression and severity, potentially informing therapeutics and vaccination strategies.
Collapse
Affiliation(s)
- Alexandra Schäfer
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA.
| | - Wakako Furuyama
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Nicholas J Catanzaro
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Cameron Nguyen
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Elaine Haddock
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Tina Thomas
- Rocky Mountain Veterinary Branch, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Miranda L Hubbard
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kendra L Gully
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Pablo Hock
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Timothy A Bell
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Gabriela E De la Cruz
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Bentley R Midkiff
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - David R Martinez
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ginger D Shaw
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Darla R Miller
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michael J Vernon
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rachel L Graham
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Dale O Cowley
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Animal Models Core Facility, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Stephanie A Montgomery
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Klaus Schughart
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Institute of Virology, University of Muenster, 48149 Muenster, Germany
| | - Fernando Pardo Manuel de Villena
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Gregory K Wilkerson
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Martin T Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, NIAID, NIH, Hamilton, MT 59840, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
3
|
Bodmer BS, Breithaupt A, Heung M, Brunetti JE, Henkel C, Müller-Guhl J, Rodríguez E, Wendt L, Winter SL, Vallbracht M, Müller A, Römer S, Chlanda P, Muñoz-Fontela C, Hoenen T, Escudero-Pérez B. In vivo characterization of the novel ebolavirus Bombali virus suggests a low pathogenic potential for humans. Emerg Microbes Infect 2023; 12:2164216. [PMID: 36580440 PMCID: PMC9858441 DOI: 10.1080/22221751.2022.2164216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ebolaviruses cause outbreaks of haemorrhagic fever in Central and West Africa. Some members of this genus such as Ebola virus (EBOV) are highly pathogenic, with case fatality rates of up to 90%, whereas others such as Reston virus (RESTV) are apathogenic for humans. Bombali virus (BOMV) is a novel ebolavirus for which complete genome sequences were recently found in free-tailed bats, although no infectious virus could be isolated. Its pathogenic potential for humans is unknown. To address this question, we first determined whether proteins encoded by the available BOMV sequence found in Chaerephon pumilus were functional in in vitro assays. The correction of an apparent sequencing error in the glycoprotein based on these data then allowed us to generate infectious BOMV using reverse genetics and characterize its infection of human cells. Furthermore, we used HLA-A2-transgenic, NOD-scid-IL-2γ receptor-knockout (NSG-A2) mice reconstituted with human haematopoiesis as a model to evaluate the pathogenicity of BOMV in vivo in a human-like immune environment. These data demonstrate that not only does BOMV show a slower growth rate than EBOV in vitro, but it also shows low pathogenicity in humanized mice, comparable to previous studies using RESTV. Taken together, these findings suggest a low pathogenic potential of BOMV for humans.
Collapse
Affiliation(s)
- B. S. Bodmer
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - A. Breithaupt
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - M. Heung
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - J. E. Brunetti
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - C. Henkel
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - J. Müller-Guhl
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany,Leibniz Institute of Virology, Hamburg, Germany
| | - E. Rodríguez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany,German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel, Braunschweig, Germany
| | - L. Wendt
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - S. L. Winter
- Schaller Research Groups, Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - M. Vallbracht
- Schaller Research Groups, Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - A. Müller
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - S. Römer
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany
| | - P. Chlanda
- Schaller Research Groups, Department of Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - C. Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany,German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel, Braunschweig, Germany
| | - T. Hoenen
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald – Insel Riems, Germany, T. Hoenen Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Südufer 10, Greifswald – Insel Riems, 17493Germany
| | - B. Escudero-Pérez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany,German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel, Braunschweig, Germany
| |
Collapse
|
4
|
Bao M, Waitkus J, Liu L, Chang Y, Xu Z, Qin P, Chen J, Du K. Micro- and nanosystems for the detection of hemorrhagic fever viruses. LAB ON A CHIP 2023; 23:4173-4200. [PMID: 37675935 DOI: 10.1039/d3lc00482a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Hemorrhagic fever viruses (HFVs) are virulent pathogens that can cause severe and often fatal illnesses in humans. Timely and accurate detection of HFVs is critical for effective disease management and prevention. In recent years, micro- and nano-technologies have emerged as promising approaches for the detection of HFVs. This paper provides an overview of the current state-of-the-art systems for micro- and nano-scale approaches to detect HFVs. It covers various aspects of these technologies, including the principles behind their sensing assays, as well as the different types of diagnostic strategies that have been developed. This paper also explores future possibilities of employing micro- and nano-systems for the development of HFV diagnostic tools that meet the practical demands of clinical settings.
Collapse
Affiliation(s)
- Mengdi Bao
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA, USA.
| | - Jacob Waitkus
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA, USA.
| | - Li Liu
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA, USA.
| | - Yu Chang
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA, USA.
| | - Zhiheng Xu
- Department of Industrial Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Peiwu Qin
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Juhong Chen
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Ke Du
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA, USA.
| |
Collapse
|
5
|
Ferraz de Arruda G, Petri G, Rodriguez PM, Moreno Y. Multistability, intermittency, and hybrid transitions in social contagion models on hypergraphs. Nat Commun 2023; 14:1375. [PMID: 36914645 PMCID: PMC10011415 DOI: 10.1038/s41467-023-37118-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
Although ubiquitous, interactions in groups of individuals are not yet thoroughly studied. Frequently, single groups are modeled as critical-mass dynamics, which is a widespread concept used not only by academics but also by politicians and the media. However, less explored questions are how a collection of groups will behave and how their intersection might change the dynamics. Here, we formulate this process as binary-state dynamics on hypergraphs. We showed that our model has a rich behavior beyond discontinuous transitions. Notably, we have multistability and intermittency. We demonstrated that this phenomenology could be associated with community structures, where we might have multistability or intermittency by controlling the number or size of bridges between communities. Furthermore, we provided evidence that the observed transitions are hybrid. Our findings open new paths for research, ranging from physics, on the formal calculation of quantities of interest, to social sciences, where new experiments can be designed.
Collapse
Affiliation(s)
| | | | | | - Yamir Moreno
- CENTAI Institute, Turin, Italy
- Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, 50018, Zaragoza, Spain
- Department of Theoretical Physics, University of Zaragoza, 50018, Zaragoza, Spain
| |
Collapse
|
6
|
John Higham D, de Kergorlay HL. Disease extinction for susceptible-infected-susceptible models on dynamic graphs and hypergraphs. CHAOS (WOODBURY, N.Y.) 2022; 32:083131. [PMID: 36049952 DOI: 10.1063/5.0093776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/06/2022] [Indexed: 06/15/2023]
Abstract
We consider stochastic, individual-level susceptible-infected-susceptible models for the spread of disease, opinion, or information on dynamic graphs and hypergraphs. We set up "snapshot" models where the interactions at any time are independently and identically sampled from an underlying distribution that represents a typical scenario. In the hypergraph case, this corresponds to a new Gilbert-style random hypergraph model. After justifying this modeling regime, we present useful mean field approximations. With an emphasis on the derivation of spectral conditions that determine long-time extinction, we give computational simulations and accompanying theoretical analysis for the exact models and their mean field approximations.
Collapse
Affiliation(s)
- Desmond John Higham
- School of Mathematics, University of Edinburgh, Edinburgh EH9 3FD, United Kingdom
| | | |
Collapse
|
7
|
Fausther-Bovendo H, Qiu X, Babuadze GG, Azizi H, Pedersen J, Wong G, Kobinger GP. Transient Liver Damage and Hemolysis Are Associated With an Inhibition of Ebola Virus Glycoprotein-Specific Antibody Response and Lymphopenia. J Infect Dis 2022; 225:1852-1855. [PMID: 34791300 PMCID: PMC9113424 DOI: 10.1093/infdis/jiab552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/27/2021] [Indexed: 11/14/2022] Open
Abstract
Numerous studies have demonstrated the importance of the adaptive immunity for survival following Ebola virus (EBOV) infection. To evaluate the contribution of tissue damage to EBOV-induced immune suppression, acute liver damage or hemolysis, 2 symptoms associated with lethal EBOV infection, were chemically induced in vaccinated mice. Results show that either liver damage or hemolysis was sufficient to inhibit the host humoral response against EBOV glycoprotein and to drastically reduce the level of circulating T cells. This study thus provides a possible mechanism for the limited specific antibody production and lymphopenia in individuals with lethal hemorrhagic fever infections.
Collapse
Affiliation(s)
- Hugues Fausther-Bovendo
- Department of Microbiology and Immunology, Faculty of Medicine, Laval University, Quebec, Canada
| | - Xiangguo Qiu
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - George Giorgi Babuadze
- Department of Microbiology and Immunology, Faculty of Medicine, Laval University, Quebec, Canada
| | - Hiva Azizi
- Department of Microbiology and Immunology, Faculty of Medicine, Laval University, Quebec, Canada
| | - Jannie Pedersen
- Department of Microbiology and Immunology, Faculty of Medicine, Laval University, Quebec, Canada
| | - Gary Wong
- Department of Microbiology and Immunology, Faculty of Medicine, Laval University, Quebec, Canada
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Canada
| | - Gary P Kobinger
- Department of Microbiology and Immunology, Faculty of Medicine, Laval University, Quebec, Canada
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School 27 of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Jeremiah Matson M, Ricotta E, Feldmann F, Massaquoi M, Sprecher A, Giuliani R, Edwards JK, Rosenke K, de Wit E, Feldmann H, Chertow DS, Munster VJ. Evaluation of viral load in patients with Ebola virus disease in Liberia: a retrospective observational study. THE LANCET MICROBE 2022; 3:e533-e542. [PMID: 35617976 PMCID: PMC9254266 DOI: 10.1016/s2666-5247(22)00065-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/15/2022] [Accepted: 03/09/2022] [Indexed: 11/30/2022] Open
Abstract
Background Viral load in patients with Ebola virus disease affects case fatality rate and is an important parameter used for diagnostic cutoffs, stratification in randomised controlled trials, and epidemiological studies. However, viral load in Ebola virus disease is currently estimated using numerous different assays and protocols that were not developed or validated for this purpose. Here, our aim was to conduct a laboratory-based re-evaluation of the viral loads of a large cohort of Liberian patients with Ebola virus disease and analyse these data in the broader context of the west Africa epidemic. Methods In this retrospective observational study, whole blood samples from patients at the Eternal Love Winning Africa Ebola treatment unit (Monrovia, Liberia) were re-extracted with an optimised protocol and analysed by droplet digital PCR (ddPCR) using a novel semi-strand specific assay to measure viral load. To allow for more direct comparisons, the ddPCR viral loads were also back-calculated to cycle threshold (Ct) values. The new viral load data were then compared with the Ct values from the original diagnostic quantitative RT-PCR (qRT-PCR) testing to identify differing trends and discrepancies. Findings Between Aug 28 and Dec 18, 2014, 727 whole blood samples from 528 individuals were collected. 463 (64%) were first-draw samples and 409 (56%) were from patients positive for Ebola virus (EBOV), species Zaire ebolavirus. Of the 307 first-draw EBOV-positive samples, 127 (41%) were from survivors and 180 (59%) were from non-survivors; 155 (50%) were women, 145 (47%) were men, and seven (2%) were not recorded, and the mean age was 29·3 (SD 15·0) years for women and 31·8 (SD 14·8) years for men. Survivors had significantly lower mean viral loads at presentation than non-survivors in both the reanalysed dataset (5·61 [95% CI 5·34–5·87] vs 7·19 [6·99–7·38] log10 EBOV RNA copies per mL; p<0·0001) and diagnostic dataset (Ct value 28·72 [27·97–29·47] vs 26·26 [25·72–26·81]; p<0·0001). However, the prognostic capacity of viral load increased with the reanalysed dataset (odds ratio [OR] of death 8·06 [95% CI 4·81–13·53], p<0·0001 for viral loads above 6·71 log10 EBOV RNA copies per mL vs OR of death 2·02 [1·27–3·20], p=0·0028 for Ct values below 27·37). Diagnostic qRT-PCR significantly (p<0·0001) underestimated viral load in both survivors and non-survivors (difference in diagnostic Ct value minus laboratory Ct value of 1·79 [95% CI 1·16–2·43] for survivors and 5·15 [4·43–5·87] for non-survivors). Six samples that were reported negative by diagnostic testing were found to be positive upon reanalysis and had high viral loads. Interpretation Inaccurate viral load estimation from diagnostic Ct values is probably multifactorial; however, unaddressed PCR inhibition from tissue damage in patients with fulminant Ebola virus disease could largely account for the discrepancies observed in our study. Testing protocols for Ebola virus disease require further standardisation and validation to produce accurate viral load estimates, minimise false negatives, and allow for reliable epidemiological investigation.
Collapse
Affiliation(s)
- M Jeremiah Matson
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA; Marshall University Joan C Edwards School of Medicine, Huntington, WV, USA
| | - Emily Ricotta
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | | | | | | | | | - Kyle Rosenke
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Emmie de Wit
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Daniel S Chertow
- Critical Care Medicine Department, Clinical Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA; Laboratory of Immunoregulation, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
| | - Vincent J Munster
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
9
|
Hickman MR, Saunders DL, Bigger CA, Kane CD, Iversen PL. The development of broad-spectrum antiviral medical countermeasures to treat viral hemorrhagic fevers caused by natural or weaponized virus infections. PLoS Negl Trop Dis 2022; 16:e0010220. [PMID: 35259154 PMCID: PMC8903284 DOI: 10.1371/journal.pntd.0010220] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The Joint Program Executive Office for Chemical, Biological, Radiological, and Nuclear Defense (JPEO-CBRND) began development of a broad-spectrum antiviral countermeasure against deliberate use of high-consequence viral hemorrhagic fevers (VHFs) in 2016. The effort featured comprehensive preclinical research, including laboratory testing and rapid advancement of lead molecules into nonhuman primate (NHP) models of Ebola virus disease (EVD). Remdesivir (GS-5734, Veklury, Gilead Sciences) was the first small molecule therapeutic to successfully emerge from this effort. Remdesivir is an inhibitor of RNA-dependent RNA polymerase, a viral enzyme that is essential for viral replication. Its robust potency and broad-spectrum antiviral activity against certain RNA viruses including Ebola virus and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) led to its clinical evaluation in randomized, controlled trials (RCTs) in human patients during the 2018 EVD outbreak in the Democratic Republic of the Congo (DRC) and the ongoing Coronavirus Disease 2019 (COVID-19) pandemic today. Remdesivir was recently approved by the US Food and Drug Administration (FDA) for the treatment of COVID-19 requiring hospitalization. Substantial gaps remain in improving the outcomes of acute viral infections for patients afflicted with both EVD and COVID-19, including how to increase therapeutic breadth and strategies for the prevention and treatment of severe disease. Combination therapy that joins therapeutics with complimentary mechanisms of action appear promising, both preclinically and in RCTs. Importantly, significant programmatic challenges endure pertaining to a clear drug and biological product development pathway for therapeutics targeting biodefense and emerging pathogens when human efficacy studies are not ethical or feasible. For example, remdesivir's clinical development was facilitated by outbreaks of Ebola and SARS-CoV-2; as such, the development pathway employed for remdesivir is likely to be the exception rather than the rule. The current regulatory licensure pathway for therapeutics targeting rare, weaponizable VHF agents is likely to require use of FDA's established Animal Rule (21 CFR 314.600-650 for drugs; 21 CFR 601.90-95 for biologics). The FDA may grant marketing approval based on adequate and well-controlled animal efficacy studies when the results of those studies establish that the drug is safe and likely to produce clinical benefit in humans. In practical terms, this is anticipated to include a series of rigorous, well-documented, animal challenge studies, to include aerosol challenge, combined with human safety data. While small clinical studies against naturally occurring, high-consequence pathogens are typically performed where possible, approval for the therapeutics currently under development against biodefense pathogens will likely require the Animal Rule pathway utilizing studies in NHPs. We review the development of remdesivir as illustrative of the effort that will be needed to field future therapeutics against highly lethal, infectious agents.
Collapse
Affiliation(s)
- Mark R. Hickman
- Joint Project Manager for Chemical, Biological, Radiological, and Nuclear Medical (JPM CBRN Medical), Fort Detrick, Maryland, United States of America
| | - David L. Saunders
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Maryland, United States of America
| | - Catherine A. Bigger
- Logistics Management International Inc, Tysons Corner, Virginia, United States of America
| | | | | |
Collapse
|
10
|
Shu Q, Kenny T, Fan J, Lyon CJ, Cazares LH, Hu TY. Species-specific quantification of circulating ebolavirus burden using VP40-derived peptide variants. PLoS Pathog 2021; 17:e1010039. [PMID: 34748613 PMCID: PMC8601621 DOI: 10.1371/journal.ppat.1010039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 11/18/2021] [Accepted: 10/14/2021] [Indexed: 11/18/2022] Open
Abstract
Six ebolavirus species are reported to date, including human pathogens Bundibugyo virus (BDBV), Ebola virus (EBOV), Sudan virus (SUDV), and Taï Forest virus (TAFV); non-human pathogen Reston virus (RESTV); and the plausible Bombali virus (BOMV). Since there are differences in the disease severity caused by different species, species identification and viral burden quantification are critical for treating infected patients timely and effectively. Here we developed an immunoprecipitation-coupled mass spectrometry (IP-MS) assay for VP40 antigen detection and quantification. We carefully selected two regions of VP40, designated as peptide 8 and peptide12 from the protein sequence that showed minor variations among Ebolavirus species through MS analysis of tryptic peptides and antigenicity prediction based on available bioinformatic tools, and generated high-quality capture antibodies pan-specific for these variant peptides. We applied this assay to human plasma spiked with recombinant VP40 protein from EBOV, SUDV, and BDBV and virus-like particles (VLP), as well as EBOV infected NHP plasma. Sequence substitutions between EBOV and SUDV, the two species with highest lethality, produced affinity variations of 2.6-fold for p8 and 19-fold for p12. The proposed IP-MS assay differentiates four of the six known EBV species in one assay, through a combination of p8 and p12 data. The IP-MS assay limit of detection (LOD) using multiple reaction monitoring (MRM) as signal readout was determined to be 28 ng/mL and 7 ng/mL for EBOV and SUDV respectively, equivalent to ~1.625-6.5×105 Geq/mL, and comparable to the LOD of lateral flow immunoassays currently used for Ebola surveillance. The two peptides of the IP-MS assay were also identified by their tandem MS spectra using a miniature MALDI-TOF MS instrument, greatly increasing the feasibility of high specificity assay in a decentralized laboratory.
Collapse
Affiliation(s)
- Qingbo Shu
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Tara Kenny
- Systems and Structural Biology Division, Protein Sciences Branch, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Jia Fan
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Christopher J. Lyon
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, Louisiana, United States of America
| | - Lisa H. Cazares
- Systems and Structural Biology Division, Protein Sciences Branch, U.S. Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, United States of America
| | - Tony Y. Hu
- Center for Cellular and Molecular Diagnostics, Department of Biochemistry and Molecular Biology, School of Medicine, Tulane University, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
11
|
Barros MT, Veletić M, Kanada M, Pierobon M, Vainio S, Balasingham I, Balasubramaniam S. Molecular Communications in Viral Infections Research: Modeling, Experimental Data, and Future Directions. IEEE TRANSACTIONS ON MOLECULAR, BIOLOGICAL, AND MULTI-SCALE COMMUNICATIONS 2021; 7:121-141. [PMID: 35782714 PMCID: PMC8544950 DOI: 10.1109/tmbmc.2021.3071780] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 12/22/2022]
Abstract
Hundreds of millions of people worldwide are affected by viral infections each year, and yet, several of them neither have vaccines nor effective treatment during and post-infection. This challenge has been highlighted by the COVID-19 pandemic, showing how viruses can quickly spread and impact society as a whole. Novel interdisciplinary techniques must emerge to provide forward-looking strategies to combat viral infections, as well as possible future pandemics. In the past decade, an interdisciplinary area involving bioengineering, nanotechnology and information and communication technology (ICT) has been developed, known as Molecular Communications. This new emerging area uses elements of classical communication systems to molecular signalling and communication found inside and outside biological systems, characterizing the signalling processes between cells and viruses. In this paper, we provide an extensive and detailed discussion on how molecular communications can be integrated into the viral infectious diseases research, and how possible treatment and vaccines can be developed considering molecules as information carriers. We provide a literature review on molecular communications models for viral infection (intra-body and extra-body), a deep analysis on their effects on immune response, how experimental can be used by the molecular communications community, as well as open issues and future directions.
Collapse
Affiliation(s)
- Michael Taynnan Barros
- CBIG/BioMediTechTampere University33014TampereFinland
- School of Computer Science and Electronic EngineeringUniversity of EssexColchesterCO4 3SQU.K.
| | - Mladen Veletić
- Intervention CentreOslo University Hospital0424OsloNorway
- Department of Electronic SystemsNorwegian University of Science and Technology7491TrondheimNorway
| | - Masamitsu Kanada
- Department of Pharmacology and ToxicologyInstitute for Quantitative Health Science and Engineering, Michigan State UniversityEast LansingMI48824USA
| | - Massimiliano Pierobon
- Department of Computer Science and EngineeringUniversity of Nebraska–LincolnLincolnNE68588USA
| | - Seppo Vainio
- InfoTech OuluKvantum Institute, Faculty of Biochemistry and Molecular Medicine, Laboratory of Developmental Biology, Oulu University90570OuluFinland
| | - Ilangko Balasingham
- Intervention CentreOslo University Hospital0424OsloNorway
- Department of Electronic SystemsNorwegian University of Science and Technology7491TrondheimNorway
| | | |
Collapse
|
12
|
Huang Y, Xiao S, Yuan Z. Comparison and Evaluation of Real-Time Taqman PCR for Detection and Quantification of Ebolavirus. Viruses 2021; 13:1575. [PMID: 34452440 PMCID: PMC8402893 DOI: 10.3390/v13081575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 01/12/2023] Open
Abstract
Given that ebolavirus causes severe and frequently lethal disease, its rapid and accurate detection using available and validated methods is essential for controlling infection. Real-time reverse-transcription PCR (RT-PCR) has proven to be an invaluable tool for ebolaviruses diagnostics. Many assays with different targets have been developed, but they have not been externally compared or validated, and limits of detection are not uniformly reported. Here we compared and evaluated the sensitivity, reproducibility and specificity of 23 in-house assays under the same conditions. Our results showed that these assays were highly gene- and species- specific when evaluated using in vitro RNA transcripts and viral RNA, and the potential limits of detection were uniformly reported ranging from 102 to 106 in vitro synthesized RNA transcripts copies perμL and 1-100 TCID50/mL. The comparison of these assays indicated that those targeting the more conservative NP gene could be the better option for EVD case definition and quantitative measurement because of its higher sensitivity for the same species. Our analysis could contribute to the standardization of ebolavirus detection and quantification assays, which can offer a better understanding of the meaning of results across laboratories and time points, as well as make them easy to implement, especially under outbreak conditions.
Collapse
Affiliation(s)
- Yi Huang
- National Biosafety Laboratory, Chinese Academy of Sciences, Wuhan 430020, China
| | - Shuqi Xiao
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430020, China;
| | - Zhiming Yuan
- National Biosafety Laboratory, Chinese Academy of Sciences, Wuhan 430020, China
| |
Collapse
|
13
|
Niemuth NA, Fallacara D, Triplett CA, Tamrakar SM, Rajbhandari A, Florence C, Ward L, Griffiths A, Carrion R, Goez-Gazi Y, Alfson KJ, Staples HM, Brasel T, Comer JE, Massey S, Smith J, Kocsis A, Lowry J, Johnston SC, Nalca A, Goff AJ, Shurtleff AC, Pitt ML, Trefry J, Fay MP. Natural history of disease in cynomolgus monkeys exposed to Ebola virus Kikwit strain demonstrates the reliability of this non-human primate model for Ebola virus disease. PLoS One 2021; 16:e0252874. [PMID: 34214118 PMCID: PMC8253449 DOI: 10.1371/journal.pone.0252874] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/24/2021] [Indexed: 12/20/2022] Open
Abstract
Filoviruses (Family Filoviridae genera Ebolavirus and Marburgvirus) are negative-stranded RNA viruses that cause severe health effects in humans and non-human primates, including death. Except in outbreak settings, vaccines and other medical countermeasures against Ebola virus (EBOV) will require testing under the FDA Animal Rule. Multiple vaccine candidates have been evaluated using cynomolgus monkeys (CM) exposed to EBOV Kikwit strain. To the best of our knowledge, however, animal model development data supporting the use of CM in vaccine research have not been submitted to the FDA. This study describes a large CM database (122 CM, 62 female and 60 male, age 2 to 9 years) and demonstrates the consistency of the CM model through time to death models and descriptive statistics. CMs were exposed to EBOV doses of 0.1 to 100,000 PFU in 33 studies conducted at three Animal Biosafety Level 4 facilities, by three exposure routes. Time to death was modeled using Cox proportional hazards models with a frailty term that incorporated study-to-study variability. Despite significant differences attributed to exposure variables, all CMs exposed to the 100 to 1,000 pfu doses commonly used in vaccine studies died or met euthanasia criteria within 21 days of exposure, median 7 days, 93% between 5 and 12 days of exposure. Moderate clinical signs were observed 4 to 5 days after exposure and preceded death or euthanasia by approximately one day. Viremia was detected within a few days of infection. Hematology indices were indicative of viremia and the propensity for hemorrhage with progression of Ebola viremia. Changes associated with coagulation parameters and platelets were consistent with coagulation disruption. Changes in leukocyte profiles were indicative of an acute inflammatory response. Increased liver enzymes were observed shortly after exposure. Taken together, these factors suggest that the cynomolgus monkey is a reliable animal model for human disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Clint Florence
- Non-Clinical Advisor for Joint Program Manager Chemical, Biological, Radiological and Nuclear Medical (JPM CBRN Medical) Joint Program Executive Office for Chemical, Biological, Radiological and Nuclear Defense (JPEO CBRND), Fort Detrick, MD, United States of America
| | - Lucy Ward
- Advanced Vaccines & Immunologics, JPM CBRN Medical, JPEO CBRND, Fort Detrick, MD, United States of America
| | - Anthony Griffiths
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, United States of America
| | - Ricardo Carrion
- Texas Biomedical Research Institute, San Antonio, TX, United States of America
| | - Yenny Goez-Gazi
- Texas Biomedical Research Institute, San Antonio, TX, United States of America
| | - Kendra J. Alfson
- Texas Biomedical Research Institute, San Antonio, TX, United States of America
| | - Hilary M. Staples
- Texas Biomedical Research Institute, San Antonio, TX, United States of America
| | - Trevor Brasel
- Department of Microbiology and Immunology/Institutional Office of Regulated Nonclinical Studies, University of Texas Medical Branch (UTMB), Galveston, TX, United States of America
| | - Jason E. Comer
- Department of Microbiology and Immunology/Institutional Office of Regulated Nonclinical Studies, University of Texas Medical Branch (UTMB), Galveston, TX, United States of America
| | - Shane Massey
- Institutional Office of Regulated Nonclinical Studies, UTMB, Galveston, TX, United States of America
| | - Jeanon Smith
- Institutional Office of Regulated Nonclinical Studies, UTMB, Galveston, TX, United States of America
| | - Andrew Kocsis
- Animal Resources Center, UTMB. Galveston, TX, United States of America
| | - Jake Lowry
- Animal Resources Center, UTMB. Galveston, TX, United States of America
| | - Sara C. Johnston
- Virology Division, United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, MD, United States of America
| | - Aysegul Nalca
- Core Support Directorate, USAMRIID, Fort Detrick, MD, United States of America
| | - Arthur J. Goff
- Commander’s Special Staff, Deputy Director Research Program Office, USAMRIID, Fort Detrick, MD, United States of America
| | - Amy C. Shurtleff
- Molecular Biology Division, USAMRIID, Fort Detrick, MD, United States of America
| | | | - John Trefry
- Vaccines/Therapeutics Division (CBM), Defense Threat Reduction Agency, Fort Belvoir, Virginia, United States of America
| | - Michael P. Fay
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
14
|
Characterization of Ebola Virus Risk to Bedside Providers in an Intensive Care Environment. Microorganisms 2021; 9:microorganisms9030498. [PMID: 33652895 PMCID: PMC7996731 DOI: 10.3390/microorganisms9030498] [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: 01/30/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 11/20/2022] Open
Abstract
Background: The 2014–2016 Ebola outbreak in West Africa recapitulated that nosocomial spread of Ebola virus could occur and that health care workers were at particular risk including notable cases in Europe and North America. These instances highlighted the need for centers to better prepare for potential Ebola virus cases; including understanding how the virus spreads and which interventions pose the greatest risk. Methods: We created a fully equipped intensive care unit (ICU), within a Biosafety Level 4 (BSL4) laboratory, and infected multiple sedated non-human primates (NHPs) with Ebola virus. While providing bedside care, we sampled blood, urine, and gastric residuals; as well as buccal, ocular, nasal, rectal, and skin swabs, to assess the risks associated with routine care. We also assessed the physical environment at end-point. Results: Although viral RNA was detectable in blood as early as three days post-infection, it was not detectable in the urine, gastric fluid, or swabs until late-stage disease. While droplet spread and fomite contamination were present on a few of the surfaces that were routinely touched while providing care in the ICU for the infected animal, these may have been abrogated through good routine hygiene practices. Conclusions: Overall this study has helped further our understanding of which procedures may pose the highest risk to healthcare providers and provides temporal evidence of this over the clinical course of disease.
Collapse
|
15
|
Nonhuman primate to human immunobridging to infer the protective effect of an Ebola virus vaccine candidate. NPJ Vaccines 2020; 5:112. [PMID: 33335092 PMCID: PMC7747701 DOI: 10.1038/s41541-020-00261-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/13/2020] [Indexed: 01/07/2023] Open
Abstract
It has been proven challenging to conduct traditional efficacy trials for Ebola virus (EBOV) vaccines. In the absence of efficacy data, immunobridging is an approach to infer the likelihood of a vaccine protective effect, by translating vaccine immunogenicity in humans to a protective effect, using the relationship between vaccine immunogenicity and the desired outcome in a suitable animal model. We here propose to infer the protective effect of the Ad26.ZEBOV, MVA-BN-Filo vaccine regimen with an 8-week interval in humans by immunobridging. Immunogenicity and protective efficacy data were obtained for Ad26.ZEBOV and MVA-BN-Filo vaccine regimens using a fully lethal EBOV Kikwit challenge model in cynomolgus monkeys (nonhuman primates [NHP]). The association between EBOV neutralizing antibodies, glycoprotein (GP)-binding antibodies, and GP-reactive T cells and survival in NHP was assessed by logistic regression analysis. Binding antibodies against the EBOV surface GP were identified as the immune parameter with the strongest correlation to survival post EBOV challenge, and used to infer the predicted protective effect of the vaccine in humans using published data from phase I studies. The human vaccine-elicited EBOV GP-binding antibody levels are in a range associated with significant protection against mortality in NHP. Based on this immunobridging analysis, the EBOV GP-specific-binding antibody levels elicited by the Ad26.ZEBOV, MVA-BN-Filo vaccine regimen in humans will likely provide protection against EBOV disease.
Collapse
|
16
|
Ebola Virus Disease Survivors Show More Efficient Antibody Immunity than Vaccinees Despite Similar Levels of Circulating Immunoglobulins. Viruses 2020; 12:v12090915. [PMID: 32825479 PMCID: PMC7552031 DOI: 10.3390/v12090915] [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] [Received: 06/30/2020] [Revised: 08/06/2020] [Accepted: 08/18/2020] [Indexed: 01/01/2023] Open
Abstract
The last seven years have seen the greatest surge of Ebola virus disease (EVD) cases in equatorial Africa, including the 2013-2016 epidemic in West Africa and the recent epidemics in the Democratic Republic of Congo (DRC). The vaccine clinical trials that took place in West Africa and the DRC, as well as follow-up studies in collaboration with EVD survivor communities, have for the first time allowed researchers to compare immune memory induced by natural infection and vaccination. These comparisons may be relevant to evaluate the putative effectiveness of vaccines and candidate medical countermeasures such as convalescent plasma transfer. In this study, we compared the long-term functionality of anti-EBOV glycoprotein (GP) antibodies from EVD survivors with that from volunteers who received the recombinant vesicular stomatitis virus vectored vaccine (rVSV-ZEBOV) during the Phase I clinical trial in Hamburg. Our study highlights important differences between EBOV vaccination and natural infection and provides a framework for comparison with other vaccine candidates.
Collapse
|
17
|
Di Paola N, Sanchez-Lockhart M, Zeng X, Kuhn JH, Palacios G. Viral genomics in Ebola virus research. Nat Rev Microbiol 2020; 18:365-378. [PMID: 32367066 PMCID: PMC7223634 DOI: 10.1038/s41579-020-0354-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2020] [Indexed: 12/20/2022]
Abstract
Filoviruses such as Ebola virus continue to pose a substantial health risk to humans. Advances in the sequencing and functional characterization of both pathogen and host genomes have provided a wealth of knowledge to clinicians, epidemiologists and public health responders during outbreaks of high-consequence viral disease. Here, we describe how genomics has been historically used to investigate Ebola virus disease outbreaks and how new technologies allow for rapid, large-scale data generation at the point of care. We highlight how genomics extends beyond consensus-level sequencing of the virus to include intra-host viral transcriptomics and the characterization of host responses in acute and persistently infected patients. Similar genomics techniques can also be applied to the characterization of non-human primate animal models and to known natural reservoirs of filoviruses, and metagenomic sequencing can be the key to the discovery of novel filoviruses. Finally, we outline the importance of reverse genetics systems that can swiftly characterize filoviruses as soon as their genome sequences are available.
Collapse
Affiliation(s)
- Nicholas Di Paola
- 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
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Gustavo Palacios
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA.
| |
Collapse
|
18
|
Forna A, Nouvellet P, Dorigatti I, Donnelly CA. Case Fatality Ratio Estimates for the 2013-2016 West African Ebola Epidemic: Application of Boosted Regression Trees for Imputation. Clin Infect Dis 2020; 70:2476-2483. [PMID: 31328221 PMCID: PMC7286386 DOI: 10.1093/cid/ciz678] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 07/17/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The 2013-2016 West African Ebola epidemic has been the largest to date with >11 000 deaths in the affected countries. The data collected have provided more insight into the case fatality ratio (CFR) and how it varies with age and other characteristics. However, the accuracy and precision of the naive CFR remain limited because 44% of survival outcomes were unreported. METHODS Using a boosted regression tree model, we imputed survival outcomes (ie, survival or death) when unreported, corrected for model imperfection to estimate the CFR without imputation, with imputation, and adjusted with imputation. The method allowed us to further identify and explore relevant clinical and demographic predictors of the CFR. RESULTS The out-of-sample performance (95% confidence interval [CI]) of our model was good: sensitivity, 69.7% (52.5-75.6%); specificity, 69.8% (54.1-75.6%); percentage correctly classified, 69.9% (53.7-75.5%); and area under the receiver operating characteristic curve, 76.0% (56.8-82.1%). The adjusted CFR estimates (95% CI) for the 2013-2016 West African epidemic were 82.8% (45.6-85.6%) overall and 89.1% (40.8-91.6%), 65.6% (61.3-69.6%), and 79.2% (45.4-84.1%) for Sierra Leone, Guinea, and Liberia, respectively. We found that district, hospitalisation status, age, case classification, and quarter (date of case reporting aggregated at three-month intervals) explained 93.6% of the variance in the naive CFR. CONCLUSIONS The adjusted CFR estimates improved the naive CFR estimates obtained without imputation and were more representative. Used in conjunction with other resources, adjusted estimates will inform public health contingency planning for future Ebola epidemics, and help better allocate resources and evaluate the effectiveness of future inventions.
Collapse
Affiliation(s)
- Alpha Forna
- Medical Research Council Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Brighton, Brighton, United Kingdom, and Imperial College London, London, United Kingdom
| | - Pierre Nouvellet
- Medical Research Council Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Brighton, Brighton, United Kingdom, and Imperial College London, London, United Kingdom
- School of Life Sciences, University of Sussex, Brighton, Brighton, United Kingdom
| | - Ilaria Dorigatti
- Medical Research Council Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Brighton, Brighton, United Kingdom, and Imperial College London, London, United Kingdom
| | - Christl A Donnelly
- Medical Research Council Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Brighton, Brighton, United Kingdom, and Imperial College London, London, United Kingdom
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Boisen ML, Uyigue E, Aiyepada J, Siddle KJ, Oestereich L, Nelson DKS, Bush DJ, Rowland MM, Heinrich ML, Eromon P, Kayode AT, Odia I, Adomeh DI, Muoebonam EB, Akhilomen P, Okonofua G, Osiemi B, Omoregie O, Airende M, Agbukor J, Ehikhametalor S, Aire CO, Duraffour S, Pahlmann M, Böhm W, Barnes KG, Mehta S, Momoh M, Sandi JD, Goba A, Folarin OA, Ogbaini-Emovan E, Asogun DA, Tobin EA, Akpede GO, Okogbenin SA, Okokhere PO, Grant DS, Schieffelin JS, Sabeti PC, Günther S, Happi CT, Branco LM, Garry RF. Field evaluation of a Pan-Lassa rapid diagnostic test during the 2018 Nigerian Lassa fever outbreak. Sci Rep 2020; 10:8724. [PMID: 32457420 PMCID: PMC7250850 DOI: 10.1038/s41598-020-65736-0] [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: 03/02/2020] [Accepted: 05/08/2020] [Indexed: 01/07/2023] Open
Abstract
Lassa virus (LASV) is the causative agent of Lassa fever (LF), an often-fatal hemorrhagic disease. LF is endemic in Nigeria, Sierra Leone and other West African countries. Diagnosis of LASV infection is challenged by the genetic diversity of the virus, which is greatest in Nigeria. The ReLASV Pan-Lassa Antigen Rapid Test (Pan-Lassa RDT) is a point-of-care, in vitro diagnostic test that utilizes a mixture of polyclonal antibodies raised against recombinant nucleoproteins of representative strains from the three most prevalent LASV lineages (II, III and IV). We compared the performance of the Pan-LASV RDT to available quantitative PCR (qPCR) assays during the 2018 LF outbreak in Nigeria. For patients with acute LF (RDT positive, IgG/IgM negative) during initial screening, RDT performance was 83.3% sensitivity and 92.8% specificity when compared to composite results of two qPCR assays. 100% of samples that gave Ct values below 22 on both qPCR assays were positive on the Pan-Lassa RDT. There were significantly elevated case fatality rates and elevated liver transaminase levels in subjects whose samples were RDT positive compared to RDT negative.
Collapse
Affiliation(s)
| | - Eghosa Uyigue
- The African Center of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Osun State, Nigeria
- Department of Biological Sciences, College of Natural Sciences, Redeemer's University, Ede, Osun State, Nigeria
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - John Aiyepada
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Katherine J Siddle
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard University, Cambridge, MA, USA
- The Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Lisa Oestereich
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg, Germany
| | | | | | | | | | - Philomena Eromon
- The African Center of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Osun State, Nigeria
| | - Adeyemi T Kayode
- The African Center of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Osun State, Nigeria
- Department of Biological Sciences, College of Natural Sciences, Redeemer's University, Ede, Osun State, Nigeria
| | - Ikponmwosa Odia
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Donatus I Adomeh
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Ekene B Muoebonam
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Patience Akhilomen
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Grace Okonofua
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Blessing Osiemi
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Omigie Omoregie
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Michael Airende
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Jacqueline Agbukor
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Solomon Ehikhametalor
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Chris Okafi Aire
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Sophie Duraffour
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg, Germany
| | - Meike Pahlmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg, Germany
| | - Wiebke Böhm
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg, Germany
| | - Kayla G Barnes
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard University, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Samar Mehta
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard University, Cambridge, MA, USA
- Beth Israel Deaconess Medical Center, Division of Infectious Diseases, Boston, MA, USA
| | - Mambu Momoh
- Eastern Polytechnic Institute, Kenema, Sierra Leone
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - John Demby Sandi
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Augustine Goba
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Onikepe A Folarin
- The African Center of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Osun State, Nigeria
- Department of Biological Sciences, College of Natural Sciences, Redeemer's University, Ede, Osun State, Nigeria
| | - Ephraim Ogbaini-Emovan
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Danny A Asogun
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Ekaete A Tobin
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - George O Akpede
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Sylvanus A Okogbenin
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Peter O Okokhere
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
- The Department of Medicine, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
- The Department of Medicine, Faculty of Clinical Sciences, Ambrose Alli University, Ekpoma, Edo State, Nigeria
| | - Donald S Grant
- Viral Hemorrhagic Fever Program, Kenema Government Hospital, Kenema, Sierra Leone
- Ministry of Health and Sanitation, Freetown, Sierra Leone
- College of Medicine and Allied Health Sciences, University of Sierra Leone, Freetown, Sierra Leone
| | - John S Schieffelin
- Sections of Infectious Disease, Departments of Pediatrics and Internal Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Pardis C Sabeti
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard University, Cambridge, MA, USA
- The Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Harvard-MIT Health Sciences and Technology, MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Stephan Günther
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Hamburg, Germany
| | - Christian T Happi
- The African Center of Excellence for Genomics of Infectious Diseases, Redeemer's University, Ede, Osun State, Nigeria.
- Department of Biological Sciences, College of Natural Sciences, Redeemer's University, Ede, Osun State, Nigeria.
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria.
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA.
| | | | - Robert F Garry
- Zalgen Labs, LLC, Germantown, MD, USA.
- Tulane Health Sciences Center, Tulane University, New Orleans, LA, USA.
- Tulane University, School of Medicine, Department of Microbiology and Immunology, New Orleans, LA, USA.
| |
Collapse
|
20
|
Ebola Virus Disease: Epidemiology, Clinical Features, Management, and Prevention. Infect Dis Clin North Am 2020; 33:953-976. [PMID: 31668200 DOI: 10.1016/j.idc.2019.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Ebola virus disease (EVD) is a deadly zoonotic disease caused by the Ebola virus. There is no specific treatment approved for EVD. Supportive care and management of complications are mainstays of treatment. Effective outbreak control requires a multidisciplinary team effort applying case management, infection prevention and control practices, surveillance and contact tracing, a good laboratory service, safe and dignified burials, and social and community mobilization. This article highlights the epidemiology, clinical features, diagnosis, management, and prevention of EVD. The emerging diagnostic technologies, rapid viral characterization, geospatial mapping of EVD transmission, and new treatments and vaccines are discussed.
Collapse
|
21
|
Kerber R, Lorenz E, Duraffour S, Sissoko D, Rudolf M, Jaeger A, Cisse SD, Camara AM, Miranda O, Castro CM, Akoi Bore J, Raymond Koundouno F, Repits J, Afrough B, Becker-Ziaja B, Hinzmann J, Mertens M, Vitoriano I, Hugh Logue C, Böttcher JP, Pallasch E, Sachse A, Bah A, Cabeza-Cabrerizo M, Nitzsche K, Kuisma E, Michel J, Holm T, Zekeng EG, Cowley LA, Garcia-Dorival I, Hetzelt N, Baum JHJ, Portmann J, Carter L, Yenamaberhan RL, Camino A, Enkirch T, Singethan K, Meisel S, Mazzarelli A, Kosgei A, Kafetzopoulou L, Rickett NY, Patrono LV, Ghebreghiorghis L, Arnold U, Colin G, Juchet S, Marchal CL, Kolie JS, Beavogui AH, Wurr S, Bockholt S, Krumkamp R, May J, Stoecker K, Fleischmann E, Ippolito G, Carroll MW, Koivogui L, Magassouba N, Keita S, Gurry C, Drury P, Diallo B, Formenty P, Wölfel R, Di Caro A, Gabriel M, Anglaret X, Malvy D, Günther S. Laboratory Findings, Compassionate Use of Favipiravir, and Outcome in Patients With Ebola Virus Disease, Guinea, 2015-A Retrospective Observational Study. J Infect Dis 2020; 220:195-202. [PMID: 30788508 PMCID: PMC6581890 DOI: 10.1093/infdis/jiz078] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 02/14/2019] [Indexed: 11/14/2022] Open
Abstract
Background In 2015, the laboratory at the Ebola treatment center in Coyah, Guinea, confirmed Ebola virus disease (EVD) in 286 patients. The cycle threshold (Ct) of an Ebola virus–specific reverse transcription–polymerase chain reaction assay and 13 blood chemistry parameters were measured on admission and during hospitalization. Favipiravir treatment was offered to patients with EVD on a compassionate-use basis. Methods To reduce biases in the raw field data, we carefully selected 163 of 286 patients with EVD for a retrospective study to assess associations between potential risk factors, alterations in blood chemistry findings, favipiravir treatment, and outcome. Results The case-fatality rate in favipiravir-treated patients was lower than in untreated patients (42.5% [31 of 73] vs 57.8% [52 of 90]; P = .053 by univariate analysis). In multivariate regression analysis, a higher Ct and a younger age were associated with survival (P < .001), while favipiravir treatment showed no statistically significant effect (P = .11). However, Kaplan-Meier analysis indicated a longer survival time in the favipiravir-treated group (P = .015). The study also showed characteristic changes in blood chemistry findings in patients who died, compared with survivors. Conclusions Consistent with the JIKI trial, this retrospective study revealed a trend toward improved survival in favipiravir- treated patients; however, the effect of treatment was not statistically significant, except for its influence on survival time.
Collapse
Affiliation(s)
- Romy Kerber
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Eva Lorenz
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,German Center for Infection Research, Braunschweig
| | - Sophie Duraffour
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Daouda Sissoko
- INSERM U1219, Bordeaux University, Bordeaux, France.,Bordeaux University Hospital, Bordeaux, France
| | - Martin Rudolf
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Anna Jaeger
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,German Center for Infection Research, Braunschweig
| | | | | | | | | | - Joseph Akoi Bore
- European Mobile Laboratory Consortium, Hamburg.,Ministry of Health Guinea, Guinea
| | | | - Johanna Repits
- European Mobile Laboratory Consortium, Hamburg.,Janssen-Cilag, Sollentuna, Sweden
| | - Babak Afrough
- European Mobile Laboratory Consortium, Hamburg.,Public Health England, Porton Down
| | - Beate Becker-Ziaja
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Julia Hinzmann
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Marc Mertens
- European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig.,Friedrich Loeffler Institute, Federal Research Institute for Animal Health, Greifswald
| | - Ines Vitoriano
- European Mobile Laboratory Consortium, Hamburg.,Public Health England, Porton Down
| | | | - Jan-Peter Böttcher
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Elisa Pallasch
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Andreas Sachse
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Amadou Bah
- European Mobile Laboratory Consortium, Hamburg.,Swiss Tropical and Public Health Institute, Basel
| | | | | | - Eeva Kuisma
- European Mobile Laboratory Consortium, Hamburg.,Public Health England, Porton Down
| | - Janine Michel
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Tobias Holm
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | | | - Lauren A Cowley
- European Mobile Laboratory Consortium, Hamburg.,Public Health England, London.,Milner Centre for Evolution, University of Bath, Bath
| | - Isabel Garcia-Dorival
- European Mobile Laboratory Consortium, Hamburg.,Institute of Infection and Global Health, University of Liverpool, Liverpool
| | - Nicole Hetzelt
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Jonathan Hans Josef Baum
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg
| | - Jasmine Portmann
- European Mobile Laboratory Consortium, Hamburg.,Federal Office for Civil Protection, Spiez Laboratory, Spiez
| | - Lisa Carter
- European Mobile Laboratory Consortium, Hamburg.,University College London, London.,World Health Organization, Geneva, Switzerland
| | - Rahel Lemma Yenamaberhan
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg
| | | | - Theresa Enkirch
- European Mobile Laboratory Consortium, Hamburg.,Division of Veterinary Medicine, Paul-Ehrlich-Institut, Langen, Germany
| | - Katrin Singethan
- European Mobile Laboratory Consortium, Hamburg.,Institute of Virology, Technische Universität München, Munich, Germany
| | - Sarah Meisel
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg
| | - Antonio Mazzarelli
- European Mobile Laboratory Consortium, Hamburg.,National Institute for Infectious Diseases "Lazzaro Spallanzani," Rome, Italy
| | - Abigail Kosgei
- European Mobile Laboratory Consortium, Hamburg.,Kenya Medical Research Institute, Nairobi, Kenya
| | - Liana Kafetzopoulou
- European Mobile Laboratory Consortium, Hamburg.,KU Leuven-University of Leuven, Rega Institute for Medical Research, Leuven, Belgium
| | - Natasha Y Rickett
- European Mobile Laboratory Consortium, Hamburg.,Institute of Infection and Global Health, University of Liverpool, Liverpool
| | - Livia Victoria Patrono
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg
| | | | - Ulrike Arnold
- European Mobile Laboratory Consortium, Hamburg.,Robert Koch Institute, Berlin
| | - Géraldine Colin
- INSERM U1219, Bordeaux University, Bordeaux, France.,Bordeaux University Hospital, Bordeaux, France.,PAC-CI, ANRS Research Site, Treichville University Hospital, Abidjan, Côte d'Ivoire
| | - Sylvain Juchet
- INSERM U1219, Bordeaux University, Bordeaux, France.,Bordeaux University Hospital, Bordeaux, France.,PAC-CI, ANRS Research Site, Treichville University Hospital, Abidjan, Côte d'Ivoire
| | | | | | | | - Stephanie Wurr
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Sabrina Bockholt
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Ralf Krumkamp
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,German Center for Infection Research, Braunschweig
| | - Jürgen May
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,German Center for Infection Research, Braunschweig
| | - Kilian Stoecker
- European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig.,Bundeswehr Institute of Microbiology, Munich, Germany
| | - Erna Fleischmann
- European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig.,Bundeswehr Institute of Microbiology, Munich, Germany
| | - Giuseppe Ippolito
- European Mobile Laboratory Consortium, Hamburg.,National Institute for Infectious Diseases "Lazzaro Spallanzani," Rome, Italy
| | - Miles W Carroll
- European Mobile Laboratory Consortium, Hamburg.,Public Health England, Porton Down.,University of Southampton, South General Hospital, Southampton, United Kingdom
| | | | - N'Faly Magassouba
- Laboratoire des Fièvres Hémorragiques en Guinée, Université Gamal Abdel Nasser de Conakry, Guinea
| | | | | | | | | | | | - Roman Wölfel
- European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig.,Bundeswehr Institute of Microbiology, Munich, Germany
| | - Antonino Di Caro
- European Mobile Laboratory Consortium, Hamburg.,National Institute for Infectious Diseases "Lazzaro Spallanzani," Rome, Italy
| | - Martin Gabriel
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| | - Xavier Anglaret
- INSERM U1219, Bordeaux University, Bordeaux, France.,Bordeaux University Hospital, Bordeaux, France.,PAC-CI, ANRS Research Site, Treichville University Hospital, Abidjan, Côte d'Ivoire
| | - Denis Malvy
- INSERM U1219, Bordeaux University, Bordeaux, France.,Bordeaux University Hospital, Bordeaux, France
| | - Stephan Günther
- Bernhard Nocht Institute for Tropical Medicine, Hamburg.,European Mobile Laboratory Consortium, Hamburg.,German Center for Infection Research, Braunschweig
| |
Collapse
|
22
|
Immune correlates of postexposure vaccine protection against Marburg virus. Sci Rep 2020; 10:3071. [PMID: 32080323 PMCID: PMC7033120 DOI: 10.1038/s41598-020-59976-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 01/28/2020] [Indexed: 12/19/2022] Open
Abstract
Postexposure immunization can prevent disease and reduce transmission following pathogen exposure. The rapid immunostimulatory properties of recombinant vesicular stomatitis virus (rVSV)-based vaccines make them suitable postexposure treatments against the filoviruses Ebola virus and Marburg virus (MARV); however, the mechanisms that drive this protection are undefined. Previously, we reported 60–75% survival of rhesus macaques treated with rVSV vectors expressing MARV glycoprotein (GP) 20–30 minutes after a low dose exposure to the most pathogenic variant of MARV, Angola. Survival in this model was linked to production of GP-specific antibodies and lower viral load. To confirm these results and potentially identify novel correlates of postexposure protection, we performed a similar experiment, but analyzed plasma cytokine levels, frequencies of immune cell subsets, and the transcriptional response to infection in peripheral blood. In surviving macaques (80–89%), we observed induction of genes mapping to antiviral and interferon-related pathways early after treatment and a higher percentage of T helper 1 (Th1) and NK cells. In contrast, the response of non-surviving macaques was characterized by hypercytokinemia; a T helper 2 signature; recruitment of low HLA-DR expressing monocytes and regulatory T-cells; and transcription of immune checkpoint (e.g., PD-1, LAG3) genes. These results suggest dysregulated immunoregulation is associated with poor prognosis, whereas early innate signaling and Th1-skewed immunity are important for survival.
Collapse
|
23
|
Jacob ST, Crozier I, Fischer WA, Hewlett A, Kraft CS, Vega MADL, Soka MJ, Wahl V, Griffiths A, Bollinger L, Kuhn JH. Ebola virus disease. Nat Rev Dis Primers 2020; 6:13. [PMID: 32080199 PMCID: PMC7223853 DOI: 10.1038/s41572-020-0147-3] [Citation(s) in RCA: 304] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2020] [Indexed: 12/16/2022]
Abstract
Ebola virus disease (EVD) is a severe and frequently lethal disease caused by Ebola virus (EBOV). EVD outbreaks typically start from a single case of probable zoonotic transmission, followed by human-to-human transmission via direct contact or contact with infected bodily fluids or contaminated fomites. EVD has a high case-fatality rate; it is characterized by fever, gastrointestinal signs and multiple organ dysfunction syndrome. Diagnosis requires a combination of case definition and laboratory tests, typically real-time reverse transcription PCR to detect viral RNA or rapid diagnostic tests based on immunoassays to detect EBOV antigens. Recent advances in medical countermeasure research resulted in the recent approval of an EBOV-targeted vaccine by European and US regulatory agencies. The results of a randomized clinical trial of investigational therapeutics for EVD demonstrated survival benefits from two monoclonal antibody products targeting the EBOV membrane glycoprotein. New observations emerging from the unprecedented 2013-2016 Western African EVD outbreak (the largest in history) and the ongoing EVD outbreak in the Democratic Republic of the Congo have substantially improved the understanding of EVD and viral persistence in survivors of EVD, resulting in new strategies toward prevention of infection and optimization of clinical management, acute illness outcomes and attendance to the clinical care needs of patients.
Collapse
Affiliation(s)
- Shevin T Jacob
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
- Global Health Security Department, Infectious Diseases Institute, Makerere University, Kampala, Uganda
| | - Ian Crozier
- Integrated Research Facility at Fort Detrick, Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research supported by the National Cancer Institute, Frederick, MD, USA
| | - William A Fischer
- Department of Medicine, Division of Pulmonary Disease and Critical Care Medicine, Chapel Hill, NC, USA
| | - Angela Hewlett
- Nebraska Biocontainment Unit, Division of Infectious Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Colleen S Kraft
- Microbiology Section, Emory Medical Laboratory, Emory University School of Medicine, Atlanta, GA, USA
| | - Marc-Antoine de La Vega
- Department of Microbiology, Immunology & Infectious Diseases, Université Laval, Quebec City, QC, Canada
| | - Moses J Soka
- Partnership for Ebola Virus Disease Research in Liberia, Monrovia Medical Units ELWA-2 Hospital, Monrovia, Liberia
| | - Victoria Wahl
- National Biodefense Analysis and Countermeasures Center, Fort Detrick, Frederick, MD, USA
| | - Anthony Griffiths
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, Boston, MA, USA
| | - Laura Bollinger
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA.
| |
Collapse
|
24
|
Postigo-Hidalgo I, Fischer C, Moreira-Soto A, Tscheak P, Nagel M, Eickmann M, Drexler JF. Pre-emptive genomic surveillance of emerging ebolaviruses. ACTA ACUST UNITED AC 2020; 25. [PMID: 31992392 PMCID: PMC6988270 DOI: 10.2807/1560-7917.es.2020.25.3.1900765] [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] [Indexed: 01/13/2023]
Abstract
Genomic surveillance during ebolavirus outbreaks to elucidate transmission chains and develop diagnostic tests is delayed by the laborious development of variant-specific laboratory assays. We developed a new protocol combining 31 parallel PCR assays with Illumina/MinION-based sequencing, allowing generic ebolavirus genomic surveillance, validated using cell culture-derived Ebola, Reston, Sudan and Taï Forest virus at concentrations compatible with patient viral loads. Our approach enables pre-emptive genomic surveillance of ongoing and future ebolavirus outbreaks irrespective of variant divergence.
Collapse
Affiliation(s)
- Ignacio Postigo-Hidalgo
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Virology, Berlin, Germany
| | - Carlo Fischer
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Virology, Berlin, Germany
| | - Andres Moreira-Soto
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Virology, Berlin, Germany
| | - Patricia Tscheak
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Virology, Berlin, Germany
| | - Michael Nagel
- Deutsche Gesellschaft für Internationale Zusammenarbeit (GIZ) GmbH, Bonn, Germany
| | - Markus Eickmann
- Institute of Virology, Philipps-Universität Marburg, Marburg, Germany.,German Centre for Infection Research (DZIF), partner site Giessen - Marburg - Langen, Germany
| | - Jan Felix Drexler
- German Centre for Infection Research (DZIF), associated partner Charité - Universitätsmedizin Berlin, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Institute of Virology, Berlin, Germany
| |
Collapse
|
25
|
Abbate JL, Becquart P, Leroy E, Ezenwa VO, Roche B. Exposure to Ebola Virus and Risk for Infection with Malaria Parasites, Rural Gabon. Emerg Infect Dis 2020; 26:229-237. [PMID: 31829919 PMCID: PMC6986822 DOI: 10.3201/eid2602.181120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
An association between malaria and risk for death among patients with Ebola virus disease has suggested within-host interactions between Plasmodium falciparum parasites and Ebola virus. To determine whether such an interaction might also influence the probability of acquiring either infection, we used a large snapshot surveillance study from rural Gabon to test if past exposure to Ebola virus is associated with current infection with Plasmodium spp. during nonepidemic conditions. We found a strong positive association, on population and individual levels, between seropositivity for antibodies against Ebola virus and the presence of Plasmodium parasites in the blood. According to a multiple regression model accounting for other key variables, antibodies against Ebola virus emerged as the strongest individual-level risk factor for acquiring malaria. Our results suggest that within-host interactions between malaria parasites and Ebola virus may underlie epidemiologic associations.
Collapse
|
26
|
Price A, Okumura A, Haddock E, Feldmann F, Meade-White K, Sharma P, Artami M, Lipkin WI, Threadgill DW, Feldmann H, Rasmussen AL. Transcriptional Correlates of Tolerance and Lethality in Mice Predict Ebola Virus Disease Patient Outcomes. Cell Rep 2020; 30:1702-1713.e6. [PMID: 32049004 PMCID: PMC11062563 DOI: 10.1016/j.celrep.2020.01.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 11/07/2019] [Accepted: 01/07/2020] [Indexed: 01/26/2023] Open
Abstract
Host response to infection is a major determinant of disease severity in Ebola virus disease (EVD), but gene expression programs associated with outcome are poorly characterized. Collaborative Cross (CC) mice develop strain-dependent EVD phenotypes of differential severity, ranging from tolerance to lethality. We screen 10 CC lines and identify clinical, virologic, and transcriptomic features that distinguish tolerant from lethal outcomes. Tolerance is associated with tightly regulated induction of immune and inflammatory responses shortly following infection, as well as reduced inflammatory macrophages and increased antigen-presenting cells, B-1 cells, and γδ T cells. Lethal disease is characterized by suppressed early gene expression and reduced lymphocytes, followed by uncontrolled inflammatory signaling, leading to death. We apply machine learning to predict outcomes with 99% accuracy in mice using transcriptomic profiles. This signature predicts outcomes in a cohort of EVD patients from western Africa with 75% accuracy, demonstrating potential clinical utility.
Collapse
Affiliation(s)
- Adam Price
- Center for Infection and Immunity, Columbia Mailman School of Public Health, New York, NY 10032, USA
| | - Atsushi Okumura
- Center for Infection and Immunity, Columbia Mailman School of Public Health, New York, NY 10032, USA; Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Elaine Haddock
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA; Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Pryanka Sharma
- Center for Infection and Immunity, Columbia Mailman School of Public Health, New York, NY 10032, USA
| | - Methinee Artami
- Center for Infection and Immunity, Columbia Mailman School of Public Health, New York, NY 10032, USA
| | - W Ian Lipkin
- Center for Infection and Immunity, Columbia Mailman School of Public Health, New York, NY 10032, USA
| | - David W Threadgill
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA; Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Angela L Rasmussen
- Center for Infection and Immunity, Columbia Mailman School of Public Health, New York, NY 10032, USA.
| |
Collapse
|
27
|
Kozak RA, Goneau LW, DeLima C, Varsaneux O, Eshaghi A, Kristjanson E, Olsha R, Safronetz D, Perusini S, Frantz C, Gubbay JB. Presence of Flavivirus Antibodies Does Not Lead to a Greater Number of Symptoms in a Small Cohort of Canadian Travelers Infected with Zika Virus. Viruses 2020; 12:v12020140. [PMID: 31991674 PMCID: PMC7077307 DOI: 10.3390/v12020140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/12/2020] [Accepted: 01/22/2020] [Indexed: 02/04/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus associated with a febrile illness as well as severe complications, including microcephaly and Guillain-Barré Syndrome. Antibody cross-reactivity between flaviviruses has been documented, and in regions where ZIKV is circulating, dengue virus (DENV) is also endemic, leaving the potential that previous exposure to DENV could alter clinical features of ZIKV infection. To investigate this, we performed a retrospective case-control study in which we compared Canadian travellers who had been infected with ZIKV and had serological findings indicating previous DENV or other flavivirus exposure (n = 16) to those without any previous exposure (n = 44). Patient samples were collected between February 2016 and September 2017 and submitted to Public Health Ontario for testing. ZIKV infection was determined using real-time RT-PCR and antibodies against DENV were identified by the plaque-reduction neutralization test. The mean time from symptom onset to sample collection was 5 days for both groups; the magnitude of viremia was not statistically different (Ct values: 35.6 vs. 34.9, p-value = 0.2). Clinical scores were also similar. Our findings indicate that previous DENV or other flavivirus exposure did not result in greater viremia or a higher illness score.
Collapse
Affiliation(s)
- Robert A. Kozak
- Division of Microbiology, Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lee W. Goneau
- Public Health Ontario, Toronto, ON M5G 1V2, Canada; (L.W.G.); (C.D.); (A.E.); (E.K.); (R.O.); (S.P.); (C.F.)
| | - Cedric DeLima
- Public Health Ontario, Toronto, ON M5G 1V2, Canada; (L.W.G.); (C.D.); (A.E.); (E.K.); (R.O.); (S.P.); (C.F.)
| | | | - AliReza Eshaghi
- Public Health Ontario, Toronto, ON M5G 1V2, Canada; (L.W.G.); (C.D.); (A.E.); (E.K.); (R.O.); (S.P.); (C.F.)
| | - Erik Kristjanson
- Public Health Ontario, Toronto, ON M5G 1V2, Canada; (L.W.G.); (C.D.); (A.E.); (E.K.); (R.O.); (S.P.); (C.F.)
| | - Romy Olsha
- Public Health Ontario, Toronto, ON M5G 1V2, Canada; (L.W.G.); (C.D.); (A.E.); (E.K.); (R.O.); (S.P.); (C.F.)
| | - David Safronetz
- Special Pathogens Program, National Microbiology Laboratory, Winnipeg, MB R3E 3R2, Canada;
| | - Stephen Perusini
- Public Health Ontario, Toronto, ON M5G 1V2, Canada; (L.W.G.); (C.D.); (A.E.); (E.K.); (R.O.); (S.P.); (C.F.)
| | - Christine Frantz
- Public Health Ontario, Toronto, ON M5G 1V2, Canada; (L.W.G.); (C.D.); (A.E.); (E.K.); (R.O.); (S.P.); (C.F.)
| | - Jonathan B. Gubbay
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Public Health Ontario, Toronto, ON M5G 1V2, Canada; (L.W.G.); (C.D.); (A.E.); (E.K.); (R.O.); (S.P.); (C.F.)
- Hospital for Sick Children, Toronto, ON M5G 1V2, Canada
- Correspondence: ; Tel.: +1-647-792-3170
| |
Collapse
|
28
|
Couturier C, Wada A, Louis K, Mistretta M, Beitz B, Povogui M, Ripaux M, Mignon C, Werle B, Lugari A, Pannetier D, Godard S, Bocquin A, Mely S, Béavogui I, Hébélamou J, Leuenberger D, Leissner P, Yamamoto T, Lécine P, Védrine C, Chaix J. Characterization and analytical validation of a new antigenic rapid diagnostic test for Ebola virus disease detection. PLoS Negl Trop Dis 2020; 14:e0007965. [PMID: 31951615 PMCID: PMC6992227 DOI: 10.1371/journal.pntd.0007965] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 01/30/2020] [Accepted: 11/27/2019] [Indexed: 01/16/2023] Open
Abstract
Hemorrhagic fever outbreaks are difficult to diagnose and control in part because of a lack of low-cost and easily accessible diagnostic structures in countries where etiologic agents are present. Furthermore, initial clinical symptoms are common and shared with other endemic diseases such as malaria or typhoid fever. Current molecular diagnostic methods such as polymerase chain reaction require trained personnel and laboratory infrastructure, hindering diagnostics at the point of need, particularly in outbreak settings. Therefore, rapid diagnostic tests such as lateral flow can be broadly deployed and are typically well-suited to rapidly diagnose hemorrhagic fever viruses, such as Ebola virus. Early detection and control of Ebola outbreaks require simple, easy-to-use assays that can detect very low amount of virus in blood. Here, we developed and characterized an immunoassay test based on immunochromatography coupled to silver amplification technology to detect the secreted glycoprotein of EBOV. The glycoprotein is among the first viral proteins to be detected in blood. This strategy aims at identifying infected patients early following onset of symptoms by detecting low amount of sGP protein in blood samples. The limit of detection achieved by this sGP-targeted kit is 2.2 x 104 genome copies/ml in plasma as assayed in a monkey analytical cohort. Clinical performance evaluation showed a specificity of 100% and a sensitivity of 85.7% when evaluated with plasma samples from healthy controls and patients infected with Zaire Ebola virus from Macenta, Guinea. This rapid and accurate diagnostic test could therefore be used in endemic countries for early detection of infected individuals in point of care settings. Moreover, it could also support efficient clinical triage in hospitals or clinical centers and thus reducing transmission rates to prevent and better manage future severe outbreaks. Ebola virus disease is a severe disease caused by Ebola virus, a member of the filovirus family, which occurs in humans and other primates. Ebola is believed to be zoonotic, however the natural reservoir is unknown. Overlapping symptoms with other endemic diseases, such as malaria and cholera, make accurate diagnostic challenging. Outbreaks of Ebola have been widespread as the consequence of the absence of available rapid, sensitive, specific, robust, and affordable licensed diagnostic test in remote areas, where outbreaks usually start. Here we have established and validated a rapid diagnostic test, which is fast, sensitive, specific, efficient, affordable, and user-friendly. Its analytical characteristics make it suitable for clinical management during Ebola virus outbreaks in remote areas. Of interest, this rapid diagnostic test detects the presence of an early viral antigen, the secreted glycoprotein, found in blood of patients shortly after infection, suggesting that it could be used to identify infected patients shortly after onset of symptoms.
Collapse
Affiliation(s)
| | - Atsuhiko Wada
- FUJIFILM, Ushijima, Kaisei-machi, Ashigarakami-gun Kanagawa, Japan
| | | | | | | | - Moriba Povogui
- Centre de Recherche Et de Formation en Infectiologie de Guinée (CERFIG), République de Guinée
| | | | | | | | | | | | | | - Anne Bocquin
- INSERM Jean Mérieux BSL4 Laboratory, LYON, France
| | | | - Ismaël Béavogui
- CHRS Macenta, c/o Mission Philafricaine, Conakry, République de Guinée
| | - Jean Hébélamou
- CHRS Macenta, c/o Mission Philafricaine, Conakry, République de Guinée
| | - David Leuenberger
- CHRS Macenta, c/o Mission Philafricaine, Conakry, République de Guinée
| | | | - Takeshi Yamamoto
- FUJIFILM, Ushijima, Kaisei-machi, Ashigarakami-gun Kanagawa, Japan
| | | | | | | |
Collapse
|
29
|
Scott JT, Sharma R, Meredith LW, Dunning J, Moore CE, Sahr F, Ward S, Goodfellow I, Horby P. Pharmacokinetics of TKM-130803 in Sierra Leonean patients with Ebola virus disease: plasma concentrations exceed target levels, with drug accumulation in the most severe patients. EBioMedicine 2020; 52:102601. [PMID: 31953031 PMCID: PMC7033524 DOI: 10.1016/j.ebiom.2019.102601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/22/2022] Open
Abstract
Background TKM-130803 is a specific anti-EBOV therapeutic comprised of two small interfering RNAs (siRNA) siLpol-2 and siVP35-2. The pharmacokinetics (PK) of these siRNAs was defined in Ebola virus disease (EVD) patients, with reference to efficacy (ET) and toxicology thresholds (TT). The relationship between PK and patient survival was explored. Methods Pharmacokinetic (PK) and pharmacodynamic (PD) data were available for seven participants with EVD in Sierra Leone who received 0·3 mg/kg of TKM-130803 by intravenous infusion over 2 h daily for up to 7 days. Plasma concentration of siRNA was compared to survival at 14 days. PK data were fitted to two-compartment models then Monte Carlo simulated PK profiles were compared to ET (Cmax 0·04–0·57 ng/mL and mean concentration 1·43 ng/mL), and TT (3000 ng/mL). Findings Viral loads (VL) were not significantly different at treatment onset or during treatment (p = 0·1) in subjects who survived or died. siRNA was in quantitative excess of virus genomes throughout treatment, but the 95% percentile exceeded TT. The maximum AUC for which the 95% percentile remained under TT was a continuous infusion of 0·15 mg/kg/day. Plasma concentration of both siRNAs were higher in subjects who died compared to subjects who survived (p<0·025 both siRNAs). Interpretation TKM-130803 was circulating in molar excess of circulating virus; a level considered needed for efficacy. Given extremely high viral loads it seems likely that the patients died because they were physiologically beyond the point of no return. Subjects who died exhibited some indication of impaired drug clearance, justifying caution in dosing strategies for such patients. This analysis has given a useful insight into the pharmacokinetics of the siRNA in the disease state and illustrates the value of designing PKPD studies into future clinical trials in epidemic situations. Funding This work was supported by the Wellcome Trust of Great Britain (grant number 106491/Z/14/Z and 097997/Z/11/A) and by the EU FP7 project PREPARE (602525). The PHE laboratory was funded by the UK Department for International Development. The funders had no role in trial design, data collection or analysis. The views expressed are those of the authors and not necessarily those of Public Health England, the Department of Health, or the EU. Trial registration Pan African Clinical Trials Registry PACTR201501000997429.
Collapse
Affiliation(s)
- Janet T Scott
- MRC-University of Glasgow Centre for Virus Research, G61 1QH, UK; NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, UK.
| | - Raman Sharma
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Luke W Meredith
- Department of Pathology, Division of Virology, University of Cambridge, Cambridge CB2 1QP, UK; Department of Public Health, University of Makeni, Makeni, Sierra Leone
| | - Jake Dunning
- National Infection Service, Public Health England, London, UK; Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Catrin E Moore
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Foday Sahr
- Republic Sierra Leone Armed Forces, Military Hosp 34, Freetown, Sierra Leone
| | - Steve Ward
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - Ian Goodfellow
- Department of Pathology, Division of Virology, University of Cambridge, Cambridge CB2 1QP, UK; Department of Public Health, University of Makeni, Makeni, Sierra Leone
| | - Peter Horby
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | -
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| |
Collapse
|
30
|
Junaid A, Tang H, van Reeuwijk A, Abouleila Y, Wuelfroth P, van Duinen V, Stam W, van Zonneveld AJ, Hankemeier T, Mashaghi A. Ebola Hemorrhagic Shock Syndrome-on-a-Chip. iScience 2019; 23:100765. [PMID: 31887664 PMCID: PMC6941864 DOI: 10.1016/j.isci.2019.100765] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/26/2019] [Accepted: 12/09/2019] [Indexed: 01/12/2023] Open
Abstract
Ebola virus, for which we lack effective countermeasures, causes hemorrhagic fever in humans, with significant case fatality rates. Lack of experimental human models for Ebola hemorrhagic fever is a major obstacle that hinders the development of treatment strategies. Here, we model the Ebola hemorrhagic syndrome in a microvessel-on-a-chip system and demonstrate its applicability to drug studies. Luminal infusion of Ebola virus-like particles leads to albumin leakage from the engineered vessels. The process is mediated by the Rho/ROCK pathway and is associated with cytoskeleton remodeling. Infusion of Ebola glycoprotein (GP1,2) generates a similar phenotype, indicating the key role of GP1,2 in this process. Finally, we measured the potency of a recently developed experimental drug FX06 and a novel drug candidate, melatonin, in phenotypic rescue. Our study confirms the effects of FX06 and identifies melatonin as an effective, safe, inexpensive therapeutic option that is worth investigating in animal models and human trials.
Collapse
Affiliation(s)
- Abidemi Junaid
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333 CC, Netherlands; Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden 2333 ZA, Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden 2333 ZA, Netherlands
| | - Huaqi Tang
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333 CC, Netherlands
| | - Anne van Reeuwijk
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333 CC, Netherlands
| | - Yasmine Abouleila
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333 CC, Netherlands
| | | | - Vincent van Duinen
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333 CC, Netherlands; Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden 2333 ZA, Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden 2333 ZA, Netherlands
| | - Wendy Stam
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden 2333 ZA, Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden 2333 ZA, Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden 2333 ZA, Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden 2333 ZA, Netherlands
| | - Thomas Hankemeier
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333 CC, Netherlands
| | - Alireza Mashaghi
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2333 CC, Netherlands.
| |
Collapse
|
31
|
Ebola virus disease: An emerging and re-emerging viral threat. J Autoimmun 2019; 106:102375. [PMID: 31806422 DOI: 10.1016/j.jaut.2019.102375] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 12/21/2022]
Abstract
The genus Ebolavirus from the family Filoviridae is composed of five species including Sudan ebolavirus, Reston ebolavirus, Bundibugyo ebolavirus, Taï Forest ebolavirus, and Ebola virus (previously known as Zaire ebolavirus). These viruses have a large non-segmented, negative-strand RNA of approximately 19 kb that encodes for glycoproteins (i.e., GP, sGP, ssGP), nucleoproteins, virion proteins (i.e., VP 24, 30,40) and an RNA dependent RNA polymerase. These viruses have become a global health concern because of mortality, their rapid dissemination, new outbreaks in West-Africa, and the emergence of a new condition known as "Post-Ebola virus disease syndrome" that resembles inflammatory and autoimmune conditions such as rheumatoid arthritis, systemic lupus erythematosus and spondyloarthritis with uveitis. However, there are many gaps in the understanding of the mechanisms that may induce the development of such autoimmune-like syndromes. Some of these mechanisms may include a high formation of neutrophil extracellular traps, an uncontrolled "cytokine storm", and the possible formation of auto-antibodies. The likely appearance of autoimmune phenomena in Ebola survivors suppose a new challenge in the management and control of this disease and opens a new field of research in a special subgroup of patients. Herein, the molecular biology, pathogenesis, clinical manifestations, and treatment of Ebola virus disease are reviewed and some strategies for control of disease are discussed.
Collapse
|
32
|
Rojek AM, Salam A, Ragotte RJ, Liddiard E, Elhussain A, Carlqvist A, Butler M, Kayem N, Castle L, Odondi L', Stepniewska K, Horby PW. A systematic review and meta-analysis of patient data from the West Africa (2013-16) Ebola virus disease epidemic. Clin Microbiol Infect 2019; 25:1307-1314. [PMID: 31284032 PMCID: PMC7116468 DOI: 10.1016/j.cmi.2019.06.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/25/2019] [Accepted: 06/28/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Over 28 000 individuals were infected with Ebola virus during the West Africa (2013-2016) epidemic, yet there has been criticism of the lack of robust clinical descriptions of Ebola virus disease (EVD) illness from that outbreak. OBJECTIVES To perform a meta-analysis of published data from the epidemic to describe the clinical presentation, evolution of disease, and predictors of mortality in individuals with EVD. To assess the quality and utility of published data for clinical and public health decision-making. DATA SOURCES Primary articles available in PubMed and published between January 2014 and May 2017. ELIGIBILITY Studies that sequentially enrolled individuals hospitalized for EVD and that reported acute clinical outcomes. METHODS We performed meta-analyses using random-effect models and assessed heterogeneity using the I2 method. We assessed data representativeness by comparing meta-analysis estimates with WHO aggregate data. We examined data utility by examining the availability and compatibility of data sets. RESULTS In all, 3653 articles were screened and 34 articles were included, representing 16 independent cohorts of patients (18 overlapping cohorts) and at least 6168 individuals. The pooled estimate for case fatality rate was 51% (95% CI 46%-56%). However, pooling of estimates for clinical presentation, progression, and predictors of mortality in individuals with EVD were hampered by significant heterogeneity, and inadequate data on clinical progression. Our assessment of data quality found that heterogeneity was largely unexplained, and data availability and compatibility were poor. CONCLUSIONS We have quantified a missed opportunity to generate reliable estimates of the clinical manifestations of EVD during the West Africa epidemic. Clinical data standards and data capture platforms are urgently needed.
Collapse
Affiliation(s)
- A M Rojek
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK.
| | - A Salam
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK; United Kingdom Public Health Rapid Support Team, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - R J Ragotte
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - E Liddiard
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - A Elhussain
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - A Carlqvist
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - M Butler
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - N Kayem
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - L Castle
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - L 'o Odondi
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| | - K Stepniewska
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK; WorldWide Antimalarial Resistance Network, Oxford, UK
| | - P W Horby
- Epidemic Diseases Research Group, University of Oxford, Oxford, UK
| |
Collapse
|
33
|
Escudero-Pérez B, Ruibal P, Rottstegge M, Lüdtke A, Port JR, Hartmann K, Gómez-Medina S, Müller-Guhl J, Nelson EV, Krasemann S, Rodríguez E, Muñoz-Fontela C. Comparative pathogenesis of Ebola virus and Reston virus infection in humanized mice. JCI Insight 2019; 4:126070. [PMID: 31550241 PMCID: PMC6948759 DOI: 10.1172/jci.insight.126070] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 09/19/2019] [Indexed: 01/14/2023] Open
Abstract
Filoviruses of the genus Ebolavirus include 6 species with marked differences in their ability to cause disease in humans. From the highly virulent Ebola virus to the seemingly nonpathogenic Reston virus, case fatality rates can range between 0% and 90%. In order to understand the molecular basis of these differences, it is imperative to establish disease models that recapitulate human disease as faithfully as possible. Nonhuman primates (NHPs) are the gold-standard models for filovirus pathogenesis, but comparative studies are skewed by the fact that Reston virus infection can be lethal for NHPs. Here we used HLA-A2-transgenic, NOD-scid-IL-2γ receptor-knockout (NSG-A2) mice reconstituted with human hematopoiesis to compare Ebola virus and Reston virus pathogenesis in a human-like environment. While markedly less pathogenic than Ebola virus, Reston virus killed 20% of infected mice, a finding that was linked to exacerbated inflammation and viral replication in the liver. In addition, the case fatality ratios of different Ebolavirus species in humans were recapitulated in the humanized mice. Our findings point to humanized mice as a putative model to test the pathogenicity of newly discovered filoviruses, and suggest that further investigations on Reston virus pathogenesis in humans are warranted.
Collapse
Affiliation(s)
- Beatriz Escudero-Pérez
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg, Hamburg, Germany
| | - Paula Ruibal
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Monika Rottstegge
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg, Hamburg, Germany
| | - Anja Lüdtke
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Julia R Port
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg, Hamburg, Germany
| | - Kristin Hartmann
- Institute for Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sergio Gómez-Medina
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg, Hamburg, Germany
| | - Jürgen Müller-Guhl
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Emily V Nelson
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg, Hamburg, Germany
| | - Susanne Krasemann
- Institute for Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Estefanía Rodríguez
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg, Hamburg, Germany
| |
Collapse
|
34
|
Speranza E, Ruibal P, Port JR, Feng F, Burkhardt L, Grundhoff A, Günther S, Oestereich L, Hiscox JA, Connor JH, Muñoz-Fontela C. T-Cell Receptor Diversity and the Control of T-Cell Homeostasis Mark Ebola Virus Disease Survival in Humans. J Infect Dis 2019; 218:S508-S518. [PMID: 29986035 DOI: 10.1093/infdis/jiy352] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Differences in T-cell phenotype, particularly the expression of markers of T-cell homeostasis, have been observed in fatal and nonfatal Ebola virus disease (EVD). However, the relationship between these markers with T-cell function and virus clearance during EVD is poorly understood. To gain biological insight into the role of T cells during EVD, combined transcriptomics and T-cell receptor sequencing was used to profile blood samples from fatal and nonfatal EVD patients from the recent West African EVD epidemic. Fatal EVD was characterized by strong T-cell activation and increased abundance of T-cell inhibitory molecules. However, the early T-cell response was oligoclonal and did not result in viral clearance. In contrast, survivors mounted highly diverse T-cell responses, maintained low levels of T-cell inhibitors, and cleared Ebola virus. Our findings highlight the importance of T-cell immunity in surviving EVD and strengthen the foundation for further research on targeting of the dendritic cell-T cell interface for postexposure immunotherapy.
Collapse
Affiliation(s)
- Emily Speranza
- Department of Microbiology, Boston University School of Medicine, Boston MA.,Department of Bioinformatics Program, Boston University, Boston MA.,Department of National Emerging Infectious Diseases Laboratories, Boston University, Boston MA.,Department of Mathematics and Statistics, Boston University, Boston MA
| | - Paula Ruibal
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Julia R Port
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner SiteHamburg, Germany
| | - Feng Feng
- Department of Microbiology, Boston University School of Medicine, Boston MA.,Department of Mathematics and Statistics, Boston University, Boston MA
| | - Lia Burkhardt
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Adam Grundhoff
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Stephan Günther
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner SiteHamburg, Germany
| | - Lisa Oestereich
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner SiteHamburg, Germany
| | - Julian A Hiscox
- Institute for Infection and Global Health, University of Liverpool, United Kingdom.,Singapore Immunology Network, A*STAR, Singapore
| | - John H Connor
- Department of Microbiology, Boston University School of Medicine, Boston MA.,Department of Bioinformatics Program, Boston University, Boston MA.,Department of National Emerging Infectious Diseases Laboratories, Boston University, Boston MA.,Department of Mathematics and Statistics, Boston University, Boston MA
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner SiteHamburg, Germany
| |
Collapse
|
35
|
Reisler RB, Kraft CS, Bavari S, Cardile AP. Clinical Laboratory Values in Human Ebola Virus Disease Support the Relevance of the Intramuscular Ebola-Kikwit Rhesus Model. Clin Infect Dis 2019; 66:1479-1480. [PMID: 29272400 DOI: 10.1093/cid/cix1025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Ronald B Reisler
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland
| | - Colleen S Kraft
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland
| | - Anthony P Cardile
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland
| |
Collapse
|
36
|
Reusken CB, Mögling R, Smit PW, Grunow R, Ippolito G, Di Caro A, Koopmans M. Status, quality and specific needs of Ebola virus diagnostic capacity and capability in laboratories of the two European preparedness laboratory networks EMERGE and EVD-LabNet. ACTA ACUST UNITED AC 2019; 23. [PMID: 29766839 PMCID: PMC5954606 DOI: 10.2807/1560-7917.es.2018.23.19.17-00404] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
From December 2013 to March 2016, West Africa experienced the largest Ebola virus (EBOV) outbreak to date, leading to a European-wide activation of laboratory preparedness and response. At the end of the outbreak, laboratories associated with the two European preparedness networks of expert laboratories EMERGE JA and EVD-LabNet were invited to participate in an assessment of the response of European laboratories to the EBOV outbreak, to identify learning points and training needs to strengthen future outbreak responses. Response aspects assessed included diagnostics, biorisk management and quality assurance. The overall coverage of EBOV diagnostics in the European Union/European Economic Area (EU/EEA) was found to be adequate although some points for quality improvement were identified. These included the need for relevant International Organization for Standardization (ISO) accreditation, the provision of EBOV external quality assessments (EQA) in periods where there is no emergency, facilitating access to controls and knowledge, biorisk management without compromising biosafety and a rapid public health response, and the need for both sustained and contingency funding for preparedness and response activities.
Collapse
Affiliation(s)
- Chantal B Reusken
- Department of Viroscience, World Health Organization Collaborating Centre for Arbovirus and Viral Haemorrhagic Fever Reference and Research, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Ramona Mögling
- Department of Viroscience, World Health Organization Collaborating Centre for Arbovirus and Viral Haemorrhagic Fever Reference and Research, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Pieter W Smit
- Department of Viroscience, World Health Organization Collaborating Centre for Arbovirus and Viral Haemorrhagic Fever Reference and Research, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | | | - Giuseppe Ippolito
- National Institute for Infectious Diseases (INMI) Lazzaro Spallanzani, Rome, Italy
| | - Antonino Di Caro
- National Institute for Infectious Diseases (INMI) Lazzaro Spallanzani, Rome, Italy
| | - Marion Koopmans
- Department of Viroscience, World Health Organization Collaborating Centre for Arbovirus and Viral Haemorrhagic Fever Reference and Research, Erasmus University Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
37
|
Abstract
The process of entry into a host cell is a key step in the life cycle of most viruses. In recent years, there has been a significant increase in our understanding of the routes and mechanisms of entry for a number of these viruses. This has led to the development of novel broad-spectrum antiviral approaches that target host cell proteins and pathways, in addition to strategies focused on individual viruses or virus families. Here we consider a number of these approaches and their broad-spectrum potential.
Collapse
Affiliation(s)
- Michela Mazzon
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Mark Marsh
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| |
Collapse
|
38
|
Brown JF, Dye JM, Tozay S, Jeh-Mulbah G, Wohl DA, Fischer WA, Cunningham CK, Rowe K, Zacharias P, van Hasselt J, Norwood DA, Thielman NM, Zak SE, Hoover DL. Anti-Ebola Virus Antibody Levels in Convalescent Plasma and Viral Load After Plasma Infusion in Patients With Ebola Virus Disease. J Infect Dis 2019; 218:555-562. [PMID: 29659889 DOI: 10.1093/infdis/jiy199] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/03/2018] [Indexed: 11/14/2022] Open
Abstract
Background Ebola virus (EBOV) neutralizing antibody in plasma may reduce viral load following administration of plasma to patients with Ebola virus disease (EVD), but measurement of these antibodies is complex. Methods Anti-EBOV antibody was measured by 2 neutralization and 2 enzyme-linked immunosorbent assays (ELISAs) in convalescent plasma (ECP) from 100 EVD survivor donors in Liberia. Viral load was assessed repetitively in patients with EVD participating in a clinical trial of enhanced standard of care plus ECP. Results All 4 anti-EBOV assays were highly concordant for detection of EBOV antibody. Antibodies were not detected in plasma specimens obtained from 15 of 100 donors, including 7 with documented EBOV-positive reverse-transcription polymerase chain reaction during EVD. Viral load was reduced following each dose in the 2 clinical trial participants who received ECP with higher antibody levels but not in the 2 who received ECP with lower antibody levels. Conclusions Recovery from EVD can occur with absence of detectable anti-EBOV antibody several months after disease onset. ELISAs may be useful to select ECP donors or identify ECP units that contain neutralizing antibody. ECP with higher anti-EBOV antibody levels may have greater effect on EBOV load-an observation that requires further investigation. Clinical Trials Registration NCT02333578.
Collapse
Affiliation(s)
- Jerry F Brown
- Eternal Love Winning Africa Hospital, Paynesville, Liberia
| | - John M Dye
- US Army Medical Research Institute of Infectious Disease, Frederick, Maryland
| | - Sam Tozay
- Eternal Love Winning Africa Hospital, Paynesville, Liberia
| | | | - David A Wohl
- University of North Carolina School of Medicine, Chapel Hill
| | | | | | - Kathleen Rowe
- Blood Centers of America, West Warwick, Rhode Island
| | - Peter Zacharias
- Safe Blood for Africa Foundation, Washington, District of Columbia, Hinckley, Ohio
| | - James van Hasselt
- Safe Blood for Africa Foundation, Washington, District of Columbia, Hinckley, Ohio
| | - David A Norwood
- US Army Medical Research Institute of Infectious Disease, Frederick, Maryland
| | | | - Samantha E Zak
- US Army Medical Research Institute of Infectious Disease, Frederick, Maryland
| | | |
Collapse
|
39
|
Racine T, Kobinger GP. Challenges and perspectives on the use of mobile laboratories during outbreaks and their use for vaccine evaluation. Hum Vaccin Immunother 2019; 15:2264-2268. [PMID: 30893007 PMCID: PMC6816390 DOI: 10.1080/21645515.2019.1597595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Mobile laboratories provide diagnostic capabilities for routine surveillance and patient identification during an outbreak. In either situation, they face many challenges including identification of the appropriate assay(s) to employ, logistical arrangements, and providing for the health and safety of the laboratory staff. Great strides have been made over the last decade in the development of mobile laboratories with assays that require minimal infrastructure and technical experience. This knowledge and expertise have been developed in partnership with many researchers and public health officials who live in regions prone to infectious disease outbreaks. Mobile laboratories should now also be used in the evaluation of novel vaccines and therapeutics in remote locations. Clinical mobile laboratories will include similar diagnostic capabilities as outbreak response mobile labs, but will also include additional point-of-care instruments operated under Good Clinical Practice guidelines. They will also operate rigorous data management plans so that the data collected will satisfy regulatory agencies during the licensure process. Failure to deploy an adequate clinical mobile laboratory when administering a novel biological product in a remote location is a significant limitation to any collected scientific data that could ultimately undermine clinical development and availability of life-saving interventions.
Collapse
Affiliation(s)
- Trina Racine
- Department of Medical Microbiology, University of Manitoba , Winnipeg , Manitoba , Canada.,Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval , Québec City , Québec , Canada
| | - Gary P Kobinger
- Department of Medical Microbiology, University of Manitoba , Winnipeg , Manitoba , Canada.,Centre de Recherche en Infectiologie, Centre Hospitalier Universitaire de Québec, Université Laval , Québec City , Québec , Canada.,Department of Immunology, University of Manitoba , Winnipeg , Manitoba , Canada.,Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine , Philadelphia , PA , USA
| |
Collapse
|
40
|
Nsio J, Kapetshi J, Makiala S, Raymond F, Tshapenda G, Boucher N, Corbeil J, Okitandjate A, Mbuyi G, Kiyele M, Mondonge V, Kikoo MJ, Van Herp M, Barboza P, Petrucci R, Benedetti G, Formenty P, Muyembe Muzinga B, Ilunga Kalenga O, Ahuka S, Fausther-Bovendo H, Ilunga BK, Kobinger GP, Muyembe JJT. 2017 Outbreak of Ebola Virus Disease in Northern Democratic Republic of Congo. J Infect Dis 2019; 221:701-706. [DOI: 10.1093/infdis/jiz107] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 03/22/2019] [Indexed: 11/12/2022] Open
Affiliation(s)
- Justus Nsio
- Ministry of Health, Kinshasa, Democratic Republic of the Congo
| | - Jimmy Kapetshi
- Institut National de Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo
| | - Sheila Makiala
- Institut National de Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo
| | | | | | | | | | | | - Gisele Mbuyi
- Ministry of Health, Kinshasa, Democratic Republic of the Congo
| | - Musa Kiyele
- Ministry of Health, Kinshasa, Democratic Republic of the Congo
| | - Vital Mondonge
- World Health Organization (WHO), Kinshasa, Democratic Republic of the Congo
| | - Marie Jose Kikoo
- World Health Organization (WHO), Kinshasa, Democratic Republic of the Congo
| | | | | | | | - Guido Benedetti
- Medical Department, Operational Centre–Brussels, MSF, Luxembourg, Luxembourg
| | | | - Baby Muyembe Muzinga
- Institut National de Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo
| | | | - Steve Ahuka
- Institut National de Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo
| | | | | | | | | |
Collapse
|
41
|
Wonderly B, Jones S, Gatton ML, Barber J, Killip M, Hudson C, Carter L, Brooks T, Simpson AJH, Semper A, Urassa W, Chua A, Perkins M, Boehme C. Comparative performance of four rapid Ebola antigen-detection lateral flow immunoassays during the 2014-2016 Ebola epidemic in West Africa. PLoS One 2019; 14:e0212113. [PMID: 30845203 PMCID: PMC6405069 DOI: 10.1371/journal.pone.0212113] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 01/28/2019] [Indexed: 11/23/2022] Open
Abstract
Background Without an effective vaccine, as was the case early in the 2014–2016 Ebola Outbreak in West Africa, disease control depends entirely on interrupting transmission through early disease detection and prompt patient isolation. Lateral Flow Immunoassays (LFI) are a potential supplement to centralized reference laboratory testing for the early diagnosis of Ebola Virus Disease (EVD). The goal of this study was to assess the performance of commercially available simple and rapid antigen detection LFIs, submitted for review to the WHO via the Emergency Use Assessment and Listing procedure. The study was performed in an Ebola Treatment Centre laboratory involved in EVD testing in Sierra Leone. In light of the current Ebola outbreak in May 2018 in the Democratic Republic of Congo, which highlights the lack of clarity in the global health community about appropriate Ebola diagnostics, our findings are increasingly critical. Methods A cross-sectional study was conducted to assess comparative performance of four LFIs for detecting EVD. LFIs were assessed against the same 328 plasma samples and 100 whole EDTA blood samples, using the altona RealStar Filovirus Screen real-time RT-PCR as the bench mark assay. The performance of the Public Health England (PHE) in-house Zaire ebolavirus-specific real time RT-PCR Trombley assay was concurrently assessed. Statistical analysis using generalized estimating equations was conducted to compare LFI performance. Findings Sensitivity and specificity varied between the LFIs, with specificity found to be significantly higher for whole EDTA blood samples compared to plasma samples in at least 2 LFIs (P≤0.003). Using the altona RT-PCR assay as the bench mark, sensitivities on plasma samples ranged from 79.53% (101/127, 95% CI: 71.46–86.17%) for the DEDIATEST EBOLA (SD Biosensor) to 98.43% (125/127, 95% CI: 94.43–99.81%) for the One step Ebola test (Intec). Specificities ranged from 80.20% (158/197, 95% CI: 74.07–88.60%) for plasma samples using the ReEBOV Antigen test Kit (Corgenix) to 100.00% (98/98, 95% CI: 96.31–100.00%) for whole blood samples using the DEDIATEST EBOLA (SD Biosensor) and SD Ebola Zaire Ag (SD Biosensor). Results also showed the Trombley RT-PCR assay had a lower limit of detection than the altona assay, with some LFIs having higher sensitivity than the altona assay when the Trombley assay was the bench mark. Interpretation All of the tested EVD LFIs may be considered suitable for use in an outbreak situation (i.e. rule out testing in communities), although they had variable performance characteristics, with none possessing both high sensitivity and specificity. The non-commercial Trombley Zaire ebolavirus RT-PCR assay warrants further investigation, as it appeared more sensitive than the current gold standard, the altona Filovirus Screen RT-PCR assay.
Collapse
Affiliation(s)
- Betsy Wonderly
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Sophie Jones
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Michelle L Gatton
- School of Public Health and Social Work, Queensland University of Technology, Brisbane, Australia
| | - John Barber
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Marian Killip
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Chris Hudson
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Lisa Carter
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Tim Brooks
- Public Health England, Porton Down, United Kingdom
| | | | | | | | - Arlene Chua
- Médecins Sans Frontières International, Geneva, Switzerland
| | - Mark Perkins
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Catharina Boehme
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| |
Collapse
|
42
|
Malvy D, McElroy AK, de Clerck H, Günther S, van Griensven J. Ebola virus disease. Lancet 2019; 393:936-948. [PMID: 30777297 DOI: 10.1016/s0140-6736(18)33132-5] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 11/12/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022]
Abstract
Ebolaviruses are pathogenic agents associated with a severe, potentially fatal, systemic disease in man and great apes. Four species of ebolaviruses have been identified in west or equatorial Africa. Once the more virulent forms enter the human population, transmission occurs primarily through contact with infected body fluids and can result in major epidemics in under-resourced settings. These viruses cause a disease characterised by systemic viral replication, immune suppression, abnormal inflammatory responses, major fluid and electrolyte losses, and high mortality. Despite recent progress on vaccines, and with no licensed prophylaxis or treatment available, case management is essentially supportive with management of severe multiple organ failure resulting from immune-mediated cell damage. The 2013-16 outbreak was classified by WHO as a Public Health Emergency of International Concern, which drew attention to the challenges of diseases caused by infections with ebolaviruses and questioned scientific, clinical, and societal preparation to handle future epidemics.
Collapse
Affiliation(s)
- Denis Malvy
- Department for Infectious and Tropical Diseases, University Hospital Centre of Bordeaux, Bordeaux, France; INSERM 1219, University of Bordeaux, Bordeaux, France.
| | - Anita K McElroy
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Stephan Günther
- Department of Virology, Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | | |
Collapse
|
43
|
Role of Type I Interferons on Filovirus Pathogenesis. Vaccines (Basel) 2019; 7:vaccines7010022. [PMID: 30791589 PMCID: PMC6466283 DOI: 10.3390/vaccines7010022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/06/2019] [Accepted: 02/15/2019] [Indexed: 01/19/2023] Open
Abstract
Filoviruses, such as Ebola and Marburg virus, encode viral proteins with the ability to counteract the type I interferon (IFN-I) response. These IFN-I antagonist proteins are crucial to ensure virus replication, prevent an antiviral state in infected and bystander cells, and impair the ability of antigen-presenting cells to initiate adaptive immune responses. However, in recent years, a number of studies have underscored the conflicting data between in vitro studies and in vivo data obtained in animal models and clinical studies during outbreaks. This review aims to summarize these data and to discuss the relative contributions of IFN-α and IFN-β to filovirus pathogenesis in animal models and humans. Finally, we evaluate the putative utilization of IFN-I in post-exposure therapy and its implications as a biomarker of vaccine efficacy.
Collapse
|
44
|
Mazzon M, Ortega-Prieto AM, Imrie D, Luft C, Hess L, Czieso S, Grove J, Skelton JK, Farleigh L, Bugert JJ, Wright E, Temperton N, Angell R, Oxenford S, Jacobs M, Ketteler R, Dorner M, Marsh M. Identification of Broad-Spectrum Antiviral Compounds by Targeting Viral Entry. Viruses 2019; 11:E176. [PMID: 30791609 PMCID: PMC6410080 DOI: 10.3390/v11020176] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/13/2019] [Accepted: 02/18/2019] [Indexed: 12/22/2022] Open
Abstract
Viruses are a major threat to human health and economic well-being. In recent years Ebola, Zika, influenza, and chikungunya virus epidemics have raised awareness that infections can spread rapidly before vaccines or specific antagonists can be made available. Broad-spectrum antivirals are drugs with the potential to inhibit infection by viruses from different groups or families, which may be deployed during outbreaks when specific diagnostics, vaccines or directly acting antivirals are not available. While pathogen-directed approaches are generally effective against a few closely related viruses, targeting cellular pathways used by multiple viral agents can have broad-spectrum efficacy. Virus entry, particularly clathrin-mediated endocytosis, constitutes an attractive target as it is used by many viruses. Using a phenotypic screening strategy where the inhibitory activity of small molecules was sequentially tested against different viruses, we identified 12 compounds with broad-spectrum activity, and found a subset blocking viral internalisation and/or fusion. Importantly, we show that compounds identified with this approach can reduce viral replication in a mouse model of Zika infection. This work provides proof of concept that it is possible to identify broad-spectrum inhibitors by iterative phenotypic screenings, and that inhibition of host-pathways critical for viral life cycles can be an effective antiviral strategy.
Collapse
Affiliation(s)
- Michela Mazzon
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Ana Maria Ortega-Prieto
- Section of Virology, Department of Medicine, School of Medicine, Imperial College London, London W2 1PG, UK.
| | - Douglas Imrie
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Christin Luft
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Lena Hess
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Stephanie Czieso
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Joe Grove
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London NW3 2QG, UK.
| | - Jessica Katy Skelton
- Section of Virology, Department of Medicine, School of Medicine, Imperial College London, London W2 1PG, UK.
| | - Laura Farleigh
- Medical Microbiology, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - Joachim J Bugert
- Medical Microbiology, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
- Bundeswehr Institute of Microbiology, 80937 Munich, Germany.
| | | | - Nigel Temperton
- Medway School of Pharmacy, University of Kent, Chatham ME4 4TB, UK.
| | - Richard Angell
- School of Pharmacy, University College London, London WC1N 1AX, UK.
| | - Sally Oxenford
- School of Pharmacy, University College London, London WC1N 1AX, UK.
| | - Michael Jacobs
- Faculty of Medical Sciences, UCL Medical School, London NW3 2QG, UK.
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| | - Marcus Dorner
- Section of Virology, Department of Medicine, School of Medicine, Imperial College London, London W2 1PG, UK.
| | - Mark Marsh
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
45
|
Intra-host Ebola viral adaption during human infection. BIOSAFETY AND HEALTH 2019; 1:14-24. [PMID: 32835207 PMCID: PMC7347341 DOI: 10.1016/j.bsheal.2019.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/14/2018] [Accepted: 12/08/2018] [Indexed: 11/25/2022] Open
Abstract
The onsite next generation sequencing (NGS) of Ebola virus (EBOV) genomes during the 2013–2016 Ebola epidemic in Western Africa provides an opportunity to trace the origin, transmission, and evolution of this virus. Herein, we have diagnosed a cohort of EBOV patients in Sierra Leone in 2015, during the late phase of the outbreak. The surviving EBOV patients had a recovery process characterized by decreasing viremia, fever, and biochemical parameters. EBOV genomes sequenced through the longitudinal blood samples of these patients showed dynamic intra-host substitutions of the virus during acute infection, including the previously described short stretches of 13 serial T>C mutations. Remarkably, within individual patients, samples collected during the early phase of infection possessed Ts at these nucleotide sites, whereas they were replaced by Cs in samples collected in the later phase, suggesting that these short stretches of T>C mutations could emerge independently. In addition, up to a total of 35 nucleotide sites spanning the EBOV genome were mutated coincidently. Our study showed the dynamic intra-host adaptation of EBOV during patient recovery and gave more insight into the complex EBOV-host interactions.
Collapse
|
46
|
Wing K, Oza S, Houlihan C, Glynn JR, Irvine S, Warrell CE, Simpson AJH, Boufkhed S, Sesay A, Vandi L, Sebba SC, Shetty P, Cummings R, Checchi F, McGowan CR. Surviving Ebola: A historical cohort study of Ebola mortality and survival in Sierra Leone 2014-2015. PLoS One 2018; 13:e0209655. [PMID: 30589913 PMCID: PMC6307710 DOI: 10.1371/journal.pone.0209655] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 12/10/2018] [Indexed: 11/30/2022] Open
Abstract
Background While a number of predictors for Ebola mortality have been identified, less is known about post-viral symptoms. The identification of acute-illness predictors for post-viral symptoms could allow the selection of patients for more active follow up in the future, and those in whom early interventions may be beneficial in the long term. Studying predictors of both mortality and post-viral symptoms within a single cohort of patients could also further our understanding of the pathophysiology of survivor sequelae. Methods/Principal findings We performed a historical cohort study using data collected as part of routine clinical care from an Ebola Treatment Centre (ETC) in Kerry Town, Sierra Leone, in order to identify predictors of mortality and of post-viral symptoms. Variables included as potential predictors were sex, age, date of admission, first recorded viral load at the ETC and symptoms (recorded upon presentation at the ETC). Multivariable logistic regression was used to identify predictors. Of 263 Ebola-confirmed patients admitted between November 2014 and March 2015, 151 (57%) survived to ETC discharge. Viral load was the strongest predictor of mortality (adjusted OR comparing high with low viral load: 84.97, 95% CI 30.87–345.94). We did not find evidence that a high viral load predicted post-viral symptoms (ocular: 1.17, 95% CI 0.35–3.97; musculoskeletal: 1.07, 95% CI 0.28–4.08). Ocular post-viral symptoms were more common in females (2.31, 95% CI 0.98–5.43) and in those who had experienced hiccups during the acute phase (4.73, 95% CI 0.90–24.73). Conclusions/Significance These findings may add epidemiological support to the hypothesis that post-viral symptoms have an immune-mediated aspect and may not only be a consequence of high viral load and disease severity.
Collapse
Affiliation(s)
- Kevin Wing
- Save the Children International, Kerry Town, Sierra Leone
- London School of Hygiene & Tropical Medicine, London, United Kingdom
- * E-mail:
| | - Shefali Oza
- Save the Children International, Kerry Town, Sierra Leone
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Catherine Houlihan
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Judith R. Glynn
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Sharon Irvine
- Save the Children International, Kerry Town, Sierra Leone
| | | | - Andrew J. H. Simpson
- Rare and Imported Pathogens Laboratory, Public Health England, Porton, Wilts, United Kingdom
| | - Sabah Boufkhed
- Save the Children International, Kerry Town, Sierra Leone
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Alieu Sesay
- Save the Children International, Kerry Town, Sierra Leone
| | - Lahai Vandi
- Save the Children International, Kerry Town, Sierra Leone
| | | | - Pranav Shetty
- Humanitarian Public Health Technical Unit, Save the Children, London, United Kingdom
| | - Rachael Cummings
- Humanitarian Public Health Technical Unit, Save the Children, London, United Kingdom
| | - Francesco Checchi
- Save the Children International, Kerry Town, Sierra Leone
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Catherine R. McGowan
- Save the Children International, Kerry Town, Sierra Leone
- London School of Hygiene & Tropical Medicine, London, United Kingdom
- Humanitarian Public Health Technical Unit, Save the Children, London, United Kingdom
| |
Collapse
|
47
|
Schindell BG, Webb AL, Kindrachuk J. Persistence and Sexual Transmission of Filoviruses. Viruses 2018; 10:E683. [PMID: 30513823 PMCID: PMC6316729 DOI: 10.3390/v10120683] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 12/27/2022] Open
Abstract
There is an increasing frequency of reports regarding the persistence of the Ebola virus (EBOV) in Ebola virus disease (EVD) survivors. During the 2014⁻2016 West African EVD epidemic, sporadic transmission events resulted in the initiation of new chains of human-to-human transmission. Multiple reports strongly suggest that these re-emergences were linked to persistent EBOV infections and included sexual transmission from EVD survivors. Asymptomatic infection and long-term viral persistence in EVD survivors could result in incidental introductions of the Ebola virus in new geographic regions and raise important national and local public health concerns. Alarmingly, although the persistence of filoviruses and their potential for sexual transmission have been documented since the emergence of such viruses in 1967, there is limited knowledge regarding the events that result in filovirus transmission to, and persistence within, the male reproductive tract. Asymptomatic infection and long-term viral persistence in male EVD survivors could lead to incidental transfer of EBOV to new geographic regions, thereby generating widespread outbreaks that constitute a significant threat to national and global public health. Here, we review filovirus testicular persistence and discuss the current state of knowledge regarding the rates of persistence in male survivors, and mechanisms underlying reproductive tract localization and sexual transmission.
Collapse
Affiliation(s)
- Brayden G Schindell
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Andrew L Webb
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Jason Kindrachuk
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| |
Collapse
|
48
|
Davey RT, Dodd L, Proschan M, Jahrling P, Hensley L, Higgs E, Lane HC. The Past Need Not Be Prologue: Recommendations for Testing and Positioning the Most-Promising Medical Countermeasures for the Next Outbreak of Ebola Virus Infection. J Infect Dis 2018; 218:S690-S697. [PMID: 30032267 PMCID: PMC6249585 DOI: 10.1093/infdis/jiy334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Background The 2013-2016 outbreak of Ebola virus disease (EVD) in West Africa led to unprecedented morbidity and mortality. Although different classes of putative antiviral agents with supportive preclinical data were available for testing, and although several attempts to perform meaningful evaluation of these agents were undertaken during the epidemic, different research methods, a lack of appropriate controls in most studies, and formidable logistical challenges to completion of studies under field conditions hampered the success of these efforts. Ultimately only 1 randomized, placebo-controlled clinical trial (PREVAIL II) was performed in this setting, and, owing to a decrease in the number of new cases available for study, it, too, ended prior to reaching definitive results. Retrospective review of the lessons learned from this outbreak argues strongly for the need for much better preparedness in terms of selecting the trial design and drug(s) for use during the next outbreak. Methods Using recent data provided by representatives from the pharmaceutical industry, clinical and laboratory subject matter experts from the National Institute of Allergy and Infectious Diseases, other US government agencies, and academic partners were consulted regarding the current state of knowledge about several lead compounds with putative activity against EVD. Consensus was sought on recommendations concerning the most promising treatment strategies against EVD that should be studied in the context of a randomized clinical trial during the next outbreak. Results Four compounds from 2 different classes (monoclonal antibody [mAb] cocktails and direct-acting antiviral agents [DAAs]) were highlighted as lead candidates, limitations in the current knowledge base about these drug classes were reviewed, and recommendations about the optimal clinical research design for studying combinations of these different agents were made. Conclusions Although achieving the desired sample size could be challenging, a randomized, controlled clinical trial based on a combination strategy of a mAb with a DAA was recommended as the most appropriate clinical trial design to be undertaken during the next outbreak of EVD.
Collapse
Affiliation(s)
- Richard T Davey
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda
| | - Lori Dodd
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda
| | - Michael Proschan
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda
| | - Peter Jahrling
- Integrated Research Facility, NIAID, NIH, Frederick, Maryland
| | - Lisa Hensley
- Integrated Research Facility, NIAID, NIH, Frederick, Maryland
| | - Elizabeth Higgs
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda
| | - H Clifford Lane
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda
| |
Collapse
|
49
|
Martins DP, Barros MT, Pierobon M, Kandhavelu M, Lio' P, Balasubramaniam S. Computational Models for Trapping Ebola Virus Using Engineered Bacteria. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 15:2017-2027. [PMID: 29994771 DOI: 10.1109/tcbb.2018.2836430] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The outbreak of the Ebola virus in recent years has resulted in numerous research initiatives to seek new solutions to contain the virus. A number of approaches that have been investigated include new vaccines to boost the immune system. An alternative post-exposure treatment is presented in this paper. The proposed approach for clearing the Ebola virus can be developed through a microfluidic attenuator, which contains the engineered bacteria that traps Ebola flowing through the blood onto its membrane. The paper presents the analysis of the chemical binding force between the virus and a genetically engineered bacterium considering the opposing forces acting on the attachment point, including hydrodynamic tension and drag force. To test the efficacy of the technique, simulations of bacterial motility within a confined area to trap the virus were performed. More than 60 percent of the displaced virus could be collected within 15 minutes. While the proposed approach currently focuses on in vitro environments for trapping the virus, the system can be further developed into a future treatment system whereby blood can be cycled out of the body into a microfluidic device that contains the engineered bacteria to trap viruses.
Collapse
|
50
|
Serological Investigation of Laboratory-Confirmed and Suspected Ebola Virus Disease Patients During the Late Phase of the Ebola Outbreak in Sierra Leone. Virol Sin 2018; 33:323-334. [PMID: 30066045 DOI: 10.1007/s12250-018-0044-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 06/29/2018] [Indexed: 10/28/2022] Open
Abstract
This study aimed to investigate the serological characteristics of Ebola virus (EBOV) infection during the late phase of the Ebola outbreak in Sierra Leone. In total, 877 blood samples from 694 suspected Ebola virus disease (EVD) cases assessed from March to December 2015, were analyzed via real-time reverse transcription polymerase chain reaction (RT-PCR) for viral RNA and enzyme-linked immunosorbent assay (ELISA) and Luminex to detect antibodies against EBOV. Viral load and EBOV-specific IgM/IgG titers displayed a declining trend during March to December 2015. Viral RNA load decreased rapidly at earlier stages after disease onset, while EBOV-specific IgM and IgG still persisted in 58.1% (18/31) and 93.5% (29/31) of the confirmed EVD patients and in 3.8% (25/663) and 17.8% (118/663) of the RNA-negative suspected patients in the later phase, respectively. Dynamic analysis of longitudinally collected samples from eight EVD patients revealed typically reversed trends of declining viral load and increasing IgM and/or IgG titers in response to the EBOV infection. The present results indicate that certain populations of Sierra Leone developed immunity to an EBOV infection in the late phase of the outbreak, providing novel insights into the risk assessment of EBOV infections among human populations.
Collapse
|