151
|
Marzi A, Menicucci AR, Engelmann F, Callison J, Horne EJ, Feldmann F, Jankeel A, Feldmann H, Messaoudi I. Protection Against Marburg Virus Using a Recombinant VSV-Vaccine Depends on T and B Cell Activation. Front Immunol 2019; 9:3071. [PMID: 30723475 PMCID: PMC6350103 DOI: 10.3389/fimmu.2018.03071] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/11/2018] [Indexed: 12/24/2022] Open
Abstract
Marburg virus (MARV) is the causative agent of hemorrhagic fever outbreaks with high case fatality rates. Closely related to Ebola virus, MARV is a filamentous virus with a negative-sense, single-stranded RNA genome. Although extensive studies on filovirus countermeasures have been conducted, there are no licensed treatments against MARV infections. An experimental vaccine based on the recombinant vesicular stomatitis virus (VSV) expressing the MARV-Musoke glycoprotein demonstrated complete protection when a single dose was administered 28 days and up to 14 months prior to MARV challenge. Here, we analyzed the protective efficacy of an updated vaccine expressing the MARV-Angola glycoprotein (VSV-MARV). A single dose of VSV-MARV given 5 weeks before challenge provided uniform protection with no detectable viremia. The vaccine induced B and T cell proliferation and, importantly, antigen-specific IgG production. Transcriptomic signatures confirm these findings and suggest innate immunity engendered by VSV-MARV may direct the development of protective humoral immunity.
Collapse
Affiliation(s)
- Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Andrea R Menicucci
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Flora Engelmann
- Department of Cell Molecular Biology, Northwestern University, Evanston, IL, United States
| | - Julie Callison
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Eva J Horne
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Allen Jankeel
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
152
|
Meyer M, Malherbe DC, Bukreyev A. Can Ebola Virus Vaccines Have Universal Immune Correlates of protection? Trends Microbiol 2019; 27:8-16. [PMID: 30201511 PMCID: PMC6309495 DOI: 10.1016/j.tim.2018.08.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/30/2018] [Accepted: 08/15/2018] [Indexed: 12/22/2022]
Abstract
Testing vaccine efficacy against the highly lethal Ebola virus (EBOV) in humans is almost impossible due to obvious ethical reasons and the sporadic nature of outbreaks. For such situations, the 'animal rule' was established, requiring the product be tested in animal models, expected to predict the response observed in humans. For vaccines, this testing aims to identify immune correlates of protection, such as antibody or cell-mediated responses. In the wake of the 2013-2016 EBOV epidemic, and despite advancement of promising candidates into clinical trials, protective correlates remain ambiguous. In the hope of identifying a reliable correlate by comparing preclinical and clinical trial data on immune responses to vaccination, we conclude that correlates are not universal for all EBOV vaccines.
Collapse
Affiliation(s)
- Michelle Meyer
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77555, USA; These authors contributed equally to this work
| | - Delphine C Malherbe
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77555, USA; These authors contributed equally to this work
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Department Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77555, USA.
| |
Collapse
|
153
|
Huttner A, Siegrist CA. Durability of single-dose rVSV-ZEBOV vaccine responses: what do we know? Expert Rev Vaccines 2018; 17:1105-1110. [PMID: 30422031 DOI: 10.1080/14760584.2018.1546582] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION The live-attenuated recombinant vesicular stomatitis virus vaccine expressing the glycoprotein (GP) of Zaire Ebola virus (rVSV-ZEBOV) has proven immunogenic in humans and effective in field studies. Yet long-term durability of vaccine responses is unknown. AREAS COVERED We survey the evidence available in the literature for the durability of human responses to rVSV-ZEBOV. We also review determinants of initial responses and of their persistence. EXPERT COMMENTARY Persistence of EBOV-GP-specific antibody responses is strong at 2 years - currently the longest post-vaccination interval studied - after a single injection. Vaccine dose predicts persistence of seropositivity, though the magnitude of antibody responses at later time points becomes less dose-dependent. Vaccine-related arthritis is a significant predictor of both persistence and magnitude of the antibody response.
Collapse
Affiliation(s)
- Angela Huttner
- a Division of Infectious Diseases , University Hospitals of Geneva , Geneva , Switzerland.,b Centre for Vaccinology , University Hospitals and University of Geneva , Geneva , Switzerland
| | - Claire-Anne Siegrist
- b Centre for Vaccinology , University Hospitals and University of Geneva , Geneva , Switzerland
| |
Collapse
|
154
|
Bramble MS, Hoff N, Gilchuk P, Mukadi P, Lu K, Doshi RH, Steffen I, Nicholson BP, Lipson A, Vashist N, Sinai C, Spencer D, Olinger G, Wemakoy EO, Illunga BK, Pettitt J, Logue J, Marchand J, Varughese J, Bennett RS, Jahrling P, Cavet G, Serafini T, Ollmann Saphire E, Vilain E, Muyembe-Tamfum JJ, Hensely LE, Simmons G, Crowe JE, Rimoin AW. Pan-Filovirus Serum Neutralizing Antibodies in a Subset of Congolese Ebolavirus Infection Survivors. J Infect Dis 2018; 218:1929-1936. [PMID: 30107445 PMCID: PMC6217721 DOI: 10.1093/infdis/jiy453] [Citation(s) in RCA: 15] [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: 06/28/2018] [Accepted: 07/31/2018] [Indexed: 11/30/2022] Open
Abstract
One year after a Zaire ebolavirus (EBOV) outbreak occurred in the Boende Health Zone of the Democratic Republic of the Congo during 2014, we sought to determine the breadth of immune response against diverse filoviruses including EBOV, Bundibugyo (BDBV), Sudan (SUDV), and Marburg (MARV) viruses. After assessing the 15 survivors, 5 individuals demonstrated some degree of reactivity to multiple ebolavirus species and, in some instances, Marburg virus. All 5 of these survivors had immunoreactivity to EBOV glycoprotein (GP) and EBOV VP40, and 4 had reactivity to EBOV nucleoprotein (NP). Three of these survivors showed serologic responses to the 3 species of ebolavirus GPs tested (EBOV, BDBV, SUDV). All 5 samples also exhibited ability to neutralize EBOV using live virus, in a plaque reduction neutralization test. Remarkably, 3 of these EBOV survivors had plasma antibody responses to MARV GP. In pseudovirus neutralization assays, serum antibodies from a subset of these survivors also neutralized EBOV, BDBV, SUDV, and Taï Forest virus as well as MARV. Collectively, these findings suggest that some survivors of naturally acquired ebolavirus infection mount not only a pan-ebolavirus response, but also in less frequent cases, a pan-filovirus neutralizing response.
Collapse
Affiliation(s)
- Matthew S Bramble
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
- Department of Genetic Medicine Research, Children’s Research Institute, Children’s National Medical Center, Washington, District of Columbia
| | - Nicole Hoff
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, and Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Patrick Mukadi
- Institut National de Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo
| | - Kai Lu
- Blood Systems Research Institute, and Department of Laboratory Medicine, University of California, San Francisco
| | - Reena H Doshi
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| | - Imke Steffen
- Blood Systems Research Institute, and Department of Laboratory Medicine, University of California, San Francisco
| | - Bradly P Nicholson
- Institute for Medical Research, Durham Veterans Affairs Medical Center, North Carolina
| | - Allen Lipson
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| | - Neerja Vashist
- Department of Genetic Medicine Research, Children’s Research Institute, Children’s National Medical Center, Washington, District of Columbia
| | - Cyrus Sinai
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| | - D’andre Spencer
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| | - Garrard Olinger
- Boston University, School of Medicine, Department of Medicine, Massachusetts
| | | | - Benoit Kebela Illunga
- Direction de la Lutte Contre les Maladies, Ministère de la Sante, Kinshasa, Democratic Republic of the Congo
| | - James Pettitt
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | - James Logue
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | - Jonathan Marchand
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | - Justin Varughese
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | - Richard S Bennett
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | - Peter Jahrling
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | | | | | - Erica Ollmann Saphire
- Skaggs Institute for Chemical Biology, La Jolla, California
- Department of Immunology and Microbial Science, Scripps Research Institute, La Jolla, California
| | - Eric Vilain
- Department of Genetic Medicine Research, Children’s Research Institute, Children’s National Medical Center, Washington, District of Columbia
| | | | - Lisa E Hensely
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
- Emerging Viral Pathogens Section, NIAID, NIH, Frederick, Maryland
| | - Graham Simmons
- Blood Systems Research Institute, and Department of Laboratory Medicine, University of California, San Francisco
| | - James E Crowe
- Vanderbilt Vaccine Center, and Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Departments of Pediatrics and Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anne W Rimoin
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| |
Collapse
|
155
|
Gupta SB, Coller BA, Feinberg M. Unprecedented pace and partnerships: the story of and lessons learned from one Ebola vaccine program. Expert Rev Vaccines 2018; 17:913-923. [PMID: 30269612 DOI: 10.1080/14760584.2018.1527692] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The Ebola epidemic in West Africa from 2014 to 2016 was unique in its size, location, and duration; this article reviews the experiences and lessons learned for one vaccine candidate developed during the outbreak and discusses critical gaps that still exist today which will need to be addressed for successful end to end emerging infectious disease vaccine product development in the future. AREAS COVERED Through the formation of numerous international partnerships, the rVSVΔG-ZEBOV-GP vaccine advanced through Phase I/II/III clinical trials which resulted in favorable Phase III efficacy results. Key lessons learned that could be used to facilitate future vaccine development efforts include sufficient preclinical work in relevant animal models, innovative partnerships created to pool resources and expertise, and 'hyper' coordination and communication among partners to build trust and ensure an adequate regulatory package needed to license a vaccine. EXPERT COMMENTARY As evidenced by the 2014-2016 outbreak in West Africa as well as the two other most recent outbreaks in the Democratic Republic of the Congo in 2018, there is an urgent need to develop new models for emerging infection vaccine development where trusted partners come together and where the development of vaccines is a shared responsibility conducted in advance of the next crisis.
Collapse
Affiliation(s)
- Swati B Gupta
- a Global Clinical Development , Merck & Co., Inc , Kenilworth , NJ , USA.,b Research Integration & Innovation , International AIDS Vaccine Initiative , New York , NY , USA
| | - Beth-Ann Coller
- a Global Clinical Development , Merck & Co., Inc , Kenilworth , NJ , USA
| | - Mark Feinberg
- a Global Clinical Development , Merck & Co., Inc , Kenilworth , NJ , USA.,c Executive Office , International AIDS Vaccine Initiative , New York , NY , USA
| |
Collapse
|
156
|
Babirye P, Musubika C, Kirimunda S, Downing R, Lutwama JJ, Mbidde EK, Weyer J, Paweska JT, Joloba ML, Wayengera M. Identity and validity of conserved B cell epitopes of filovirus glycoprotein: towards rapid diagnostic testing for Ebola and possibly Marburg virus disease. BMC Infect Dis 2018; 18:498. [PMID: 30285648 PMCID: PMC6171133 DOI: 10.1186/s12879-018-3409-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 09/23/2018] [Indexed: 11/13/2022] Open
Abstract
Background Ebolavirus and Marburgvirus are genera of the virus family Filoviridae. Filoviruses cause rare but fatal viral hemorrhagic fevers (VHFs) in remote villages of equatorial Africa with potential for regional and international spread. Point-of-care (POC) rapid diagnostic tests (RDTs) are critical for early epidemic detection, reponse and control. There are 2 RDTs for Zaire ebolavirus (EBOV), but not other Ebolavirus spp. or Marburg marburgvirus (MARV). We validate 3 conserved B cell epitopes of filovirus glycoprotein (GP) using ebola virus diseases (EVD) survivor samples, towards devising pan-filovirus RDTs. Methods In-silico Immuno-informatics:- (a) multiple and basic local alignments of amino-acid sequences of filovirus (4 Ebolavirus spp. & MARV) Gp1, 2 and epitope prediction and conservation analyses within context of ClusterW, BLAST-P and the immune epitope database analysis resource (IEDB-AR); alongside (b) in-vitro enzyme immuno-assays (EIAs) for SUDV Gp1, 2 antigen and host-specific antibodies (IgM and IgG) among 94 gamma irradiated EVD survivor serum and 9 negative controls. Results Linear B cell epitopes were present across the entire length of all Gp1, 2, most lying in the region between amino acids positioned 350 and 500. Three seperate epitopes 97/80_GAFFLYDRLAST, 39_YEAGEWAENCY and 500_CGLRQLANETTQALQLFLRATTELR (designated UG-Filo-Peptide− 1, 2 and 3 respectively) were conserved within all studied filovirus species Gp1, 2. Gp1, 2 host specific IgM levels were comparably low (av. ODs < 0.04 [95% CI: 0.02837 to 0.04033]) among the 9 negative controls and 57 survivor samples analyzed. Host specific IgG levels, on the other hand, were elevated (av. ODs > 1.7525 [95% CI: 0.3010 to 3.1352]) among the 92 survivor samples relative to the 9 negative controls (av. ODs < 0.2.321 [95% CI: -0.7596 to 0.5372]). Filovirus Gp1, 2 antigen was not detected [av. ODs < 0.20] within EVD survivor serum relative to recombinant protein positive controls [av. ODs = 0.50]. Conclusions These conserved B cell epitopes of filovirus Gp1, 2 and their derivative antibodies are promising for research and development of RDTs for EVD, with potential for extension to detect MVD. Electronic supplementary material The online version of this article (10.1186/s12879-018-3409-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peace Babirye
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P o Box 7072, Kampala, Uganda
| | - Carol Musubika
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P o Box 7072, Kampala, Uganda
| | - Samuel Kirimunda
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P o Box 7072, Kampala, Uganda
| | - Robert Downing
- Arbovirology and Filovirology Laboratories/Centers for Disease Control-CDC, Uganda Virus Research Institute (UVRI), Entebbe, Uganda
| | - Julian J Lutwama
- Arbovirology and Filovirology Laboratories/Centers for Disease Control-CDC, Uganda Virus Research Institute (UVRI), Entebbe, Uganda
| | - Edward K Mbidde
- Arbovirology and Filovirology Laboratories/Centers for Disease Control-CDC, Uganda Virus Research Institute (UVRI), Entebbe, Uganda
| | - Jacqueline Weyer
- Center for Emerging Zoonotic Diseases, National Institute for Communicable Diseases, Johanesburg, South Africa
| | - Janusz T Paweska
- Center for Emerging Zoonotic Diseases, National Institute for Communicable Diseases, Johanesburg, South Africa
| | - Moses L Joloba
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P o Box 7072, Kampala, Uganda
| | - Misaki Wayengera
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P o Box 7072, Kampala, Uganda. .,Unit of Genetics & Genomics & Department of Pathology, School of Biomedical Sciences, College of Health Sciences, Makerere University, P o Box 7072, Kampala, Uganda.
| |
Collapse
|
157
|
Medaglini D, Santoro F, Siegrist CA. Correlates of vaccine-induced protective immunity against Ebola virus disease. Semin Immunol 2018; 39:65-72. [DOI: 10.1016/j.smim.2018.07.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/10/2018] [Accepted: 07/13/2018] [Indexed: 10/28/2022]
|
158
|
Rauch S, Jasny E, Schmidt KE, Petsch B. New Vaccine Technologies to Combat Outbreak Situations. Front Immunol 2018; 9:1963. [PMID: 30283434 PMCID: PMC6156540 DOI: 10.3389/fimmu.2018.01963] [Citation(s) in RCA: 360] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/09/2018] [Indexed: 01/07/2023] Open
Abstract
Ever since the development of the first vaccine more than 200 years ago, vaccinations have greatly decreased the burden of infectious diseases worldwide, famously leading to the eradication of small pox and allowing the restriction of diseases such as polio, tetanus, diphtheria, and measles. A multitude of research efforts focuses on the improvement of established and the discovery of new vaccines such as the HPV (human papilloma virus) vaccine in 2006. However, radical changes in the density, age distribution and traveling habits of the population worldwide as well as the changing climate favor the emergence of old and new pathogens that bear the risk of becoming pandemic threats. In recent years, the rapid spread of severe infections such as HIV, SARS, Ebola, and Zika have highlighted the dire need for global preparedness for pandemics, which necessitates the extremely rapid development and comprehensive distribution of vaccines against potentially previously unknown pathogens. What is more, the emergence of antibiotic resistant bacteria calls for new approaches to prevent infections. Given these changes, established methods for the identification of new vaccine candidates are no longer sufficient to ensure global protection. Hence, new vaccine technologies able to achieve rapid development as well as large scale production are of pivotal importance. This review will discuss viral vector and nucleic acid-based vaccines (DNA and mRNA vaccines) as new approaches that might be able to tackle these challenges to global health.
Collapse
|
159
|
Abstract
The West African Ebola virus (EBOV) epidemic has fast-tracked countermeasures for this rare, emerging zoonotic pathogen. Until 2013-2014, most EBOV vaccine candidates were stalled between the preclinical and clinical milestones on the path to licensure, because of funding problems, lack of interest from pharmaceutical companies, and competing priorities in public health. The unprecedented and devastating epidemic propelled vaccine candidates toward clinical trials that were initiated near the end of the active response to the outbreak. Those trials did not have a major impact on the epidemic but provided invaluable data on vaccine safety, immunogenicity, and, to a limited degree, even efficacy in humans. There are plenty of lessons to learn from these trials, some of which are addressed in this review. Better preparation is essential to executing an effective response to EBOV in the future; yet, the first indications of waning interest are already noticeable.
Collapse
Affiliation(s)
- Heinz Feldmann
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA;
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba 93E 0J9, Canada
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA
| | - Andrea Marzi
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA;
| |
Collapse
|
160
|
Gross L, Lhomme E, Pasin C, Richert L, Thiebaut R. Ebola vaccine development: Systematic review of pre-clinical and clinical studies, and meta-analysis of determinants of antibody response variability after vaccination. Int J Infect Dis 2018; 74:83-96. [PMID: 29981944 DOI: 10.1016/j.ijid.2018.06.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/20/2018] [Accepted: 06/28/2018] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES For Ebola vaccine development, antibody response is a major endpoint although its determinants are not well known. We aimed to review Ebola vaccine studies and to assess factors associated with antibody response variability in humans. METHODS We searched PubMed and Scopus for preventive Ebola vaccine studies in humans or non-human primates (NHP), published up to February 2018. For each vaccination group with Ebola Zaire antibody titre measurements after vaccination, data about antibody response and its potential determinants were extracted. A random-effects meta-regression was conducted including human groups with at least 8 individuals. RESULTS We reviewed 49 studies (202 vaccination groups including 74 human groups) with various vaccine platforms and antigen inserts. Mean antibody titre was slightly higher in NHP (3.10, 95% confidence interval [293; 327]) than in humans (2.75 [257; 293]). Vaccine platform (p<0·001) and viral strain used for antibody detection (p<0·001) were associated with antibody response in humans, but adjusted heterogeneity remained at 95%. CONCLUSIONS Various platforms have been evaluated in humans, including Ad26, Ad5, ChimpAd3, DNA, MVA, and VSV. In addition to platforms, viral strain used for antibody detection influences antibody response. However, variability remained mostly unexplained. Therefore, comparison of vaccine immunogenicity needs randomised controlled trials.
Collapse
Affiliation(s)
- Lise Gross
- SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France
| | - Edouard Lhomme
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France; Pôle de Santé Publique, CHU de Bordeaux, Bordeaux, F-33000, France
| | - Chloé Pasin
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France
| | - Laura Richert
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France; Pôle de Santé Publique, CHU de Bordeaux, Bordeaux, F-33000, France
| | - Rodolphe Thiebaut
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France; Pôle de Santé Publique, CHU de Bordeaux, Bordeaux, F-33000, France.
| |
Collapse
|
161
|
A novel electrochemical DNA biosensor for Ebola virus detection. Anal Biochem 2018; 557:151-155. [DOI: 10.1016/j.ab.2018.06.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/30/2018] [Accepted: 06/12/2018] [Indexed: 12/13/2022]
|
162
|
Öhlund P, García-Arriaza J, Zusinaite E, Szurgot I, Männik A, Kraus A, Ustav M, Merits A, Esteban M, Liljeström P, Ljungberg K. DNA-launched RNA replicon vaccines induce potent anti-Ebolavirus immune responses that can be further improved by a recombinant MVA boost. Sci Rep 2018; 8:12459. [PMID: 30127450 PMCID: PMC6102224 DOI: 10.1038/s41598-018-31003-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 08/07/2018] [Indexed: 12/28/2022] Open
Abstract
There are currently no licensed therapeutic treatment or preventive vaccines against Ebolavirus disease, and the 2013-2016 West African outbreak of Ebolavirus disease spread rapidly and resulted in almost 30,000 cases and more than 11,000 deaths. However, the devastating outbreak has spurred the development of novel Ebolavirus vaccines. Here, we demonstrate that alphavirus-based DNA-launched self-replicating RNA replicon vaccines (DREP) encoding either the glycoprotein (GP) gene or co-expressing the GP and VP40 genes of Sudan or Zaire Ebolavirus are immunogenic in mice inducing both binding and neutralizing antibodies as well as CD8 T cell responses. In addition, antibodies were cross-reactive against another Ebolavirus, although the specificity was higher for the vaccination antigen. DREP vaccines were more immunogenic than recombinant MVA vaccines expressing the same Ebolavirus antigens. However, a DREP prime followed by an MVA boost immunization regimen improved vaccine immunogenicity as compared to DREP and MVA homologous prime-boost immunizations. Moreover, we show that a bivalent approach targeting both Sudan and Zaire Ebolavirus can be employed without significant loss of immunity. This opens for further investigation of a pan-Ebolavirus or even a pan-filovirus vaccine.
Collapse
Affiliation(s)
- Pontus Öhlund
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Biomedical Science and Veterinary Public Health, Virology Unit, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Juan García-Arriaza
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Eva Zusinaite
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Inga Szurgot
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Andres Männik
- Icosagen Cell Factory OÜ, Ülenurme vald, Tartumaa, Estonia
| | - Annette Kraus
- Department of Microbiology, Public Health Agency of Sweden, Solna, Sweden
| | - Mart Ustav
- Icosagen Cell Factory OÜ, Ülenurme vald, Tartumaa, Estonia
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Peter Liljeström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Karl Ljungberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
163
|
Brett-Major D, Lawler J. Catching Chances: The Movement to Be on the Ground and Research Ready before an Outbreak. Viruses 2018; 10:E439. [PMID: 30126221 PMCID: PMC6116208 DOI: 10.3390/v10080439] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/04/2018] [Accepted: 08/14/2018] [Indexed: 12/17/2022] Open
Abstract
After more than 28,000 Ebola virus disease cases and at least 11,000 deaths in West Africa during the 2014⁻2016 epidemic, the world remains without a licensed vaccine or therapeutic broadly available and demonstrated to alleviate suffering. This deficiency has been felt acutely in the two, short, following years with two Ebola virus outbreaks in the Democratic Republic of Congo (DRC), and a Marburg virus outbreak in Uganda. Despite billions of U.S. dollars invested in developing medical countermeasures for filoviruses in the antecedent decades, resulting in an array of preventative, diagnostic, and therapeutic products, none are available on commercial shelves. This paper explores why just-in-time research efforts in the field during the West Africa epidemic failed, as well as some recent initiatives to prevent similarly lost opportunities.
Collapse
Affiliation(s)
- David Brett-Major
- Department of Preventive Medicine and Biostatistics, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, MD 20814, USA.
| | - James Lawler
- Global Center for Health Security, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
164
|
Liu Y, Ye L, Lin F, Gomaa Y, Flyer D, Carrion R, Patterson JL, Prausnitz MR, Smith G, Glenn G, Wu H, Compans RW, Yang C. Intradermal immunization by Ebola virus GP subunit vaccines using microneedle patches protects mice against lethal EBOV challenge. Sci Rep 2018; 8:11193. [PMID: 30046140 PMCID: PMC6060117 DOI: 10.1038/s41598-018-29135-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/17/2018] [Indexed: 11/09/2022] Open
Abstract
Development of a safe and efficacious filovirus vaccine is of high importance to public health. In this study, we compared immune responses induced by Ebola virus (EBOV) glycoprotein (GP) subunit vaccines via intradermal immunization with microneedle (MN) patches and the conventional intramuscular (IM) injection in mice, which showed that MN delivery of GP induced higher levels and longer lasting antibody responses against GP than IM injection. Further, we found that EBOV GP in formulation with a saponin-based adjuvant, Matrix-M, can be efficiently loaded onto MN patches. Co-delivery of Matrix-M with GP significantly enhanced induction of antibody responses by MN delivery, as also observed for IM injection. Results from challenge studies showed that all mice that received the GP/adjuvant formulation by MN or IM immunizations were protected from lethal EBOV challenge. Further, 4 out of 5 mice vaccinated by MN delivery of unadjuvanted GP also survived the challenge, whereas only 1 out of 5 mice vaccinated by IM injection of unadjuvanted GP survived the challenge. These results demonstrate that MN patch delivery of EBOV GP subunit vaccines, which is expected to enable improved safety and thermal stability, can confer effective protection against EBOV infection that is superior to IM vaccination.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Viral/genetics
- Antibodies, Viral/immunology
- Antibody Formation/immunology
- Ebola Vaccines/immunology
- Ebolavirus/immunology
- Ebolavirus/pathogenicity
- Glycoproteins/administration & dosage
- Glycoproteins/immunology
- Hemorrhagic Fever, Ebola/genetics
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/prevention & control
- Humans
- Immunization
- Injections, Intradermal
- Mice
- Vaccination
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Subunit/therapeutic use
Collapse
Affiliation(s)
- Ying Liu
- Key Laboratory of Special Animal Epidemic Disease, Ministry of Agriculture of China, Institute of Special Economic Animals and Plants, Chinese Academy of Agricultural Sciences CAAS, Changchun, Jilin 130112, P. R. China
- Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA, 30322, USA
| | - Ling Ye
- Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA, 30322, USA
| | - Fang Lin
- Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA, 30322, USA
- Central Laboratory, Tangdu Hospital at the Fourth Military Medical University, Xi'An, 710038, China
| | - Yasmine Gomaa
- Georgia Institute of Technology, 311 Ferst Drive, Atlanta, GA, 30332, USA
| | - David Flyer
- Novavax Inc., 20 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Ricardo Carrion
- Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, TX, 78227, USA
| | - Jean L Patterson
- Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, TX, 78227, USA
| | - Mark R Prausnitz
- Georgia Institute of Technology, 311 Ferst Drive, Atlanta, GA, 30332, USA
| | - Gale Smith
- Novavax Inc., 20 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Gregory Glenn
- Novavax Inc., 20 Firstfield Road, Gaithersburg, MD, 20878, USA
| | - Hua Wu
- Key Laboratory of Special Animal Epidemic Disease, Ministry of Agriculture of China, Institute of Special Economic Animals and Plants, Chinese Academy of Agricultural Sciences CAAS, Changchun, Jilin 130112, P. R. China
| | - Richard W Compans
- Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA, 30322, USA
| | - Chinglai Yang
- Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA, 30322, USA.
| |
Collapse
|
165
|
Emanuel J, Callison J, Dowd KA, Pierson TC, Feldmann H, Marzi A. A VSV-based Zika virus vaccine protects mice from lethal challenge. Sci Rep 2018; 8:11043. [PMID: 30038228 PMCID: PMC6056530 DOI: 10.1038/s41598-018-29401-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/05/2018] [Indexed: 01/25/2023] Open
Abstract
Infection with Zika virus (ZIKV) is commonly mild in humans but has been associated with alarming negative health outcomes including Guillain-Barré syndrome in adults and microcephaly in fetuses. As such, developing a vaccine for ZIKV is a global public health priority. Recombinant vesicular stomatitis virus (VSV) expressing the Ebola virus (EBOV) glycoprotein (GP) has been successfully used as a vaccine platform in the past. In this study, two novel VSV-ZIKV vaccines were generated utilizing the favorable immune targeting of the existing VSV-EBOV vector. In addition to the EBOV GP, these new vaccines express the full-length pre-membrane and envelope proteins or pre-membrane and truncated soluble envelope proteins as antigens. Efficacy testing of both of the VSV vectors against ZIKV was conducted in IFNAR−/− mice and resulted in uniform protection when a single dose was administered 28 days prior to lethal challenge. Furthermore, this vaccine is fast-acting and can uniformly protect mice from lethal disease when administered as late as 3 days prior to ZIKV challenge. Thus, VSV-ZIKV vectors are promising vaccine candidates and should move forward along the licensure pathway.
Collapse
Affiliation(s)
- Jackson Emanuel
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Julie Callison
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kimberly A Dowd
- Laboratory of Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Theodore C Pierson
- Laboratory of Viral Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
166
|
Gristwood A. Fresh approaches to vaccine development: New financial and economic models are required to bring more vaccines against a wider range of diseases to the market. EMBO Rep 2018; 19:embr.201846675. [PMID: 30002117 DOI: 10.15252/embr.201846675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
167
|
Abstract
The 2014 western Africa Ebola virus (EBOV) epidemic was unprecedented in magnitude, infecting over 28,000 and causing over 11,000 deaths. During this outbreak, multiple instances of EBOV sexual transmission were reported, including cases where the infectious individual had recovered from EBOV disease months before transmission. Potential human host factors in EBOV sexual transmission remain unstudied. Several basic seminal amyloids, most notably semen-derived enhancer of viral infection (SEVI), enhance in vitro infection by HIV and several other viruses. To test the ability of these peptides to enhance EBOV infection, viruses bearing the EBOV glycoprotein (EboGP) were preincubated with physiological concentrations of SEVI before infection of physiologically relevant cell lines and primary cells. Preincubation with SEVI significantly increased EboGP-mediated infectivity and replication in epithelium- and monocyte-derived cell lines. This enhancement was dependent upon amyloidogenesis and positive charge, and infection results were observed with both viruses carrying EboGP and authentic EBOV as well as with semen. SEVI enhanced binding of virus to cells and markedly increased its subsequent internalization. SEVI also stimulated uptake of a fluid phase marker by macropinocytosis, a critical mechanism by which cells internalize EBOV. We report a previously unrecognized ability of SEVI and semen to significantly alter viral physical properties critical for transmissibility by increasing the stability of EboGP-bearing recombinant viruses during incubation at elevated temperature and providing resistance to desiccation. Given the potential for EBOV sexual transmission to spark new transmission chains, these findings represent an important interrogation of factors potentially important for this EBOV transmission route.
Collapse
|
168
|
Suder E, Furuyama W, Feldmann H, Marzi A, de Wit E. The vesicular stomatitis virus-based Ebola virus vaccine: From concept to clinical trials. Hum Vaccin Immunother 2018; 14:2107-2113. [PMID: 29757706 PMCID: PMC6183239 DOI: 10.1080/21645515.2018.1473698] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/30/2018] [Indexed: 10/25/2022] Open
Abstract
The devastating Ebola virus (EBOV) epidemic in West Africa in 2013-2016 accelerated the progress of several vaccines and antivirals through clinical trials, including the replication-competent vesicular stomatitis virus-based vaccine expressing the EBOV glycoprotein (VSV-EBOV). Extensive preclinical testing in animal models demonstrated the prophylactic and post-exposure efficacy of this vaccine, identified the mechanism of protection, and suggested it was safe for human use. Based on these data, VSV-EBOV was extensively tested in phase 1-3 clinical trials in North America, Europe and Africa. Although some side effects of vaccination were observed, these clinical trials showed that the VSV-EBOV was safe and immunogenic in humans. Moreover, the data supported the use of VSV-EBOV as an emergency vaccine in individuals at risk for Ebola virus disease. In this review, we summarize the results of the extensive preclinical and clinical testing of the VSV-EBOV vaccine.
Collapse
MESH Headings
- Animals
- Clinical Trials as Topic
- Drug Carriers
- Drug Evaluation, Preclinical
- Drug-Related Side Effects and Adverse Reactions/epidemiology
- Drug-Related Side Effects and Adverse Reactions/pathology
- Ebola Vaccines/administration & dosage
- Ebola Vaccines/genetics
- Ebola Vaccines/immunology
- Ebola Vaccines/isolation & purification
- Hemorrhagic Fever, Ebola/prevention & control
- Humans
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/isolation & purification
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/isolation & purification
- Vesiculovirus/genetics
Collapse
Affiliation(s)
- Ellen Suder
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Wakako Furuyama
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Emmie de Wit
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| |
Collapse
|
169
|
Singh K, Marasini B, Chen X, Spearman P. A novel Ebola virus antibody-dependent cell-mediated cytotoxicity (Ebola ADCC) assay. J Immunol Methods 2018; 460:10-16. [PMID: 29894746 DOI: 10.1016/j.jim.2018.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 05/03/2018] [Accepted: 06/06/2018] [Indexed: 12/21/2022]
Abstract
Ebolaviruses are highly virulent pathogens that cause Ebola viral disease (EVD). Data from non-human primate (NHP) models and from human survivors of EVD suggest that anti-Ebola antibodies play an integral role in protection. Antibody-dependent cell-mediated cytotoxicity (ADCC) is a potential mechanism through which anti-Ebola antibodies may mediate protection. We developed a robust Ebola-specific ADCC assay for use in ongoing trials of Ebola vaccines. Stable cell lines for inducible Zaire ebolavirus glycoprotein (EBOV GP) expression were developed to provide a uniform source of target cells in the assay, and were combined with an existing human natural killer (NK) cell line as the effector cell. When applied to commercially available anti-EBOV GP monoclonal antibodies, the assay clearly differentiated antibody with high ADCC activity from those with low or no ADCC activity. Anti-EBOV ADCC activity was also detected in plasma samples from rhesus macaques immunized with a candidate Ebola vaccine. The Ebola ADCC assay reported here will be a useful tool in studying the functionality of anti-EBOV GP antibodies elicited by Ebola vaccines in ongoing and future clinical trials.
Collapse
Affiliation(s)
- Karnail Singh
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Bishal Marasini
- Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Xuemin Chen
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Paul Spearman
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Division of Infectious Diseases, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.
| |
Collapse
|
170
|
Locher S, Schweneker M, Hausmann J, Zimmer G. Immunogenicity of propagation-restricted vesicular stomatitis virus encoding Ebola virus glycoprotein in guinea pigs. J Gen Virol 2018; 99:866-879. [PMID: 29869979 PMCID: PMC6152369 DOI: 10.1099/jgv.0.001085] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Vesicular stomatitis virus (VSV) expressing the Ebola virus (EBOV) glycoprotein (GP) in place of the VSV glycoprotein G (VSV/EBOV-GP) is a promising EBOV vaccine candidate which has already entered clinical phase 3 studies. Although this chimeric virus was tolerated overall by volunteers, it still caused viremia and adverse effects such as fever and arthritis, suggesting that it might not be sufficiently attenuated. In this study, the VSV/EBOV-GP vector was further modified in order to achieve attenuation while maintaining immunogenicity. All recombinant VSV constructs were propagated on VSV G protein expressing helper cells and used to immunize guinea pigs via the intramuscular route. The humoral immune response was analysed by EBOV-GP-specific fluorescence-linked immunosorbent assay, plaque reduction neutralization test and in vitro virus-spreading inhibition test that employed recombinant VSV/EBOV-GP expressing either green fluorescent protein or secreted Nano luciferase. Most modified vector constructs induced lower levels of protective antibodies than the parental VSV/EBOV-GP or a recombinant modified vaccinia virus Ankara vector encoding full-length EBOV-GP. However, the VSV/EBOV-GP(F88A) mutant was at least as immunogenic as the parental vaccine virus although it was highly propagation-restricted. This finding suggests that VSV-vectored vaccines need not be propagation-competent to induce a robust humoral immune response. However, VSV/EBOV-GP(F88A) rapidly reverted to a fully propagation-competent virus indicating that a single-point mutation is not sufficient to maintain the propagation-restricted phenotype.
Collapse
Affiliation(s)
- Samira Locher
- Institut für Virologie und Immunologie (IVI), Sensemattstrasse 293, CH-3147 Mittelhäusern, Switzerland
| | - Marc Schweneker
- Bavarian Nordic GmbH, Fraunhoferstraße 13, D-82152 Martinsried, Germany
| | - Jürgen Hausmann
- Bavarian Nordic GmbH, Fraunhoferstraße 13, D-82152 Martinsried, Germany
| | - Gert Zimmer
- Institut für Virologie und Immunologie (IVI), Sensemattstrasse 293, CH-3147 Mittelhäusern, Switzerland
| |
Collapse
|
171
|
Lehrer AT, Wong TAS, Lieberman MM, Humphreys T, Clements DE, Bakken RR, Hart MK, Pratt WD, Dye JM. Recombinant proteins of Zaire ebolavirus induce potent humoral and cellular immune responses and protect against live virus infection in mice. Vaccine 2018; 36:3090-3100. [PMID: 28216187 PMCID: PMC8426131 DOI: 10.1016/j.vaccine.2017.01.068] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/17/2017] [Accepted: 01/30/2017] [Indexed: 01/26/2023]
Abstract
Infections with filoviruses in humans are highly virulent, causing hemorrhagic fevers which result in up to 90% mortality. In addition to natural infections, the ability to use these viruses as bioterrorist weapons is of significant concern. Currently, there are no licensed vaccines or therapeutics available to combat these infections. The pathogenesis of disease involves the dysregulation of the host's immune system, which results in impairment of the innate and adaptive immune responses, with subsequent development of lymphopenia, thrombocytopenia, hemorrhage, and death. Questions remain with regard to the few survivors of infection, who manage to mount an effective adaptive immune response. These questions concern the humoral and cellular components of this response, and whether such a response can be elicited by an appropriate prophylactic vaccine. The data reported herein describe the production and evaluation of a recombinant subunit Ebola virus vaccine candidate consisting of insect cell expressed Zaire ebolavirus (EBOV) surface glycoprotein (GP) and the matrix proteins VP24 and VP40. The recombinant subunit proteins are shown to be highly immunogenic in mice, yielding both humoral and cellular responses, as well as highly efficacious, providing up to 100% protection against a lethal challenge with live virus. These results demonstrate proof of concept for such a recombinant non-replicating vaccine candidate in the mouse model of EBOV which helps to elucidate immune correlates of protection and warrants further development.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- Ebola Vaccines/immunology
- Ebolavirus
- Female
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/prevention & control
- Immunity, Cellular
- Immunity, Humoral
- Mice
- Mice, Inbred BALB C
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/immunology
- Viral Envelope Proteins/immunology
- Viral Matrix Proteins/immunology
- Viral Proteins/immunology
Collapse
Affiliation(s)
- Axel T Lehrer
- PanThera Biopharma, LLC, Aiea, HI 96701, United States.
| | | | | | | | | | - Russell R Bakken
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| | - Mary Kate Hart
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| | - William D Pratt
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| | - John M Dye
- US Army Medical Research Institute for Infectious Diseases, Fort Detrick, MD 21702, United States
| |
Collapse
|
172
|
Zemp F, Rajwani J, Mahoney DJ. Rhabdoviruses as vaccine platforms for infectious disease and cancer. Biotechnol Genet Eng Rev 2018; 34:122-138. [PMID: 29781359 DOI: 10.1080/02648725.2018.1474320] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The family Rhabdoviridae (RV) comprises a large, genetically diverse collection of single-stranded, negative sense RNA viruses from the order Mononegavirales. Several RV members are being developed as live-attenuated vaccine vectors for the prevention or treatment of infectious disease and cancer. These include the prototype recombinant Vesicular Stomatitis Virus (rVSV) and the more recently developed recombinant Maraba Virus, both species within the genus Vesiculoviridae. A relatively strong safety profile in humans, robust immunogenicity and genetic malleability are key features that make the RV family attractive vaccine platforms. Currently, the rVSV vector is in preclinical development for vaccination against numerous high-priority infectious diseases, with clinical evaluation underway for HIV/AIDS and Ebola virus disease. Indeed, the success of the rVSV-ZEBOV vaccine during the 2014-15 Ebola virus outbreak in West Africa highlights the therapeutic potential of rVSV as a vaccine vector for acute, life-threatening viral illnesses. The rVSV and rMaraba platforms are also being tested as 'oncolytic' cancer vaccines in a series of phase 1-2 clinical trials, after being proven effective at eliciting immune-mediated tumour regression in preclinical mouse models. In this review, we discuss the biological and genetic features that make RVs attractive vaccine platforms and the development and ongoing testing of rVSV and rMaraba strains as vaccine vectors for infectious disease and cancer.
Collapse
Affiliation(s)
- Franz Zemp
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada
| | - Jahanara Rajwani
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada.,d Department of Biochemistry and Molecular Biology, Faculty of Medicine , University of Calgary , Calgary , Canada
| | - Douglas J Mahoney
- a Alberta Children's Hospital Research Institute , Calgary , Canada.,b Charbonneau Cancer Research Institute , Calgary , Canada.,c Department of Microbiology, Immunology and Infectious Disease , Faculty of Medicine , University of Calgary , Calgary , Canada.,d Department of Biochemistry and Molecular Biology, Faculty of Medicine , University of Calgary , Calgary , Canada
| |
Collapse
|
173
|
Distinct Immunogenicity and Efficacy of Poxvirus-Based Vaccine Candidates against Ebola Virus Expressing GP and VP40 Proteins. J Virol 2018. [PMID: 29514907 DOI: 10.1128/jvi.00363-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Zaire and Sudan ebolavirus species cause a severe disease in humans and nonhuman primates (NHPs) characterized by a high mortality rate. There are no licensed therapies or vaccines against Ebola virus disease (EVD), and the recent 2013 to 2016 outbreak in West Africa highlighted the need for EVD-specific medical countermeasures. Here, we generated and characterized head-to-head the immunogenicity and efficacy of five vaccine candidates against Zaire ebolavirus (EBOV) and Sudan ebolavirus (SUDV) based on the highly attenuated poxvirus vector modified vaccinia virus Ankara (MVA) expressing either the virus glycoprotein (GP) or GP together with the virus protein 40 (VP40) forming virus-like particles (VLPs). In a human monocytic cell line, the different MVA vectors (termed MVA-EBOVs and MVA-SUDVs) triggered robust innate immune responses, with production of beta interferon (IFN-β), proinflammatory cytokines, and chemokines. Additionally, several innate immune cells, such as dendritic cells, neutrophils, and natural killer cells, were differentially recruited in the peritoneal cavity of mice inoculated with MVA-EBOVs. After immunization of mice with a homologous prime/boost protocol (MVA/MVA), total IgG antibodies against GP or VP40 from Zaire and Sudan ebolavirus were differentially induced by these vectors, which were mainly of the IgG1 and IgG3 isotypes. Remarkably, an MVA-EBOV construct coexpressing GP and VP40 protected chimeric mice challenged with EBOV to a greater extent than a vector expressing GP alone. These results support the consideration of MVA-EBOVs and MVA-SUDVs expressing GP and VP40 and producing VLPs as best-in-class potential vaccine candidates against EBOV and SUDV.IMPORTANCE EBOV and SUDV cause a severe hemorrhagic fever affecting humans and NHPs. Since their discovery in 1976, they have caused several sporadic epidemics, with the recent outbreak in West Africa from 2013 to 2016 being the largest and most severe, with more than 11,000 deaths being reported. Although some vaccines are in advanced clinical phases, less expensive, safer, and more effective licensed vaccines are desirable. We generated and characterized head-to-head the immunogenicity and efficacy of five novel vaccines against EBOV and SUDV based on the poxvirus MVA expressing GP or GP and VP40. The expression of GP and VP40 leads to the formation of VLPs. These MVA-EBOV and MVA-SUDV recombinants triggered robust innate and humoral immune responses in mice. Furthermore, MVA-EBOV recombinants expressing GP and VP40 induced high protection against EBOV in a mouse challenge model. Thus, MVA expressing GP and VP40 and producing VLPs is a promising vaccine candidate against EBOV and SUDV.
Collapse
|
174
|
Rechtien A, Richert L, Lorenzo H, Martrus G, Hejblum B, Dahlke C, Kasonta R, Zinser M, Stubbe H, Matschl U, Lohse A, Krähling V, Eickmann M, Becker S, Thiébaut R, Altfeld M, Addo MM. Systems Vaccinology Identifies an Early Innate Immune Signature as a Correlate of Antibody Responses to the Ebola Vaccine rVSV-ZEBOV. Cell Rep 2018; 20:2251-2261. [PMID: 28854372 PMCID: PMC5583508 DOI: 10.1016/j.celrep.2017.08.023] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 06/21/2017] [Accepted: 08/02/2017] [Indexed: 01/15/2023] Open
Abstract
Predicting vaccine efficacy remains a challenge. We used a systems vaccinology approach to identify early innate immune correlates of antibody induction in humans receiving the Ebola vaccine rVSV-ZEBOV. Blood samples from days 0, 1, 3, 7, and 14 were analyzed for changes in cytokine levels, innate immune cell subsets, and gene expression. Integrative statistical analyses with cross-validation identified a signature of 5 early innate markers correlating with antibody titers on day 28 and beyond. Among those, IP-10 on day 3 and MFI of CXCR6 on NK cells on day 1 were independent correlates. Consistently, we found an early gene expression signature linked to IP-10. This comprehensive characterization of early innate immune responses to the rVSV-ZEBOV vaccine in humans revealed immune signatures linked to IP-10. These results suggest correlates of vaccine-induced antibody induction and provide a rationale to explore strategies for augmenting the effectiveness of vaccines through manipulation of IP-10. 5 early innate markers correlate with antibody response to Ebola vaccine rVSV-ZEBOV IP-10 on day 3 after vaccination is an independent correlate of antibody induction RNA-seq analysis identifies early gene expression signature linked to IP-10
Collapse
Affiliation(s)
- Anne Rechtien
- University Medical Center Hamburg-Eppendorf, 1(st) Department of Medicine, 20246 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 20246 Hamburg, Germany; Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany.
| | - Laura Richert
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany; INSERM U1219, INRIA SISTM, Bordeaux University, Bordeaux, France; Vaccine Research Institute, Creteil, France
| | - Hadrien Lorenzo
- INSERM U1219, INRIA SISTM, Bordeaux University, Bordeaux, France; Vaccine Research Institute, Creteil, France
| | - Gloria Martrus
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Boris Hejblum
- INSERM U1219, INRIA SISTM, Bordeaux University, Bordeaux, France; Vaccine Research Institute, Creteil, France
| | - Christine Dahlke
- University Medical Center Hamburg-Eppendorf, 1(st) Department of Medicine, 20246 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 20246 Hamburg, Germany
| | - Rahel Kasonta
- University Medical Center Hamburg-Eppendorf, 1(st) Department of Medicine, 20246 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 20246 Hamburg, Germany
| | - Madeleine Zinser
- University Medical Center Hamburg-Eppendorf, 1(st) Department of Medicine, 20246 Hamburg, Germany
| | - Hans Stubbe
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 20246 Hamburg, Germany; Division of Infectious Diseases, Department of Medicine IV, LMU, 80336 Munich, Germany
| | - Urte Matschl
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Ansgar Lohse
- University Medical Center Hamburg-Eppendorf, 1(st) Department of Medicine, 20246 Hamburg, Germany
| | - Verena Krähling
- Philipps University Marburg, Institute for Virology, 35043 Marburg, Germany
| | - Markus Eickmann
- Philipps University Marburg, Institute for Virology, 35043 Marburg, Germany
| | - Stephan Becker
- Philipps University Marburg, Institute for Virology, 35043 Marburg, Germany
| | | | - Rodolphe Thiébaut
- INSERM U1219, INRIA SISTM, Bordeaux University, Bordeaux, France; Vaccine Research Institute, Creteil, France
| | - Marcus Altfeld
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Marylyn M Addo
- University Medical Center Hamburg-Eppendorf, 1(st) Department of Medicine, 20246 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, 20246 Hamburg, Germany.
| |
Collapse
|
175
|
Callendret B, Vellinga J, Wunderlich K, Rodriguez A, Steigerwald R, Dirmeier U, Cheminay C, Volkmann A, Brasel T, Carrion R, Giavedoni LD, Patterson JL, Mire CE, Geisbert TW, Hooper JW, Weijtens M, Hartkoorn-Pasma J, Custers J, Grazia Pau M, Schuitemaker H, Zahn R. A prophylactic multivalent vaccine against different filovirus species is immunogenic and provides protection from lethal infections with Ebolavirus and Marburgvirus species in non-human primates. PLoS One 2018; 13:e0192312. [PMID: 29462200 PMCID: PMC5819775 DOI: 10.1371/journal.pone.0192312] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/22/2018] [Indexed: 12/11/2022] Open
Abstract
The search for a universal filovirus vaccine that provides protection against multiple filovirus species has been prompted by sporadic but highly lethal outbreaks of Ebolavirus and Marburgvirus infections. A good prophylactic vaccine should be able to provide protection to all known filovirus species and as an upside potentially protect from newly emerging virus strains. We investigated the immunogenicity and protection elicited by multivalent vaccines expressing glycoproteins (GP) from Ebola virus (EBOV), Sudan virus (SUDV), Taï Forest virus (TAFV) and Marburg virus (MARV). Immune responses against filovirus GP have been associated with protection from disease. The GP antigens were expressed by adenovirus serotypes 26 and 35 (Ad26 and Ad35) and modified Vaccinia virus Ankara (MVA) vectors, all selected for their strong immunogenicity and good safety profile. Using fully lethal NHP intramuscular challenge models, we assessed different vaccination regimens for immunogenicity and protection from filovirus disease. Heterologous multivalent Ad26-Ad35 prime-boost vaccination regimens could give full protection against MARV (range 75%-100% protection) and EBOV (range 50% to 100%) challenge, and partial protection (75%) against SUDV challenge. Heterologous multivalent Ad26-MVA prime-boost immunization gave full protection against EBOV challenge in a small cohort study. The use of such multivalent vaccines did not show overt immune interference in comparison with monovalent vaccines. Multivalent vaccines induced GP-specific antibody responses and cellular IFNγ responses to each GP expressed by the vaccine, and cross-reactivity to TAFV GP was detected in a trivalent vaccine expressing GP from EBOV, SUDV and MARV. In the EBOV challenge studies, higher humoral EBOV GP-specific immune responses (p = 0.0004) were associated with survival from EBOV challenge and less so for cellular immune responses (p = 0.0320). These results demonstrate that it is feasible to generate a multivalent filovirus vaccine that can protect against lethal infection by multiple members of the filovirus family.
Collapse
Affiliation(s)
| | - Jort Vellinga
- Janssen Vaccines & Prevention B.V., Leiden, Netherlands
| | | | | | | | | | | | | | - Trevor Brasel
- University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Ricardo Carrion
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Luis D. Giavedoni
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Jean L. Patterson
- Department of Virology and Immunology, Texas Biomedical Research Institute, San Antonio, Texas, United States of America
| | - Chad E. Mire
- University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Thomas W. Geisbert
- University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jay W. Hooper
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Mo Weijtens
- Janssen Vaccines & Prevention B.V., Leiden, Netherlands
| | | | | | | | | | - Roland Zahn
- Janssen Vaccines & Prevention B.V., Leiden, Netherlands
| |
Collapse
|
176
|
Wong G, Mendoza EJ, Plummer FA, Gao GF, Kobinger GP, Qiu X. From bench to almost bedside: the long road to a licensed Ebola virus vaccine. Expert Opin Biol Ther 2018; 18:159-173. [PMID: 29148858 PMCID: PMC5841470 DOI: 10.1080/14712598.2018.1404572] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION The Ebola virus (EBOV) disease epidemic during 2014-16 in West Africa has accelerated the clinical development of several vaccine candidates that have demonstrated efficacy in the gold standard nonhuman primate (NHP) model, namely cynomolgus macaques. AREAS COVERED This review discusses the pre-clinical research and if available, clinical evaluation of the currently available EBOV vaccine candidates, while emphasizing the translatability of pre-clinical data generated in the NHP model to clinical data in humans. EXPERT OPINION Despite the existence of many successful EBOV vaccine candidates in the pre-clinical stages, only two platforms became the focus of Phase 2/3 efficacy trials in Liberia, Sierra Leone, and Guinea near the peak of the epidemic: the Vesicular stomatitis virus (VSV)-vectored vaccine and the chimpanzee adenovirus type 3 (ChAd3)-vectored vaccine. The results of three distinct clinical trials involving these candidates may soon pave the way for a licensed, safe and efficacious EBOV vaccine to help combat future epidemics.
Collapse
Affiliation(s)
- Gary Wong
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People’s Hospital, Shenzhen, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, Winnipeg, MB, Canada
| | - Emelissa J. Mendoza
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | | | - George F. Gao
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People’s Hospital, Shenzhen, China
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, China
| | - Gary P. Kobinger
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Département de microbiologie-infectiologie et d’immunologie, Universite Laval, Quebec, QC, Canada
| | - Xiangguo Qiu
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, Winnipeg, MB, Canada
| |
Collapse
|
177
|
|
178
|
Tumor Necrosis Factor-Mediated Survival of CD169 + Cells Promotes Immune Activation during Vesicular Stomatitis Virus Infection. J Virol 2018; 92:JVI.01637-17. [PMID: 29142134 PMCID: PMC5774891 DOI: 10.1128/jvi.01637-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/01/2017] [Indexed: 12/14/2022] Open
Abstract
Innate immune activation is essential to mount an effective antiviral response and to prime adaptive immunity. Although a crucial role of CD169+ cells during vesicular stomatitis virus (VSV) infections is increasingly recognized, factors regulating CD169+ cells during viral infections remain unclear. Here, we show that tumor necrosis factor is produced by CD11b+ Ly6C+ Ly6G+ cells following infection with VSV. The absence of TNF or TNF receptor 1 (TNFR1) resulted in reduced numbers of CD169+ cells and in reduced type I interferon (IFN-I) production during VSV infection, with a severe disease outcome. Specifically, TNF triggered RelA translocation into the nuclei of CD169+ cells; this translocation was inhibited when the paracaspase MALT-1 was absent. Consequently, MALT1 deficiency resulted in reduced VSV replication, defective innate immune activation, and development of severe disease. These findings indicate that TNF mediates the maintenance of CD169+ cells and innate and adaptive immune activation during VSV infection.IMPORTANCE Over the last decade, strategically placed CD169+ metallophilic macrophages in the marginal zone of the murine spleen and lymph nodes (LN) have been shown to play a very important role in host defense against viral pathogens. CD169+ macrophages have been shown to activate innate and adaptive immunity via "enforced virus replication," a controlled amplification of virus particles. However, the factors regulating the CD169+ macrophages remain to be studied. In this paper, we show that after vesicular stomatitis virus infection, phagocytes produce tumor necrosis factor (TNF), which signals via TNFR1, and promote enforced virus replication in CD169+ macrophages. Consequently, lack of TNF or TNFR1 resulted in defective immune activation and VSV clearance.
Collapse
|
179
|
Single-Dose Trivalent VesiculoVax Vaccine Protects Macaques from Lethal Ebolavirus and Marburgvirus Challenge. J Virol 2018; 92:JVI.01190-17. [PMID: 29142131 DOI: 10.1128/jvi.01190-17] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/18/2017] [Indexed: 01/08/2023] Open
Abstract
Previous studies demonstrated that a single intramuscular (i.m.) dose of an attenuated recombinant vesicular stomatitis virus (rVSV) vector (VesiculoVax vector platform; rVSV-N4CT1) expressing the glycoprotein (GP) from the Mayinga strain of Zaire ebolavirus (EBOV) protected nonhuman primates (NHPs) from lethal challenge with EBOV strains Kikwit and Makona. Here, we studied the immunogenicities of an expanded range of attenuated rVSV vectors expressing filovirus GP in mice. Based on data from those studies, an optimal attenuated trivalent rVSV vector formulation was identified that included rVSV vectors expressing EBOV, Sudan ebolavirus (SUDV), and the Angola strain of Marburg marburgvirus (MARV) GPs. NHPs were vaccinated with a single dose of the trivalent formulation, followed by lethal challenge 28 days later with each of the three corresponding filoviruses. At day 14 postvaccination, a serum IgG response specific for all three GPs was detected in all the vaccinated macaques. A modest and balanced cell-mediated immune response specific for each GP was also detected in a majority of the vaccinated macaques. No matter the level of total GP-specific immune response detected postvaccination, all the vaccinated macaques were protected from disease and death following lethal challenge with each of the three filoviruses. These findings indicate that vaccination with a single dose of attenuated rVSV-N4CT1 vectors each expressing a single filovirus GP may provide protection against the filoviruses most commonly responsible for outbreaks of hemorrhagic fever in sub-Saharan Africa.IMPORTANCE The West African Ebola virus Zaire outbreak in 2013 showed that the disease was not only a regional concern, but a worldwide problem, and highlighted the need for a safe and efficacious vaccine to be administered to the populace. However, other endemic pathogens, like Ebola virus Sudan and Marburg, also pose an important health risk to the public and therefore require development of a vaccine prior to the occurrence of an outbreak. The significance of our research was the development of a blended trivalent filovirus vaccine that elicited a balanced immune response when administered as a single dose and provided complete protection against a lethal challenge with all three filovirus pathogens.
Collapse
|
180
|
Abstract
Immunization has played a large role in substantially reducing the infected and death tolls from infectious diseases. In the case of emerging diseases, the identity of the pathogen responsible, as well as the time and location for the next outbreak, cannot be accurately predicted using current means. Coupled with disjointed efforts towards the development of vaccines and a lack of funds and desire to advance promising products against known emerging pathogens to clinical trials, there has been a shortage of approved products ready for emergency use. Recent outbreaks have exposed these weaknesses, and the Coalition for Epidemic Preparedness Innovations (CEPI) was created in 2016 to address these issues. In this commentary, we discuss the establishment of such a global vaccine fund, and provide some additional points to consider for stimulating further discussion on this comprehensive, ambitious initiative.
Collapse
Affiliation(s)
- Gary Wong
- a Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People's Hospital , Shenzhen , China
| | - Xiangguo Qiu
- b Special Pathogens Program, National Microbiology Laboratory , Winnipeg , Canada.,c Department of Medical Microbiology , University of Manitoba , Winnipeg , Canada
| |
Collapse
|
181
|
Bergren NA, Miller MR, Monath TP, Kading RC. Assessment of the ability of V920 recombinant vesicular stomatitis-Zaire ebolavirus vaccine to replicate in relevant arthropod cell cultures and vector species. Hum Vaccin Immunother 2018; 14:994-1002. [PMID: 29206076 PMCID: PMC5893201 DOI: 10.1080/21645515.2017.1412898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
V920, rVSVΔG-ZEBOV-GP, is a recombinant vesicular stomatitis-Zaire ebolavirus vaccine which has shown an acceptable safety profile and provides a protective immune response against Ebola virus disease (EVD) induced by Zaire ebolavirus in humans. The purpose of this study was to determine whether the V920 vaccine is capable of replicating in arthropod cell cultures of relevant vector species and of replicating in live mosquitoes. While the V920 vaccine replicated well in Vero cells, no replication was observed in Anopheles or Aedes mosquito, Culicoides biting midge, or Lutzomyia sand fly cells, nor in live Culex or Aedes mosquitoes following exposure through intrathoracic inoculation or feeding on a high-titer infectious blood meal. The insect taxa selected for use in this study represent actual and potential epidemic vectors of VSV. V920 vaccine inoculated into Cx. quinquefasciatus and Ae. aegypti mosquitoes demonstrated persistence of replication-competent virus following inoculation, consistent with the recognized biological stability of the vaccine, but no evidence for active virus replication in live mosquitoes was observed. Following administration of an infectious blood meal to Ae. aegypti and Cx. quinquefasciatus mosquitoes at a titer several log10 PFU more concentrated than would be observed in vaccinated individuals, no infection or dissemination of V920 was observed in either mosquito species. In vitro and in vivo data gathered during this study support minimal risk of the vector-borne potential of the V920 vaccine.
Collapse
Affiliation(s)
- Nicholas A Bergren
- a Arthropod-borne Infectious Diseases Laboratory, Department of Microbiology , Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Fort Collins , CO , USA
| | - Megan R Miller
- a Arthropod-borne Infectious Diseases Laboratory, Department of Microbiology , Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Fort Collins , CO , USA
| | - Thomas P Monath
- b BioProtection Systems, New Link Genetics Corporation , Ames , IA , USA
| | - Rebekah C Kading
- a Arthropod-borne Infectious Diseases Laboratory, Department of Microbiology , Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Fort Collins , CO , USA
| |
Collapse
|
182
|
|
183
|
Lambe T, Bowyer G, Ewer KJ. A review of Phase I trials of Ebola virus vaccines: what can we learn from the race to develop novel vaccines? Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0295. [PMID: 28396468 PMCID: PMC5394635 DOI: 10.1098/rstb.2016.0295] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2016] [Indexed: 11/23/2022] Open
Abstract
Sporadic outbreaks of Ebola virus infection have been documented since the mid-Seventies and viral exposure can lead to lethal haemorrhagic fever with case fatalities as high as 90%. There is now a comprehensive body of data from both ongoing and completed clinical trials assessing various vaccine strategies, which were rapidly advanced through clinical trials in response to the 2013–2016 Ebola virus disease (EVD) public health emergency. Careful consideration of immunogenicity post vaccination is essential but has been somewhat stifled because of the wide array of immunological assays and outputs that have been used in the numerous clinical trials. We discuss here the different aspects of the immune assays currently used in the Phase I clinical trials for Ebola virus vaccines, and draw comparisons across the immune outputs where possible; various trials have examined both cellular and humoral immunity in European and African cohorts. Assessment of the safety data, the immunological outputs and the ease of field deployment for the various vaccine modalities will help both the scientific community and policy-makers prioritize and potentially license vaccine candidates. If this can be achieved, the next outbreak of Ebola virus, or other emerging pathogen, can be more readily contained and will not have such widespread and devastating consequences. This article is part of the themed issue ‘The 2013–2016 West African Ebola epidemic: data, decision-making and disease control’.
Collapse
Affiliation(s)
- Teresa Lambe
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Georgina Bowyer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
184
|
Gomes MF, de la Fuente-Núñez V, Saxena A, Kuesel AC. Protected to death: systematic exclusion of pregnant women from Ebola virus disease trials. Reprod Health 2017; 14:172. [PMID: 29297366 PMCID: PMC5751665 DOI: 10.1186/s12978-017-0430-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND For 30 years, women have sought equal opportunity to be included in trials so that drugs are equitably studied in women as well as men; regulatory guidelines have changed accordingly. Pregnant women, however, continue to be excluded from trials for non-obstetric conditions, though they have been included for trials of life-threatening diseases because prospects for maternal survival outweighed potential fetal risks. Ebola virus disease is a life-threatening infection without approved treatments or vaccines. Previous Ebola virus (EBOV) outbreak data showed 89-93% maternal and 100% fetal/neonatal mortality. Early in the 2013-2016 EBOV epidemic, an expert panel pointed to these high mortality rates and the need to prioritize and preferentially allocate unregistered interventions in favor of pregnant women (and children). Despite these recommendations and multiple ethics committee requests for their inclusion on grounds of justice, equity, and medical need, pregnant women were excluded from all drug and vaccine trials in the affected countries, either without justification or on grounds of potential fetal harm. An opportunity to offer pregnant women the same access to potentially life-saving interventions as others, and to obtain data to inform their future use, was lost. Once again, pregnant women were denied autonomy and their right to decide. CONCLUSION We recommend that, without clear justification for exclusion, pregnant women are included in clinical trials for EBOV and other life-threatening conditions, with lay language on risks and benefits in information documents, so that pregnant women can make their own decision to participate. Their automatic exclusion from trials for other conditions should be questioned.
Collapse
Affiliation(s)
| | | | - Abha Saxena
- Department for Information Evidence and Research, World Health Organization, Geneva, Switzerland
| | - Annette C. Kuesel
- UNICEF/UNDP/World Bank/WHO Special Programme for Research and Training in Tropical Diseases, World Health Organization, Geneva, Switzerland
| |
Collapse
|
185
|
Jin Y, Lei C, Hu D, Dimitrov DS, Ying T. Human monoclonal antibodies as candidate therapeutics against emerging viruses. Front Med 2017; 11:462-470. [PMID: 29159596 PMCID: PMC7088856 DOI: 10.1007/s11684-017-0596-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 10/10/2017] [Indexed: 12/22/2022]
Abstract
The emergence of new pathogens, such as severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and Ebola virus, poses serious challenges to global public health and highlights the urgent need for novel antiviral approaches. Monoclonal antibodies (mAbs) have been successfully used to treat various diseases, particularly cancer and immunological disorders. Antigen-specific mAbs have been isolated using several different approaches, including hybridoma, transgenic mice, phage display, yeast display, and single B-cell isolation. Consequently, an increasing number of mAbs, which exhibit high potency against emerging viruses in vitro and in animal models of infection, have been developed. In this paper, we summarize historical trends and recent developments in mAb discovery, compare the advantages and disadvantages of various approaches to mAb production, and discuss the potential use of such strategies for the development of antivirals against emerging diseases. We also review the application of recently developed human mAbs against SARS-CoV, MERS-CoV, and Ebola virus and discuss prospects for the development of mAbs as therapeutic agents against emerging viral diseases.
Collapse
Affiliation(s)
- Yujia Jin
- Key Laboratory of Medical Molecular Virology of the Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Cheng Lei
- Key Laboratory of Medical Molecular Virology of the Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Dan Hu
- Key Laboratory of Medical Molecular Virology of the Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Dimiter S Dimitrov
- Protein Interactions Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA.
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology of the Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
186
|
Singh RK, Dhama K, Malik YS, Ramakrishnan MA, Karthik K, Khandia R, Tiwari R, Munjal A, Saminathan M, Sachan S, Desingu PA, Kattoor JJ, Iqbal HMN, Joshi SK. Ebola virus - epidemiology, diagnosis, and control: threat to humans, lessons learnt, and preparedness plans - an update on its 40 year's journey. Vet Q 2017; 37:98-135. [PMID: 28317453 DOI: 10.1080/01652176.2017.1309474] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Ebola virus (EBOV) is an extremely contagious pathogen and causes lethal hemorrhagic fever disease in man and animals. The recently occurred Ebola virus disease (EVD) outbreaks in the West African countries have categorized it as an international health concern. For the virus maintenance and transmission, the non-human primates and reservoir hosts like fruit bats have played a vital role. For curbing the disease timely, we need effective therapeutics/prophylactics, however, in the absence of any approved vaccine, timely diagnosis and monitoring of EBOV remains of utmost importance. The technologically advanced vaccines like a viral-vectored vaccine, DNA vaccine and virus-like particles are underway for testing against EBOV. In the absence of any effective control measure, the adaptation of high standards of biosecurity measures, strict sanitary and hygienic practices, strengthening of surveillance and monitoring systems, imposing appropriate quarantine checks and vigilance on trade, transport, and movement of visitors from EVD endemic countries remains the answer of choice for tackling the EBOV spread. Herein, we converse with the current scenario of EBOV giving due emphasis on animal and veterinary perspectives along with advances in diagnosis and control strategies to be adopted, lessons learned from the recent outbreaks and the global preparedness plans. To retrieve the evolutionary information, we have analyzed a total of 56 genome sequences of various EBOV species submitted between 1976 and 2016 in public databases.
Collapse
Affiliation(s)
- Raj Kumar Singh
- a ICAR-Indian Veterinary Research Institute , Bareilly , India
| | - Kuldeep Dhama
- b Division of Pathology, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | - Yashpal Singh Malik
- c Division of Biological Standardization, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | | | - Kumaragurubaran Karthik
- e Divison of Bacteriology and Mycology, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | - Rekha Khandia
- f Department of Biochemistry and Genetics , Barkatullah University , Bhopal , India
| | - Ruchi Tiwari
- g Department of Veterinary Microbiology and Immunology , College of Veterinary Sciences, Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan (DUVASU) , Mathura , India
| | - Ashok Munjal
- f Department of Biochemistry and Genetics , Barkatullah University , Bhopal , India
| | - Mani Saminathan
- b Division of Pathology, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | - Swati Sachan
- h Immunology Section, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | | | - Jobin Jose Kattoor
- c Division of Biological Standardization, ICAR-Indian Veterinary Research Institute , Bareilly , India
| | - Hafiz M N Iqbal
- i School of Engineering and Science, Tecnologico de Monterrey , Monterrey , Mexico
| | - Sunil Kumar Joshi
- j Cellular Immunology Lab , Frank Reidy Research Center for Bioelectrics , School of Medical Diagnostics & Translational Sciences, Old Dominion University , Norfolk , VA , USA
| |
Collapse
|
187
|
Ebola virus disease: an update on post-exposure prophylaxis. THE LANCET. INFECTIOUS DISEASES 2017; 18:e183-e192. [PMID: 29153266 DOI: 10.1016/s1473-3099(17)30677-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/20/2017] [Accepted: 10/03/2017] [Indexed: 12/11/2022]
Abstract
The massive outbreak of Ebola virus disease in west Africa between 2013 and 2016 resulted in intense efforts to evaluate the efficacy of several specific countermeasures developed through years of preclinical work, including the first clinical trials for therapeutics and vaccines. In this Review, we discuss how the experience and data generated from that outbreak have helped to advance the understanding of the use of these countermeasures for post-exposure prophylaxis against Ebola virus infection. In future outbreaks, post-exposure prophylaxis could play an important part in reducing community transmission of Ebola virus by providing more immediate protection than does immunisation as well as providing additional protection for health-care workers who are inadvertently exposed over the course of their work. We propose provisional guidance for use of post-exposure prophylaxis in Ebola virus disease and identify the priorities for future preparedness and further research.
Collapse
|
188
|
Affiliation(s)
- Francis A Plummer
- Department of Medical Microbiology (Plummer), University of Manitoba College of Medicine, Winnipeg, Man.; Cognoveritas Consulting (Jones), Winnipeg, Man.
| | - Steven M Jones
- Department of Medical Microbiology (Plummer), University of Manitoba College of Medicine, Winnipeg, Man.; Cognoveritas Consulting (Jones), Winnipeg, Man
| |
Collapse
|
189
|
Speranza E, Connor JH. Host Transcriptional Response to Ebola Virus Infection. Vaccines (Basel) 2017; 5:E30. [PMID: 28930167 PMCID: PMC5620561 DOI: 10.3390/vaccines5030030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/12/2017] [Accepted: 09/12/2017] [Indexed: 01/09/2023] Open
Abstract
Ebola virus disease (EVD) is a serious illness that causes severe disease in humans and non-human primates (NHPs) and has mortality rates up to 90%. EVD is caused by the Ebolavirus and currently there are no licensed therapeutics or vaccines to treat EVD. Due to its high mortality rates and potential as a bioterrorist weapon, a better understanding of the disease is of high priority. Multiparametric analysis techniques allow for a more complete understanding of a disease and the host response. Analysis of RNA species present in a sample can lead to a greater understanding of activation or suppression of different states of the immune response. Transcriptomic analyses such as microarrays and RNA-Sequencing (RNA-Seq) have been important tools to better understand the global gene expression response to EVD. In this review, we outline the current knowledge gained by transcriptomic analysis of EVD.
Collapse
Affiliation(s)
- Emily Speranza
- Department of Microbiology, Bioinformatics Program, National Emerging Infectious Disease Laboratories, Boston University, Boston, MA 02118, USA.
| | - John H Connor
- Department of Microbiology, Bioinformatics Program, National Emerging Infectious Disease Laboratories, Boston University, Boston, MA 02118, USA.
| |
Collapse
|
190
|
Racine T, Kobinger GP, Arts EJ. Development of an HIV vaccine using a vesicular stomatitis virus vector expressing designer HIV-1 envelope glycoproteins to enhance humoral responses. AIDS Res Ther 2017; 14:55. [PMID: 28893277 PMCID: PMC5594459 DOI: 10.1186/s12981-017-0179-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 08/22/2017] [Indexed: 11/30/2022] Open
Abstract
Vesicular stomatitis virus (VSV), like many other Rhabdoviruses, have become the focus of intense research over the past couple of decades based on their suitability as vaccine vectors, transient gene delivery systems, and as oncolytic viruses for cancer therapy. VSV as a vaccine vector platform has multiple advantages over more traditional viral vectors including low level, non-pathogenic replication in diverse cell types, ability to induce both humoral and cell-mediate immune responses, and the remarkable expression of foreign proteins cloned into multiple intergenic sites in the VSV genome. The utility and safety of VSV as a vaccine vector was recently demonstrated near the end of the recent Ebola outbreak in West Africa where VSV pseudotyped with the Ebola virus (EBOV) glycoprotein was proven safe in humans and provided protective efficacy against EBOV in a human phase III clinical trial. A team of Canadian scientists, led by Dr. Gary Kobinger, is now working with International AIDS Vaccine Initiative (IAVI) in developing a VSV-based HIV vaccine that will combine unique Canadian research on the HIV-1 Env glycoprotein and on the VSV vaccine vector. The goal of this collaboration is to develop a vaccine with a robust and potent anti-HIV immune response with an emphasis on generating quality antibodies to protect against HIV challenges.
Collapse
|
191
|
Dutta P, Halder AK, Basu S, Kundu M. A survey on Ebola genome and current trends in computational research on the Ebola virus. Brief Funct Genomics 2017; 17:374-380. [DOI: 10.1093/bfgp/elx020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
192
|
Windows of opportunity for Ebola virus infection treatment and vaccination. Sci Rep 2017; 7:8975. [PMID: 28827623 PMCID: PMC5567060 DOI: 10.1038/s41598-017-08884-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/19/2017] [Indexed: 12/23/2022] Open
Abstract
Ebola virus (EBOV) infection causes a high death toll, killing a high proportion of EBOV-infected patients within 7 days. Comprehensive data on EBOV infection are fragmented, hampering efforts in developing therapeutics and vaccines against EBOV. Under this circumstance, mathematical models become valuable resources to explore potential controlling strategies. In this paper, we employed experimental data of EBOV-infected nonhuman primates (NHPs) to construct a mathematical framework for determining windows of opportunity for treatment and vaccination. Considering a prophylactic vaccine based on recombinant vesicular stomatitis virus expressing the EBOV glycoprotein (rVSV-EBOV), vaccination could be protective if a subject is vaccinated during a period from one week to four months before infection. For the case of a therapeutic vaccine based on monoclonal antibodies (mAbs), a single dose might resolve the invasive EBOV replication even if it was administrated as late as four days after infection. Our mathematical models can be used as building blocks for evaluating therapeutic and vaccine modalities as well as for evaluating public health intervention strategies in outbreaks. Future laboratory experiments will help to validate and refine the estimates of the windows of opportunity proposed here.
Collapse
|
193
|
Vujadinovic M, Wunderlich K, Callendret B, Koning M, Vermeulen M, Sanders B, van der Helm E, Gecgel A, Spek D, de Boer K, Stalknecht M, Serroyen J, Grazia Pau M, Schuitemaker H, Zahn R, Custers J, Vellinga J. Adenoviral Type 35 and 26 Vectors with a Bidirectional Expression Cassette in the E1 Region Show an Improved Genetic Stability Profile and Potent Transgene-Specific Immune Response. Hum Gene Ther 2017; 29:337-351. [PMID: 28816084 DOI: 10.1089/hum.2017.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Genetic vaccines based on replication-incompetent adenoviral (AdV) vectors are currently in clinical development. Monovalent AdV vectors express one antigen from an expression cassette placed in most cases in the E1 region. For many vaccines, inclusion of several antigens is necessary in order to raise protective immunity and/or target more than one pathogen or pathogen strain. On the basis of the current technology, a mix of several monovalent vectors can be employed. However, a mix of the standard monovalent AdV vectors may not be optimal with respect to manufacturing costs and the final dose per vector in humans. Alternatively, a variety of bivalent recombinant AdV vector approaches is described in the literature. It remains unclear whether all strategies are equally suitable for clinical development while preserving all the beneficial properties of the monovalent AdV (e.g., immunogenic potency). Therefore, a thorough assessment of different bivalent AdV strategies was performed in a head-to-head fashion compared with the monovalent benchmark. The vectors were tested for rescue efficiency, genetic stability, transgene expression, and potency to induce transgene-specific immune responses. We report that the vector expressing multiple antigens from a bidirectional expression cassette in E1 shows a better genetic stability profile and a potent transgene-specific immune response compared with the other tested bivalent vectors.
Collapse
Affiliation(s)
- Marija Vujadinovic
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Kerstin Wunderlich
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Benoit Callendret
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Marina Koning
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Mark Vermeulen
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Barbara Sanders
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Esmeralda van der Helm
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Adile Gecgel
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Dirk Spek
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Karin de Boer
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Masha Stalknecht
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jan Serroyen
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Maria Grazia Pau
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Hanneke Schuitemaker
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Roland Zahn
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jerome Custers
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jort Vellinga
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| |
Collapse
|
194
|
Vesicular Stomatitis Virus Pseudotyped with Ebola Virus Glycoprotein Serves as a Protective, Noninfectious Vaccine against Ebola Virus Challenge in Mice. J Virol 2017; 91:JVI.00479-17. [PMID: 28615211 DOI: 10.1128/jvi.00479-17] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/07/2017] [Indexed: 11/20/2022] Open
Abstract
The recent Ebola virus (EBOV) epidemic in West Africa demonstrates the potential for a significant public health burden caused by filoviral infections. No vaccine or antiviral is currently FDA approved. To expand the vaccine options potentially available, we assessed protection conferred by an EBOV vaccine composed of vesicular stomatitis virus pseudovirions that lack native G glycoprotein (VSVΔG) and bear EBOV glycoprotein (GP). These pseudovirions mediate a single round of infection. Both single-dose and prime/boost vaccination regimens protected mice against lethal challenge with mouse-adapted Ebola virus (ma-EBOV) in a dose-dependent manner. The prime/boost regimen provided significantly better protection than a single dose. As N-linked glycans are thought to shield conserved regions of the EBOV GP receptor-binding domain (RBD), thereby blocking epitopes within the RBD, we also tested whether VSVΔG bearing EBOV GPs that lack GP1 N-linked glycans provided effective immunity against challenge with ma-EBOV or a more distantly related virus, Sudan virus. Using a prime/boost strategy, high doses of GP/VSVΔG partially or fully denuded of N-linked glycans on GP1 protected mice against ma-EBOV challenge, but these mutants were no more effective than wild-type (WT) GP/VSVΔG and did not provide cross protection against Sudan virus. As reported for other EBOV vaccine platforms, the protection conferred correlated with the quantity of EBOV GP-specific Ig produced but not with the production of neutralizing antibodies. Our results show that EBOV GP/VSVΔG pseudovirions serve as a successful vaccination platform in a rodent model of Ebola virus disease and that GP1 N-glycan loss does not influence immunogenicity or vaccination success.IMPORTANCE The West African Ebola virus epidemic was the largest to date, with more than 28,000 people infected. No FDA-approved vaccines are yet available, but in a trial vaccination strategy in West Africa, recombinant, infectious VSV encoding the Ebola virus glycoprotein effectively prevented virus-associated disease. VSVΔG pseudovirion vaccines may prove as efficacious and have better safety, but they have not been tested to date. Thus, we tested the efficacy of VSVΔG pseudovirions bearing Ebola virus glycoprotein as a vaccine platform. We found that wild-type Ebola virus glycoprotein, in the context of this platform, provides robust protection of EBOV-challenged mice. Further, we found that removal of the heavy glycan shield surrounding conserved regions of the glycoprotein does not enhance vaccine efficacy.
Collapse
|
195
|
Venkatraman N, Silman D, Folegatti PM, Hill AVS. Vaccines against Ebola virus. Vaccine 2017; 36:5454-5459. [PMID: 28780120 DOI: 10.1016/j.vaccine.2017.07.054] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/15/2017] [Accepted: 07/17/2017] [Indexed: 11/29/2022]
Abstract
We have just witnessed the largest and most devastating outbreak of Ebola virus disease, which highlighted the urgent need for development of an efficacious vaccine that could be used to curtail future outbreaks. Prior to 2014, there had been limited impetus worldwide to develop a vaccine since the virus was first discovered in 1976. Though too many lives were lost during this outbreak, it resulted in the significantly accelerated clinical development of a number of candidate vaccines through an extraordinary collaborative global effort coordinated by the World Health Organisation (WHO) and involving a number of companies, trial centres, funders, global stakeholders and agencies. We have acquired substantial safety and immunogenicity data on a number of vaccines in Caucasian and African populations. The rapid pace of events led to the initiation of the landmark efficacy trial testing the rVSV-vectored vaccine, which showed high level efficacy in an outbreak setting when deployed using an innovative ring vaccination strategy. Though the Public Health Emergency of International Concern (PHEIC) declared by the WHO has now been lifted, the global scientific community faces numerous challenges ahead to ensure that there is a licensed, deployable vaccine available for use in future outbreaks for at least the Zaire and Sudan strains of Ebola virus. There remain several unanswered questions on the durability of protection, mechanistic immunological correlates and preferred deployment strategies. This review outlines a brief history of the development of Ebola vaccines, the significant progress made since the scale of the outbreak became apparent, some lessons learnt and how they could shape future development of vaccines and the management of similar outbreaks.
Collapse
Affiliation(s)
- Navin Venkatraman
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom.
| | - Daniel Silman
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Pedro M Folegatti
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Adrian V S Hill
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
196
|
Abstract
The filoviruses, Ebola virus (EBOV), and Marburg virus (MARV), are among the most pathogenic viruses known to man and the causative agents of viral hemorrhagic fever outbreaks in Africa with case fatality rates of up to 90%. Nearly 30,000 infections were observed in the latest EBOV epidemic in West Africa; previous outbreaks were much smaller, typically only affecting less than a few hundred people. Compared to other diseases such as AIDS or Malaria with millions of cases annually, filovirus hemorrhagic fever (FHF) is one of the neglected infectious diseases. There are no licensed vaccines or therapeutics available to treat EBOV and MARV infections; therefore, these pathogens can only be handled in maximum containment laboratories and are classified as select agents. Under these limitations, a very few laboratories worldwide conducted basic research and countermeasure development for EBOV and MARV since their respective discoveries in 1967 (MARV) and 1976 (EBOV). In this review, we discuss several vaccine platforms against EBOV and MARV, which have been assessed for their protective efficacy in animal models of FHF. The focus is on the most promising approaches, which were accelerated in clinical development (phase I-III trials) during the EBOV epidemic in West Africa.
Collapse
Affiliation(s)
- Pierce Reynolds
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| |
Collapse
|
197
|
ElSherif MS, Brown C, MacKinnon-Cameron D, Li L, Racine T, Alimonti J, Rudge TL, Sabourin C, Silvera P, Hooper JW, Kwilas SA, Kilgore N, Badorrek C, Ramsey WJ, Heppner DG, Kemp T, Monath TP, Nowak T, McNeil SA, Langley JM, Halperin SA. Assessing the safety and immunogenicity of recombinant vesicular stomatitis virus Ebola vaccine in healthy adults: a randomized clinical trial. CMAJ 2017. [PMID: 28630358 DOI: 10.1503/cmaj.170074] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND The 2013-2016 Ebola virus outbreak in West Africa was the most widespread in history. In response, alive attenuated recombinant vesicular stomatitis virus (rVSV) vaccine expressing Zaire Ebolavirus glycoprotein (rVSVΔG-ZEBOV-GP) was evaluated in humans. METHODS In a phase 1, randomized, dose-ranging, observer-blind, placebo-controlled trial, healthy adults aged 18-65 years were randomized into 4 groups of 10 to receive one of 3 vaccine doses or placebo. Follow-up visits spanned 180 days postvaccination for safety monitoring, immunogenicity testing and any rVSV virus shedding. RESULTS Forty participants were injected with rVSVΔG-ZEBOV-GP vaccine (n = 30) or saline placebo (n = 10). No serious adverse events related to the vaccine or participant withdrawals were reported. Solicited adverse events during the 14-day follow-up period were mild to moderate and self-limited, with the exception of injection-site pain and headache. Viremia following vaccination was transient and no longer detectable after study day 3, with no virus shedding in saliva or urine. All vaccinated participants developed serum immunoglobulin G (IgG), as measured by Ebola virus envelope glycoprotein-based enzyme-linked immunosorbent assay (ELISA). Immunogenicity was comparable across all dose groups, and sustained IgG titers were detectable through to the last visit, at study day 180. INTERPRETATION In this phase 1 study, there were no safety concerns after a single dose of rVSVΔG-ZEBOV-GP vaccine. IgG ELISA showed persistent high titers at 180 days postimmunization. There was a period of reactogenicity, but in general, the vaccine was well tolerated. This study provides evidence of the safety and immunogenicity of rVSVΔG-ZEBOV-GP vaccine and importance of its further investigation. Trial registration: Clinical-Trials.gov no., NCT02374385.
Collapse
Affiliation(s)
- May S ElSherif
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Catherine Brown
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Donna MacKinnon-Cameron
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Li Li
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Trina Racine
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Judie Alimonti
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Thomas L Rudge
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Carol Sabourin
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Peter Silvera
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Jay W Hooper
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Steven A Kwilas
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Nicole Kilgore
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Christopher Badorrek
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - W Jay Ramsey
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - D Gray Heppner
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Tracy Kemp
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Thomas P Monath
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Teresa Nowak
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Shelly A McNeil
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Joanne M Langley
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass
| | - Scott A Halperin
- Canadian Center for Vaccinology (ElSherif, Brown, MacKinnon-Cameron, Li, McNeil, Langley, Halperin), IWK Health Centre and Nova Scotia Health Authority, Dalhousie University, Halifax, NS; National Microbiology Laboratory (Racine, Alimonti), Winnipeg, Man.; Battelle Biomedical Research Center (Rudge, Sabourin), Columbus, Ohio; United States Army Medical Research Institute of Infectious Disease (Silvera, Hooper, Kwilas), Fort Detrick, Md.; Joint Program Executive Office for Chemical and Biological Defense Medical Countermeasure Systems' Joint Vaccine Acquisition Program (Kilgore, Badorrek), Fort Detrick, Md.; BioProtection Systems/NewLink Genetics Corporation (Ramsey, Heppner, Kemp, Monath), Ames, Iowa; Veristat LLC (Nowak), Southborough, Mass.
| | | |
Collapse
|
198
|
Alirol E, Kuesel AC, Guraiib MM, dela Fuente-Núñez V, Saxena A, Gomes MF. Ethics review of studies during public health emergencies - the experience of the WHO ethics review committee during the Ebola virus disease epidemic. BMC Med Ethics 2017; 18:43. [PMID: 28651650 PMCID: PMC5485606 DOI: 10.1186/s12910-017-0201-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 06/08/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Between 2013 and 2016, West Africa experienced the largest ever outbreak of Ebola Virus Disease. In the absence of registered treatments or vaccines to control this lethal disease, the World Health Organization coordinated and supported research to expedite identification of interventions that could control the outbreak and improve future control efforts. Consequently, the World Health Organization Research Ethics Review Committee (WHO-ERC) was heavily involved in reviews and ethics discussions. It reviewed 24 new and 22 amended protocols for research studies including interventional (drug, vaccine) and observational studies. WHO-ERC REVIEWS WHO-ERC provided the reviews within on average 6 working days. The WHO-ERC often could not provide immediate approval of protocols for reasons which were not Ebola Virus Disease specific but related to protocol inconsistencies, missing information and complex informed consents. WHO-ERC considerations on Ebola Virus Disease specific issues (benefit-risk assessment, study design, exclusion of pregnant women and children from interventional studies, data and sample sharing, collaborative partnerships including international and local researchers and communities, community engagement and participant information) are presented. CONCLUSIONS To accelerate study approval in future public health emergencies, we recommend: (1) internally consistent and complete submissions with information documents in language participants are likely to understand, (2) close collaboration between local and international researchers from research inception, (3) generation of template agreements for data and sample sharing and use during the ongoing global consultations on bio-banks, (4) formation of Joint Scientific Advisory and Data Safety Review Committees for all studies linked to a particular intervention or group of interventions, (5) formation of a Joint Ethics Review Committee with representatives of the Ethics Committees of all institutions and countries involved to strengthen reviews through the different perspectives provided without the 'opportunity costs' for time to final approval of multiple, independent reviews, (6) direct information exchange between the chairs of advisory, safety review and ethics committees, (7) more Ethics Committee support for investigators than is standard and (8) a global consultation on criteria for inclusion of pregnant women and children in interventional studies for conditions which put them at particularly high risk of mortality or other irreversible adverse outcomes under standard-of-care.
Collapse
Affiliation(s)
- Emilie Alirol
- Global Antibiotics Research and Development Partnership (GARDP), Drugs for Neglected Diseases initiative (DNDi), 15 chemin Louis Dunant, 1202 Geneva, Switzerland
| | - Annette C. Kuesel
- World Health Organization, UNICEF/UNDP/World Bank/WHO Special Programme for Research and Training in Tropical Diseases, 20 Avenue Appia, 1211, 27 Geneva, Switzerland
| | - Maria Magdalena Guraiib
- World Health Organization, Department for Information Evidence and Research, 20 Avenue Appia, 1211, 27 Geneva, Switzerland
| | - Vânia dela Fuente-Núñez
- World Health Organization, Department for Information Evidence and Research, 20 Avenue Appia, 1211, 27 Geneva, Switzerland
| | - Abha Saxena
- World Health Organization, Department for Information Evidence and Research, 20 Avenue Appia, 1211, 27 Geneva, Switzerland
| | - Melba F. Gomes
- World Health Organization, 20 Avenue Appia, 1211, 27 Geneva, Switzerland
| |
Collapse
|
199
|
Coller BAG, Blue J, Das R, Dubey S, Finelli L, Gupta S, Helmond F, Grant-Klein RJ, Liu K, Simon J, Troth S, VanRheenen S, Waterbury J, Wivel A, Wolf J, Heppner DG, Kemp T, Nichols R, Monath TP. Clinical development of a recombinant Ebola vaccine in the midst of an unprecedented epidemic. Vaccine 2017. [PMID: 28647166 DOI: 10.1016/j.vaccine.2017.05.097] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The 2014-2016 Ebola outbreak caused over 28,000 cases and 11,000 deaths. Merck & Co. Inc., Kenilworth, NJ USA and NewLink Genetics are working with private and public partners to develop and license an Ebola vaccine that was evaluated extensively during the outbreak. The vaccine referred to as V920 is a recombinant vesicular stomatitis virus (rVSV) in which the VSV-G envelope glycoprotein (GP) is completely replaced by the Zaire ebolavirus GP (rVSVΔG-ZEBOV-GP). Eight Phase I and four Phase II/III clinical trials enrolling approximately 17,000 subjects were conducted in parallel to the outbreak to assess the safety, immunogenicity, and/or efficacy of V920. Immunogenicity data demonstrate that anti-GP antibodies are generally detectable by ELISA by 14days postvaccination with up to 100% seroconversion observed by 28days post dose. In addition, the results of a ring vaccination trial conducted by the WHO and their partners in Guinea suggest robust vaccine efficacy within 10days of receipt of a single dose of vaccine. The vaccine is generally well-tolerated when administered to healthy, non-pregnant adults. The development of this vaccine candidate in the context of this unprecedented epidemic has involved the close cooperation of large number of international partners and highlights what we as a public health community can accomplish when working together towards a common goal. Study identification: V920-001 to V920-012. CLINICALTRIALS.GOV identifiers: NCT02269423; NCT02280408; NCT02374385; NCT02314923; NCT02287480; NCT02283099; NCT02296983; NCT02344407; NCT02378753; NCT02503202.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - D Gray Heppner
- NewLink Genetics, Inc./BioProtection Systems, Ames, IA, USA
| | - Tracy Kemp
- NewLink Genetics, Inc./BioProtection Systems, Ames, IA, USA
| | - Rick Nichols
- NewLink Genetics, Inc./BioProtection Systems, Ames, IA, USA
| | | |
Collapse
|
200
|
Heppner DG, Kemp TL, Martin BK, Ramsey WJ, Nichols R, Dasen EJ, Link CJ, Das R, Xu ZJ, Sheldon EA, Nowak TA, Monath TP. Safety and immunogenicity of the rVSV∆G-ZEBOV-GP Ebola virus vaccine candidate in healthy adults: a phase 1b randomised, multicentre, double-blind, placebo-controlled, dose-response study. THE LANCET. INFECTIOUS DISEASES 2017; 17:854-866. [PMID: 28606591 DOI: 10.1016/s1473-3099(17)30313-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND The 2014 Zaire Ebola virus outbreak highlighted the need for a safe, effective vaccine with a rapid onset of protection. We report the safety and immunogenicity of the recombinant vesicular stomatitis virus-Zaire Ebola virus envelope glycoprotein vaccine (rVSV∆G-ZEBOV-GP) across a 6 log10 dose range in two sequential cohorts. METHODS In this phase 1b double-blind, placebo-controlled, dose-response study we enrolled and randomly assigned healthy adults (aged 18-61 years) at eight study sites in the USA to receive a single injection of vaccine or placebo, administered by intramuscular injection. In cohort 1, participants were assigned to receive 3 × 103, 3 × 104, 3 × 105, or 3 × 106 PFU doses of rVSV∆G-ZEBOV-GP or placebo. In cohort 2, participants were assigned to receive 3 × 106, 9 × 106, 2 × 107, or 1 × 108 PFU doses of rVSV∆G-ZEBOV-GP or placebo. Participants were centrally allocated by the study statistician to vaccine groups or placebo through computer-generated randomisation lists. The primary safety outcome was incidence of adverse events within 14 days in the modified intention-to-treat population (all randomly assigned participants who received vaccine or placebo), and the primary outcome for immunogenicity was IgG ELISA antibody titres at day 28 in the per-protocol population. Surveillance was enhanced for arthritis and dermatitis through to day 56. This study is registered with ClinicalTrials.gov, number NCT02314923. FINDINGS Between Dec 26, 2014, and June 8, 2015, 513 participants were enrolled and randomly assigned; one was not immunised because of unsuccessful phlebotomy. In cohort 1, 256 participants received vaccine (3 × 103 [n=64], 3 × 104 [n=64], 3 × 105 [n=64], or 3 × 106 PFU [n=64]) and 74 received placebo. In cohort 2, 162 participants received vaccine (3 × 106 [n=20], 9 × 106 [n=47], 2 × 107 [n=47], or 1 × 108 PFU [n=48]) and 20 received placebo. Most adverse events occurred in the first day after vaccination, and were mild to moderate in intensity, of a short duration, and more frequent at high vaccine doses (9 × 106 PFU and greater). At the 2 × 107 PFU dose (used in phase 3 trials), the most common local adverse events versus placebo within the first 14 days were arm pain (57·4% [27 of 47] vs 7·4% [seven of 94]) and local tenderness (59·6% [28 of 47] vs 8·5% [eight of 94]). The most common systemic adverse events at the 2 × 107 PFU dose versus placebo, occurring in the first 14 days, were headache (46·8% [22 of 47] vs 27·7% [26 of 94]), fatigue (38·3% [18 of 47] vs 19·1% [18 of 94]), myalgia (34·0% [16 of 47] vs 10·6% [10 of 94]), subjective fever (29·8% [14 of 47] vs 2·1% [two of 94]), shivering or chills (27·7% [13 of 47] vs 7·4% [seven of 94]), sweats (23·4% [11 of 47] vs 3·2% [three of 94]), joint aches and pain (19·1% [nine of 47] vs 7·4% [seven of 94]), objective fever (14·9% [seven of 47] vs 1·1% [one of 94]), and joint tenderness or swelling (14·9% [seven of 47] vs 2·1% [two of 94]). Self-limited, post-vaccination arthritis occurred in 4·5% (19 of 418) of vaccinees (median onset 12·0 days [IQR 10-14]; median duration 8·0 days [6-15]) versus 3·2% (three of 94) of controls (median onset 15·0 days [6-20]; median duration 47·0 days [37-339]), with no apparent dose relationship. Post-vaccination dermatitis occurred in 5·7% (24 of 418) of vaccinees (median onset 9·0 days [IQR 2-12]; median duration 7·0 days [4-9]) versus 3·2% (three of 94) of controls (median onset 5·0 days [3-53]; median duration 33·0 days [5-370]). A low-level, transient, dose-dependent viraemia occurred in concert with early reactogenicity. Antibody responses were observed in most participants by day 14. IgG and neutralising antibody titres were dose-related (p=0·0003 for IgG ELISA and p<0·0001 for the 60% plaque-reduction neutralisation test [PRNT60] by linear trend). On day 28 at the 2 × 107 PFU dose, the geometric mean IgG ELISA endpoint titre was 1624 (95% CI 1146-2302) and seroconversion was 95·7% (95% CI 85·5-98·8); the geometric mean neutralising antibody titre by PRNT60 was 250 (176-355) and seroconversion was 95·7% (85·5-98·8). These robust immunological responses were sustained for 1 year. INTERPRETATION rVSV∆G-ZEBOV-GP was well tolerated and stimulated a rapid onset of binding and neutralising antibodies, which were maintained through to day 360. The immunogenicity results support selection of the 2 × 107 PFU dose. FUNDING Biomedical Advanced Research and Development Authority, US Department of Health and Human Services.
Collapse
Affiliation(s)
- D Gray Heppner
- Bioprotection Systems, NewLink Genetics, Ames, IA, USA; Devens, MA, USA.
| | - Tracy L Kemp
- Bioprotection Systems, NewLink Genetics, Ames, IA, USA; Devens, MA, USA
| | - Brian K Martin
- Bioprotection Systems, NewLink Genetics, Ames, IA, USA; Devens, MA, USA
| | - William J Ramsey
- Bioprotection Systems, NewLink Genetics, Ames, IA, USA; Devens, MA, USA
| | - Richard Nichols
- Bioprotection Systems, NewLink Genetics, Ames, IA, USA; Devens, MA, USA
| | - Emily J Dasen
- Bioprotection Systems, NewLink Genetics, Ames, IA, USA; Devens, MA, USA
| | - Charles J Link
- Bioprotection Systems, NewLink Genetics, Ames, IA, USA; Devens, MA, USA
| | | | | | | | | | - Thomas P Monath
- Bioprotection Systems, NewLink Genetics, Ames, IA, USA; Devens, MA, USA
| | | |
Collapse
|