1
|
Lacabaratz C, Durand M, Wiedemann A, Foucat E, Surénaud M, Krief C, Guillaumat L, Robinson C, Luhn K, Bockstal V, Thiébaut R, Richert L, Lévy Y. Innate and Cellular Immune Response to the Ebola Vaccine Ad26.ZEBOV, MVA-BN-Filo: An Ancillary Study of the EBL2001 Phase 2 Trial. J Infect Dis 2025; 231:230-240. [PMID: 39012798 DOI: 10.1093/infdis/jiae360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND The EBL2001 phase 2 trial tested the 2-dose Ad26.ZEBOV, MVA-BN-Filo Ebola vaccine in Europe. Safety and humoral immunogenicity assessments led to European Union market authorization in 2020. Complementary analyses of immune responses are warranted to better characterize vaccine effects. METHODS We conducted an ancillary study to analyze changes in the serum and cellular responses. Serum biomarkers of activation/inflammation were evaluated using a Luminex assay. Vaccine-elicited T-cell responses and functions were evaluated by assessing their phenotype, cytokine production, proliferation, and cytotoxic potential. Integrated data analysis was performed through correlation and principal component analysis of serum biomarkers and cellular immune responses. RESULTS Forty-eight volunteers were included. The Ad26.ZEBOV, MVA-BN-Filo vaccine elicited (1) serum increase of inflammatory/activation markers mainly at 1 day after the Ad26.ZEBOV vaccine; and (2) durable EBOV-specific T-cell proliferation and CD8+ T cells exhibiting a cytotoxic phenotype after Ad26.ZEBOV prime, after MVA-BN-Filo boost, and 6 months postvaccination. Integrated analysis revealed correlations between (1) EBOV-specific CD8+ T-cell proliferation and cytotoxic phenotype; and (2) high EBOV-specific CD8+ T-cell cytotoxic phenotype and low inflammatory marker IL-8 at day 1 postvaccination. DISCUSSION This study provides unique insights into the in vivo contribution of proliferation/cytotoxic CD8+ T cells and inflammation to the Ad26.ZEBOV, MVA-BN-Filo vaccine-induced potency. Clinical Trials Registration. NCT02416453.
Collapse
Affiliation(s)
- Christine Lacabaratz
- Vaccine Research Institute, Créteil, France
- INSERM Unité U955, Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
- Faculté de Médecine, Université Paris-Est, Créteil, France
| | - Mélany Durand
- Vaccine Research Institute, Créteil, France
- Population Health Research Center, Université Bordeaux, INSERM UMR 1219, Statistics in System Biology and Translational Medicine (SISTM), Institut national de recherche en sciences et technologies du numérique (INRIA), Bordeaux, France
| | - Aurélie Wiedemann
- Vaccine Research Institute, Créteil, France
- INSERM Unité U955, Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
- Faculté de Médecine, Université Paris-Est, Créteil, France
| | - Emile Foucat
- Vaccine Research Institute, Créteil, France
- INSERM Unité U955, Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
- Faculté de Médecine, Université Paris-Est, Créteil, France
| | - Mathieu Surénaud
- Vaccine Research Institute, Créteil, France
- INSERM Unité U955, Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
- Faculté de Médecine, Université Paris-Est, Créteil, France
| | - Corinne Krief
- Vaccine Research Institute, Créteil, France
- INSERM Unité U955, Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
- Faculté de Médecine, Université Paris-Est, Créteil, France
| | - Lydia Guillaumat
- Vaccine Research Institute, Créteil, France
- INSERM Unité U955, Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
- Faculté de Médecine, Université Paris-Est, Créteil, France
| | | | - Kerstin Luhn
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | - Viki Bockstal
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | - Rodolphe Thiébaut
- Vaccine Research Institute, Créteil, France
- Population Health Research Center, Université Bordeaux, INSERM UMR 1219, Statistics in System Biology and Translational Medicine (SISTM), Institut national de recherche en sciences et technologies du numérique (INRIA), Bordeaux, France
- Department of Medical Information and CIC-EC 1401/Euclid F-CRIN, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Laura Richert
- Vaccine Research Institute, Créteil, France
- Population Health Research Center, Université Bordeaux, INSERM UMR 1219, Statistics in System Biology and Translational Medicine (SISTM), Institut national de recherche en sciences et technologies du numérique (INRIA), Bordeaux, France
- Department of Medical Information and CIC-EC 1401/Euclid F-CRIN, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Yves Lévy
- Vaccine Research Institute, Créteil, France
- INSERM Unité U955, Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
- Faculté de Médecine, Université Paris-Est, Créteil, France
- Service d'Immunologie Clinique, Groupe Henri-Mondor Albert-Chenevier, Assistance Publique-Hôpitaux de Paris, Créteil, France
| |
Collapse
|
2
|
Watson CH, Gsell PS, Hall Y, Camacho A, Riveros X, Enwere G, Vicari A, Nadlaou SD, Toure A, Sani IM, Diallo A, Kolie C, Duraffour S, Ifono K, Maomou A, Dore K, Djidonou HA, Bagayoko A, Damey PP, Camara MN, Diallo FB, Oumar FT, Toure K, Diaby ML, Sylla L, Conde D, Kaba IL, Tipton T, Eggo RM, Marks M, Roberts CH, Strecker T, Günther S, Keita S, Edmunds WJ, Carroll MW, Henao-Restrepo AM. rVSV-ZEBOV vaccination in people with pre-existing immunity to Ebolavirus: an open-label safety and immunogenicity study in Guinean communities affected by Ebola virus disease (l'essai proches). BMC Med 2024; 22:523. [PMID: 39511527 PMCID: PMC11545826 DOI: 10.1186/s12916-024-03726-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Zaire Ebolavirus disease (EVD) outbreaks can be controlled using rVSV-ZEBOV vaccination and other public health measures. People in high-risk areas may have pre-existing antibodies from asymptomatic Ebolavirus exposure that might affect response to rVSV-ZEBOV. Therefore, we assessed the impact pre-existing immunity had on post-vaccination IgG titre, virus neutralisation, and reactogenicity following vaccination. METHODS In this prospective cohort study, 2115 consenting close contacts ("proches") of EVD survivors were recruited. Proches were vaccinated with rVSV-ZEBOV and followed up for 28 days for safety and immunogenicity. Anti-GP IgG titre at baseline and day 28 was assessed by ELISA. Samples from a representative subset were evaluated using live virus neutralisation. RESULTS Ten percent were seropositive at baseline. At day 28, IgG in baseline seronegative (GMT 0.106 IU/ml, 95% CI: 0.100 to 0.113) and seropositive (GMT 0.237 IU/ml, 0.210 to 0.267) participants significantly increased from baseline (both p < 0.0001). There was strong correlation between antibody titres and virus neutralisation in day 28 samples (Spearman's rho 0.75). Vaccinees with baseline IgG antibodies against Zaire Ebolavirus had similar safety profiles to those without detectable antibodies (63.6% vs 66.1% adults experienced any adverse event; 49.1% vs 60.9% in children), with almost all adverse events graded as mild. No serious adverse events were attributed to vaccination. No EVD survivors tested positive for Ebolavirus by RT-PCR. CONCLUSIONS These data add further evidence of rVSV-ZEBOV safety and immunogenicity, including in people with pre-existing antibodies from suspected natural ZEBOV infection whose state does not blunt rVSV-ZEBOV immune response. Pre-vaccination serological screening is not required.
Collapse
Affiliation(s)
| | | | - Yper Hall
- UK Health Security Agency, London, UK
| | | | | | | | | | | | | | | | | | - Cece Kolie
- World Health Organization, Geneva, Switzerland
| | | | - Kékoura Ifono
- Bernhard-Nocht-Institut Für Tropenmedizin, Hamburg, Germany
| | | | - Kassie Dore
- World Health Organization, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Michael Marks
- London School of Hygiene and Tropical Medicine, London, UK
| | | | | | | | | | - W John Edmunds
- London School of Hygiene and Tropical Medicine, London, UK
| | | | | |
Collapse
|
3
|
Zhang Z, Yang W, Chen Z, Chi H, Wu S, Zheng W, Jin R, Wang B, Wang Y, Huo N, Zhang J, Song X, Xu L, Zhang J, Hou L, Chen W. A causal multiomics study discriminates the early immune features of Ad5-vectored Ebola vaccine recipients. Innovation (N Y) 2024; 5:100603. [PMID: 38745762 PMCID: PMC11092886 DOI: 10.1016/j.xinn.2024.100603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 02/28/2024] [Indexed: 05/16/2024] Open
Abstract
The vaccine-induced innate immune response is essential for the generation of an antibody response. To date, how Ad5-vectored vaccines are influenced by preexisting anti-Ad5 antibodies during activation of the early immune response remains unclear. Here, we investigated the specific alterations in GP1,2-specific IgG-related elements of the early immune response at the genetic, molecular, and cellular levels on days 0, 1, 3, and 7 after Ad5-EBOV vaccination. In a causal multiomics analysis, distinct early immune responses associated with GP1,2-specific IgG were observed in Ad5-EBOV recipients with a low level of preexisting anti-Ad5 antibodies. This study revealed the correlates of the Ad5-EBOV-induced IgG response and provided mechanistic evidence for overcoming preexisting Ad5 immunity during the administration of Ad5-vectored vaccines.
Collapse
Affiliation(s)
- Zhe Zhang
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Wenjing Yang
- Department of Intelligent Data Science, College of Computer Science and Technology, National University of Defense Technology, Changsha 410073, China
| | - Zhengshan Chen
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Haoang Chi
- Department of Intelligent Data Science, College of Computer Science and Technology, National University of Defense Technology, Changsha 410073, China
- Intelligent Game and Decision Lab, Academy of Military Science, Beijing 100091, China
| | - Shipo Wu
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Wanru Zheng
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Ruochun Jin
- Department of Intelligent Data Science, College of Computer Science and Technology, National University of Defense Technology, Changsha 410073, China
| | - Busen Wang
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Yudong Wang
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Nan Huo
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Jinlong Zhang
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Xiaohong Song
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Liyang Xu
- Department of Intelligent Data Science, College of Computer Science and Technology, National University of Defense Technology, Changsha 410073, China
| | - Jun Zhang
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Lihua Hou
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| | - Wei Chen
- Institute of Biotechnology, Academy of Military Medical Sciences, Beijing 100071, China
| |
Collapse
|
4
|
Martinez-Murillo PA, Huttner A, Lemeille S, Medaglini D, Ottenhoff THM, Harandi AM, Didierlaurent AM, Siegrist CA. Refined innate plasma signature after rVSVΔG-ZEBOV-GP immunization is shared among adult cohorts in Europe and North America. Front Immunol 2024; 14:1279003. [PMID: 38235127 PMCID: PMC10791923 DOI: 10.3389/fimmu.2023.1279003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/07/2023] [Indexed: 01/19/2024] Open
Abstract
Background During the last decade Ebola virus has caused several outbreaks in Africa. The recombinant vesicular stomatitis virus-vectored Zaire Ebola (rVSVΔG-ZEBOV-GP) vaccine has proved safe and immunogenic but is reactogenic. We previously identified the first innate plasma signature response after vaccination in Geneva as composed of five monocyte-related biomarkers peaking at day 1 post-immunization that correlates with adverse events, biological outcomes (haematological changes and viremia) and antibody titers. In this follow-up study, we sought to identify additional biomarkers in the same Geneva cohort and validate those identified markers in a US cohort. Methods Additional biomarkers were identified using multiplexed protein biomarker platform O-link and confirmed by Luminex. Principal component analysis (PCA) evaluated if these markers could explain a higher variability of the vaccine response (and thereby refined the initial signature). Multivariable and linear regression models evaluated the correlations of the main components with adverse events, biological outcomes, and antibody titers. External validation of the refined signature was conducted in a second cohort of US vaccinees (n=142). Results Eleven additional biomarkers peaked at day 1 post-immunization: MCP2, MCP3, MCP4, CXCL10, OSM, CX3CL1, MCSF, CXCL11, TRAIL, RANKL and IL15. PCA analysis retained three principal components (PC) that accounted for 79% of the vaccine response variability. PC1 and PC2 were very robust and had different biomarkers that contributed to their variability. PC1 better discriminated different doses, better defined the risk of fever and myalgia, while PC2 better defined the risk of headache. We also found new biomarkers that correlated with reactogenicity, including transient arthritis (MCP-2, CXCL10, CXCL11, CX3CL1, MCSF, IL-15, OSM). Several innate biomarkers are associated with antibody levels one and six months after vaccination. Refined PC1 correlated strongly in both data sets (Geneva: r = 0.97, P < 0.001; US: r = 0.99, P< 0.001). Conclusion Eleven additional biomarkers refined the previously found 5-biomarker Geneva signature. The refined signature better discriminated between different doses, was strongly associated with the risk of adverse events and with antibody responses and was validated in a separate cohort.
Collapse
Affiliation(s)
- Paola Andrea Martinez-Murillo
- Center of Vaccinology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Angela Huttner
- Center for Vaccinology, Geneva University Hospitals, Geneva, Switzerland
- Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for Clinical Research, Geneva University Hospitals, Geneva, Switzerland
| | - Sylvain Lemeille
- Center of Vaccinology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Ali M. Harandi
- Department of Microbiology and Immunology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Vaccine Evaluation Centre, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Arnaud M. Didierlaurent
- Center of Vaccinology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Claire-Anne Siegrist
- Center of Vaccinology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Center for Vaccinology, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
5
|
Gonzalez Dias Carvalho PC, Dominguez Crespo Hirata T, Mano Alves LY, Moscardini IF, do Nascimento APB, Costa-Martins AG, Sorgi S, Harandi AM, Ferreira DM, Vianello E, Haks MC, Ottenhoff THM, Santoro F, Martinez-Murillo P, Huttner A, Siegrist CA, Medaglini D, Nakaya HI. Baseline gene signatures of reactogenicity to Ebola vaccination: a machine learning approach across multiple cohorts. Front Immunol 2023; 14:1259197. [PMID: 38022684 PMCID: PMC10663260 DOI: 10.3389/fimmu.2023.1259197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The rVSVDG-ZEBOV-GP (Ervebo®) vaccine is both immunogenic and protective against Ebola. However, the vaccine can cause a broad range of transient adverse reactions, from headache to arthritis. Identifying baseline reactogenicity signatures can advance personalized vaccinology and increase our understanding of the molecular factors associated with such adverse events. Methods In this study, we developed a machine learning approach to integrate prevaccination gene expression data with adverse events that occurred within 14 days post-vaccination. Results and Discussion We analyzed the expression of 144 genes across 343 blood samples collected from participants of 4 phase I clinical trial cohorts: Switzerland, USA, Gabon, and Kenya. Our machine learning approach revealed 22 key genes associated with adverse events such as local reactions, fatigue, headache, myalgia, fever, chills, arthralgia, nausea, and arthritis, providing insights into potential biological mechanisms linked to vaccine reactogenicity.
Collapse
Affiliation(s)
| | - Thiago Dominguez Crespo Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Leandro Yukio Mano Alves
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | - André G. Costa-Martins
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Artificial Intelligence and Analytics Department, Institute for Technological Research, São Paulo, Brazil
| | - Sara Sorgi
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Ali M. Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Vaccine Evaluation Center, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Daniela M. Ferreira
- Oxford Vaccine Group, University of Oxford, Oxford, United Kingdom
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Eleonora Vianello
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Mariëlle C. Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Francesco Santoro
- Laboratory of Molecular Microbiology and Biotechnology (LAMMB), Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Angela Huttner
- Centre for Vaccinology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Infectious Diseases Service, Geneva University Hospitals, Geneva, Switzerland
| | - Claire-Anne Siegrist
- Centre for Vaccinology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Donata Medaglini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Helder I. Nakaya
- Scientific Platform Pasteur-University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
6
|
Blengio F, Hocini H, Richert L, Lefebvre C, Durand M, Hejblum B, Tisserand P, McLean C, Luhn K, Thiebaut R, Levy Y. Identification of early gene expression profiles associated with long-lasting antibody responses to the Ebola vaccine Ad26.ZEBOV/MVA-BN-Filo. Cell Rep 2023; 42:113101. [PMID: 37691146 DOI: 10.1016/j.celrep.2023.113101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/24/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023] Open
Abstract
Ebola virus disease is a severe hemorrhagic fever with a high fatality rate. We investigate transcriptome profiles at 3 h, 1 day, and 7 days after vaccination with Ad26.ZEBOV and MVA-BN-Filo. 3 h after Ad26.ZEBOV injection, we observe an increase in genes related to antigen presentation, sensing, and T and B cell receptors. The highest response occurs 1 day after Ad26.ZEBOV injection, with an increase of the gene expression of interferon-induced antiviral molecules, monocyte activation, and sensing receptors. This response is regulated by the HESX1, ATF3, ANKRD22, and ETV7 transcription factors. A plasma cell signature is observed on day 7 post-Ad26.ZEBOV vaccination, with an increase of CD138, MZB1, CD38, CD79A, and immunoglobulin genes. We have identified early expressed genes correlated with the magnitude of the antibody response 21 days after the MVA-BN-Filo and 364 days after Ad26.ZEBOV vaccinations. Our results provide early gene signatures that correlate with vaccine-induced Ebola virus glycoprotein-specific antibodies.
Collapse
Affiliation(s)
- Fabiola Blengio
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Hakim Hocini
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Laura Richert
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; University Bordeaux, Department of Public Health, INSERM Bordeaux Population Health Research Centre, Inria SISTM, UMR 1219, Bordeaux, France; CHU de Bordeaux, Pôle de Santé Publique, Service d'Information Médicale, Bordeaux, France
| | - Cécile Lefebvre
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Mélany Durand
- University Bordeaux, Department of Public Health, INSERM Bordeaux Population Health Research Centre, Inria SISTM, UMR 1219, Bordeaux, France; CHU de Bordeaux, Pôle de Santé Publique, Service d'Information Médicale, Bordeaux, France
| | - Boris Hejblum
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; University Bordeaux, Department of Public Health, INSERM Bordeaux Population Health Research Centre, Inria SISTM, UMR 1219, Bordeaux, France; CHU de Bordeaux, Pôle de Santé Publique, Service d'Information Médicale, Bordeaux, France
| | - Pascaline Tisserand
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France
| | - Chelsea McLean
- Janssen Vaccines & Prevention, B.V. Archimediesweg, Leiden, the Netherlands
| | - Kerstin Luhn
- Janssen Vaccines & Prevention, B.V. Archimediesweg, Leiden, the Netherlands
| | - Rodolphe Thiebaut
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; University Bordeaux, Department of Public Health, INSERM Bordeaux Population Health Research Centre, Inria SISTM, UMR 1219, Bordeaux, France; CHU de Bordeaux, Pôle de Santé Publique, Service d'Information Médicale, Bordeaux, France.
| | - Yves Levy
- Vaccine Research Institute, Université Paris-Est Créteil, Faculté de Médecine, INSERM U955, Team 16, Créteil, France; Assistance Publique-Hôpitaux de Paris, Groupe Henri-Mondor Albert-Chenevier, Service Immunologie Clinique, Créteil, France.
| |
Collapse
|
7
|
Monath TP, Nichols R, Feldmann F, Griffin A, Haddock E, Callison J, Meade-White K, Okumura A, Lovaglio J, Hanley PW, Clancy CS, Shaia C, Rida W, Fusco J. Immunological correlates of protection afforded by PHV02 live, attenuated recombinant vesicular stomatitis virus vector vaccine against Nipah virus disease. Front Immunol 2023; 14:1216225. [PMID: 37731485 PMCID: PMC10507387 DOI: 10.3389/fimmu.2023.1216225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/16/2023] [Indexed: 09/22/2023] Open
Abstract
Introduction Immune correlates of protection afforded by PHV02, a recombinant vesicular stomatitis (rVSV) vector vaccine against Nipah virus (NiV) disease, were investigated in the African green monkey (AGM) model. Neutralizing antibody to NiV has been proposed as the principal mediator of protection against future NiV infection. Methods Two approaches were used to determine the correlation between neutralizing antibody levels and outcomes following a severe (1,000 median lethal doses) intranasal/intratracheal (IN/IT) challenge with NiV (Bangladesh): (1) reduction in vaccine dose given 28 days before challenge and (2) challenge during the early phase of the antibody response to the vaccine. Results Reduction in vaccine dose to very low levels led to primary vaccine failure rather than a sub-protective level of antibody. All AGMs vaccinated with the nominal clinical dose (2 × 107 pfu) at 21, 14, or 7 days before challenge survived. AGMs vaccinated at 21 days before challenge had neutralizing antibodies (geometric mean titer, 71.3). AGMs vaccinated at 7 or 14 days before challenge had either undetectable or low neutralizing antibody titers pre-challenge but had a rapid rise in titers after challenge that abrogated the NiV infection. A simple logistic regression model of the combined studies was used, in which the sole explanatory variable was pre-challenge neutralizing antibody titers. For a pre-challenge titer of 1:5, the predicted survival probability is 100%. The majority of animals with pre-challenge neutralizing titer of ≥1:20 were protected against pulmonary infiltrates on thoracic radiograms, and a majority of those with titers ≥1:40 were protected against clinical signs of illness and against a ≥fourfold antibody increase following challenge (indicating sterile immunity). Controls receiving rVSV-Ebola vaccine rapidly succumbed to NiV challenge, eliminating the innate immunity stimulated by the rVSV vector as a contributor to survival in monkeys challenged as early as 7 days after vaccination. Discussion and conclusion It was concluded that PHV02 vaccine elicited a rapid onset of protection and that any detectable level of neutralizing antibody was a functional immune correlate of survival.
Collapse
Affiliation(s)
- Thomas P. Monath
- Crozet Biopharma LLC, Lexington, MA, United States
- Public Health Vaccines Inc., Cambridge, MA, 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
| | - Amanda Griffin
- Laboratory of Virology, Division of Intramural Studies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Elaine Haddock
- Laboratory of Virology, Division of Intramural Studies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Julie Callison
- Laboratory of Virology, Division of Intramural Studies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Studies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Atsushi Okumura
- Laboratory of Virology, Division of Intramural Studies, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Patrick W. Hanley
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Chad S. Clancy
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States
| | - Wasima Rida
- Biostatistics Consultant, Arlington, VA, United States
| | - Joan Fusco
- Public Health Vaccines Inc., Cambridge, MA, United States
| |
Collapse
|
8
|
Amai M, Nojima M, Yuki Y, Kiyono H, Nagamura F. A review of criteria strictness in "Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials". Vaccine 2023; 41:5622-5629. [PMID: 37532612 DOI: 10.1016/j.vaccine.2023.07.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/25/2023] [Accepted: 07/29/2023] [Indexed: 08/04/2023]
Abstract
To assess safety in vaccine development, stricter grading scales, such as the "Toxicity Grading Scale for Healthy Adult and Adolescent Volunteers Enrolled in Preventive Vaccine Clinical Trials" issued by the U.S. Food and Drug Administration (FDA grading scale), are required. However, concern exists that their strictness may lead to an overestimation of some adverse events (AEs). We analyzed the details of AEs in a phase I clinical trial of a preventive vaccine for infectious diseases. In this trial, we observed the high occurrence of Grade 1 or greater AEs in hemoglobin changes from baseline value, and hypernatremia, and hypokalemia by FDA grading scale. The range considered as non-AE according to the FDA grading scale shifted or became narrower when compared to reference intervals, especially for a Japanese cohort. For sodium grading, the criterion for hypernatremia was around 2 to mEq/L lower than the upper limit of most standards in several countries. Also, the criterion for hypokalemia was around 0.2 mEq/L higher than the lower limit of most standards. Regarding a decrease in hemoglobin from baseline, the criterion of "any decrease" used for a Grade 1 AE was too strict and we suggest this be omitted. Upper and lower limits of AE criteria for sodium and potassium should be equal to, or 10-20% above, the reference interval consistent with other toxicities determined by laboratory tests. Consideration should be given to the issues surrounding the criteria that determine AEs before conducting clinical trials.
Collapse
Affiliation(s)
- Motoki Amai
- Center for Translational Research, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masanori Nojima
- Center for Translational Research, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Advanced Medicine Promotion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Yoshikazu Yuki
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; HanaVax Inc., Chiba, Japan
| | - Hiroshi Kiyono
- HanaVax Inc., Chiba, Japan; Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Future Medicine Education and Research Organization, Chiba University, Chiba, Japan; CU-UCSD Center for Mucosal Immunology, Allergy, and Vaccine (cMAV), Departments of Medicine and Pathology, University of California, San Diego, CA, USA
| | - Fumitaka Nagamura
- Center for Translational Research, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Advanced Medicine Promotion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
9
|
Zhang HQ, Zhang QY, Yuan ZM, Zhang B. The potential epidemic threat of Ebola virus and the development of a preventive vaccine. JOURNAL OF BIOSAFETY AND BIOSECURITY 2023; 5:67-78. [DOI: 10.1016/j.jobb.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
10
|
Wang S, Liang B, Wang W, Li L, Feng N, Zhao Y, Wang T, Yan F, Yang S, Xia X. Viral vectored vaccines: design, development, preventive and therapeutic applications in human diseases. Signal Transduct Target Ther 2023; 8:149. [PMID: 37029123 PMCID: PMC10081433 DOI: 10.1038/s41392-023-01408-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/06/2023] [Accepted: 03/14/2023] [Indexed: 04/09/2023] Open
Abstract
Human diseases, particularly infectious diseases and cancers, pose unprecedented challenges to public health security and the global economy. The development and distribution of novel prophylactic and therapeutic vaccines are the prioritized countermeasures of human disease. Among all vaccine platforms, viral vector vaccines offer distinguished advantages and represent prominent choices for pathogens that have hampered control efforts based on conventional vaccine approaches. Currently, viral vector vaccines remain one of the best strategies for induction of robust humoral and cellular immunity against human diseases. Numerous viruses of different families and origins, including vesicular stomatitis virus, rabies virus, parainfluenza virus, measles virus, Newcastle disease virus, influenza virus, adenovirus and poxvirus, are deemed to be prominent viral vectors that differ in structural characteristics, design strategy, antigen presentation capability, immunogenicity and protective efficacy. This review summarized the overall profile of the design strategies, progress in advance and steps taken to address barriers to the deployment of these viral vector vaccines, simultaneously highlighting their potential for mucosal delivery, therapeutic application in cancer as well as other key aspects concerning the rational application of these viral vector vaccines. Appropriate and accurate technological advances in viral vector vaccines would consolidate their position as a leading approach to accelerate breakthroughs in novel vaccines and facilitate a rapid response to public health emergencies.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Bo Liang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Weiqi Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ling Li
- China National Research Center for Exotic Animal Diseases, China Animal Health and Epidemiology Center, Qingdao, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China.
| |
Collapse
|
11
|
Crozier I, Britson KA, Wolfe DN, Klena JD, Hensley LE, Lee JS, Wolfraim LA, Taylor KL, Higgs ES, Montgomery JM, Martins KA. The Evolution of Medical Countermeasures for Ebola Virus Disease: Lessons Learned and Next Steps. Vaccines (Basel) 2022; 10:1213. [PMID: 36016101 PMCID: PMC9415766 DOI: 10.3390/vaccines10081213] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/26/2022] Open
Abstract
The Ebola virus disease outbreak that occurred in Western Africa from 2013-2016, and subsequent smaller but increasingly frequent outbreaks of Ebola virus disease in recent years, spurred an unprecedented effort to develop and deploy effective vaccines, therapeutics, and diagnostics. This effort led to the U.S. regulatory approval of a diagnostic test, two vaccines, and two therapeutics for Ebola virus disease indications. Moreover, the establishment of fieldable diagnostic tests improved the speed with which patients can be diagnosed and public health resources mobilized. The United States government has played and continues to play a key role in funding and coordinating these medical countermeasure efforts. Here, we describe the coordinated U.S. government response to develop medical countermeasures for Ebola virus disease and we identify lessons learned that may improve future efforts to develop and deploy effective countermeasures against other filoviruses, such as Sudan virus and Marburg virus.
Collapse
Affiliation(s)
- Ian Crozier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA;
| | - Kyla A. Britson
- U.S. Department of Health and Human Services (DHHS), Assistant Secretary for Preparedness and Response (ASPR), Biomedical Advanced Research and Development Authority (BARDA), Washington, DC 20201, USA; (K.A.B.); (D.N.W.); (J.S.L.)
- U.S. Department of Health and Human Services (DHHS), Assistant Secretary for Preparedness and Response (ASPR), Biomedical Advanced Research and Development Authority (BARDA), Oak Ridge Institute for Science and Education (ORISE) Postdoctoral Fellow, Oak Ridge, TN 37831, USA
| | - Daniel N. Wolfe
- U.S. Department of Health and Human Services (DHHS), Assistant Secretary for Preparedness and Response (ASPR), Biomedical Advanced Research and Development Authority (BARDA), Washington, DC 20201, USA; (K.A.B.); (D.N.W.); (J.S.L.)
| | - John D. Klena
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA; (J.D.K.); (J.M.M.)
| | - Lisa E. Hensley
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, Fort Detrick, MD 12116, USA;
| | - John S. Lee
- U.S. Department of Health and Human Services (DHHS), Assistant Secretary for Preparedness and Response (ASPR), Biomedical Advanced Research and Development Authority (BARDA), Washington, DC 20201, USA; (K.A.B.); (D.N.W.); (J.S.L.)
| | - Larry A. Wolfraim
- U.S. Department of Health and Human Services (DHHS), National Institutes of Health (NIH), National Institute of Allergy and Infectious Diseases (NIAID), Rockville, MD 20852, USA; (L.A.W.); (K.L.T.); (E.S.H.)
| | - Kimberly L. Taylor
- U.S. Department of Health and Human Services (DHHS), National Institutes of Health (NIH), National Institute of Allergy and Infectious Diseases (NIAID), Rockville, MD 20852, USA; (L.A.W.); (K.L.T.); (E.S.H.)
| | - Elizabeth S. Higgs
- U.S. Department of Health and Human Services (DHHS), National Institutes of Health (NIH), National Institute of Allergy and Infectious Diseases (NIAID), Rockville, MD 20852, USA; (L.A.W.); (K.L.T.); (E.S.H.)
| | - Joel M. Montgomery
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA; (J.D.K.); (J.M.M.)
| | - Karen A. Martins
- U.S. Department of Health and Human Services (DHHS), Assistant Secretary for Preparedness and Response (ASPR), Biomedical Advanced Research and Development Authority (BARDA), Washington, DC 20201, USA; (K.A.B.); (D.N.W.); (J.S.L.)
| |
Collapse
|
12
|
Vianello E, Gonzalez-Dias P, van Veen S, Engele CG, Quinten E, Monath TP, Medaglini D, Santoro F, Huttner A, Dubey S, Eichberg M, Ndungu FM, Kremsner PG, Essone PN, Agnandji ST, Siegrist CA, Nakaya HI, Ottenhoff THM, Haks MC. Transcriptomic signatures induced by the Ebola virus vaccine rVSVΔG-ZEBOV-GP in adult cohorts in Europe, Africa, and North America: a molecular biomarker study. THE LANCET. MICROBE 2022; 3:e113-e123. [PMID: 35544042 PMCID: PMC7613316 DOI: 10.1016/s2666-5247(21)00235-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/05/2021] [Accepted: 08/13/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND A recombinant vesicular stomatitis virus vector expressing the Zaire Ebola virus glycoprotein (rVSVΔG-ZEBOV-GP) vaccine has been reported as safe, immunogenic, and highly protective in a ring vaccination trial. We aimed to identify transcriptomic immune response biomarker signatures induced by vaccination and associated signatures with its immunogenicity and reactogenicity to better understand the potential mechanisms of action of the vaccine. METHODS 354 healthy adult volunteers were vaccinated in randomised, double-blind, placebo-controlled trials in Europe (Geneva, Switzerland [November, 2014, to January, 2015]) and North America (USA [Dec 5, 2014, to June 23, 2015]), and dose-escalation trials in Africa (Lambaréné, Gabon [November, 2014, to January, 2015], and Kilifi, Kenya [December, 2014, to January, 2015]) using different doses of the recombinant vesicular stomatitis virus vector expressing the Zaire Ebola virus glycoprotein (rVSVΔG-ZEBOV-GP; 3 × 105 to 1 × 108 plaque-forming units [pfu]). Longitudinal transcriptomic responses (days 0, 1, 2, 3, 7, 14, and 28) were measured in whole blood using a targeted gene expression profiling platform (dual-colour reverse-transcriptase multiplex ligation-dependent probe amplification) focusing on 144 immune-related genes. The effect of time and dose on transcriptomic response was also assessed. Logistic regression with lasso regularisation was applied to identify host signatures with optimal discriminatory capability of vaccination at day 1 or day 7 versus baseline, whereas random-effects models and recursive feature elimination combined with regularised logistic regression were used to associate signatures with immunogenicity and reactogenicity. FINDINGS Our results indicated that perturbation of gene expression peaked on day 1 and returned to baseline levels between day 7 and day 28. The magnitude of the response was dose-dependent, with vaccinees receiving a high dose (≥9 × 106 pfu) of rVSVΔG-ZEBOV-GP exhibiting the largest amplitude. The most differentially expressed genes that were significantly upregulated following vaccination consisted of type I and II interferon-related genes and myeloid cell-associated markers, whereas T cell, natural killer cell, and cytotoxicity-associated genes were downregulated. A gene signature associated with immunogenicity (common to all four cohorts) was identified correlating gene expression profiles with ZEBOV-GP antibody titres and a gene signatures associated with reactogenicity (Geneva cohort) was identified correlating gene expression profiles with an adverse event (ie, arthritis). INTERPRETATION Collectively, our results identify and cross-validate immune-related transcriptomic signatures induced by rVSVΔG-ZEBOV-GP vaccination in four cohorts of adult participants from different genetic and geographical backgrounds. These signatures will aid in the rational development, testing, and evaluation of novel vaccines and will allow evaluation of the effect of host factors such as age, co-infection, and comorbidity on responses to vaccines. FUNDING Innovative Medicines Initiative 2 Joint Undertaking.
Collapse
Affiliation(s)
- Eleonora Vianello
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.
| | - Patricia Gonzalez-Dias
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Suzanne van Veen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Carmen G Engele
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Edwin Quinten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Sclavo Vaccines Association, Siena, Italy
| | - Francesco Santoro
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Angela Huttner
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland; Center for Vaccinology, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Sheri Dubey
- Department of Vaccine and Biologics Research, Merck Research Laboratories, West Point, PA, USA
| | - Michael Eichberg
- Department of Vaccine and Biologics Research, Merck Research Laboratories, West Point, PA, USA
| | - Francis M Ndungu
- Department of Biosciences, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon; Institut für Tropenmedizin, Universitätsklinikum Tübingen, and German Center for Infection Research, Tübingen, Germany
| | - Paulin N Essone
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon; Institut für Tropenmedizin, Universitätsklinikum Tübingen, and German Center for Infection Research, Tübingen, Germany
| | - Claire-Anne Siegrist
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland; Center for Vaccinology, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Scientific Platform Pasteur-USP, São Paulo, Brazil
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
13
|
Bai Y, Wang Q, Liu M, Bian L, Liu J, Gao F, Mao Q, Wang Z, Wu X, Xu M, Liang Z. The next major emergent infectious disease: reflections on vaccine emergency development strategies. Expert Rev Vaccines 2022; 21:471-481. [PMID: 35080441 DOI: 10.1080/14760584.2022.2027240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Major emergent infectious diseases (MEID) pose the most serious threat to human health. The research proposes targeted response strategies for the prevention and control of potential MEID. AREAS COVERED Based on the analysis of infectious diseases, this research analyzes pandemics that have a high probability of occurrence and aims to synthesize the past experience and lessons learned of controlling infectious diseases such as coronavirus, influenza, Ebola, etc. In addition, by integrating major infectious disease response guidelines developed by WHO, the European Union, the United States, and the United Kingdom, we intend to bring forward national vaccine R&D development strategies for emergency use. EXPERT OPINION We advise to establish and improve existing laws, regulations, and also prevention and control systems for the emergent R&D and application of vaccines in response to potential infectious diseases. The strategies would not only help increase the various abilities in response to the research, development, evaluation, production, and supervision of emergency vaccines, but also establish surrogate endpoint of immunogenicity protection in early clinical studies to enable a rapid evaluation of the efficacy of emergency vaccines.
Collapse
Affiliation(s)
- Yu Bai
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Qian Wang
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Mingchen Liu
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Lianlian Bian
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Jianyang Liu
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Fan Gao
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Qunying Mao
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Zhongfang Wang
- Guangzhou Laboratory. No. 9 XingDaoHuanBei Road, Guangzhou, China
| | - Xing Wu
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Miao Xu
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| | - Zhenglun Liang
- Institute of Biological Products, Division of Hepatitis and Enterovirus Vaccines, National Institutes for Food and Drug Control, Beijing, China.,NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, Beijing, China.,NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing, China
| |
Collapse
|
14
|
Pinski AN, Messaoudi I. Therapeutic vaccination strategies against EBOV by rVSV-EBOV-GP: the role of innate immunity. Curr Opin Virol 2021; 51:179-189. [PMID: 34749265 DOI: 10.1016/j.coviro.2021.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/30/2022]
Abstract
Zaire Ebola virus (EBOV) is a member of the Filoviridae family. Infection with EBOV causes Ebola virus disease (EVD) characterized by excessive inflammation, lymphocyte death, coagulopathy, and multi-organ failure. In 2019, the FDA-approved the first anti-EBOV vaccine, rVSV-EBOV-GP (Ervebo® by Merck). This live-recombinant vaccine confers both prophylactic and therapeutic protection to nonhuman primates and humans. While mechanisms conferring prophylactic protection are well-investigated, those underlying protection conferred shortly before and after exposure to EBOV remain poorly understood. In this review, we review data from in vitro and in vivo studies analyzing early immune responses to rVSV-EBOV-GP and discuss the role of innate immune activation in therapeutic protection.
Collapse
Affiliation(s)
- Amanda N Pinski
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA; Center for Virus Research, University of California, Irvine, Irvine, CA, USA; Institute for Immunology, University of California, Irvine, Irvine, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
15
|
Risk factors for treatment failure in women with uncomplicated lower urinary tract infection. PLoS One 2021; 16:e0256464. [PMID: 34464397 PMCID: PMC8407559 DOI: 10.1371/journal.pone.0256464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/06/2021] [Indexed: 11/19/2022] Open
Abstract
Given rising antibiotic resistance and increasing use of delayed prescription for uncomplicated lower urinary tract infections (UTI), patients at risk for treatment failure should be identified early. We assessed risk factors for clinical and microbiological failure in women with lower UTI. This case-control study nested within a randomized clinical trial included all women in the per-protocol population (PPP), those in the PPP with microbiologically confirmed UTI, and those in the PPP with UTI due to Escherichia coli. Cases were women who experienced clinical and/or microbiologic failure; controls were those who did not. Risk factors for failure were assessed using multivariate logistic regression. In the PPP, there were 152 clinical cases for 307 controls. Among 340 women with microbiologically confirmed UTI, 126 and 102 cases with clinical and microbiological failure were considered with, respectively, 214 and 220 controls. Age ≥52 years was independently associated with clinical (adjusted OR 3.01; 95%CI 1.84-4.98) and microbiologic failure (aOR 2.55; 95%CI 1.54-4.25); treatment with fosfomycin was associated with clinical failure (aOR 2.35; 95%CI 1.47-3.80). The association with age persisted among all women, and women with E. coli-related UTI. Diabetes was not an independent risk factor, nor were other comorbidities. Postmenopausal age emerged as an independent risk factor for both clinical and microbiological treatment failure in women with lower UTI and should be considered to define women at-risk for non-spontaneous remission, and thus for delayed antibiotic therapy; diabetes mellitus was not associated with failure.
Collapse
|
16
|
Fehér C, Pastor-lbáñez R, Leal L, Plana M, Arnedo M, van den Ham HJ, Andeweg AC, Gruters RA, Díez-Fuertes F, Alcamí J, Aloy P, García F. Association of Transcriptomic Signatures of Inflammatory Response with Viral Control after Dendritic Cell-Based Therapeutic Vaccination in HIV-1 Infected Individuals. Vaccines (Basel) 2021; 9:vaccines9070799. [PMID: 34358215 PMCID: PMC8310264 DOI: 10.3390/vaccines9070799] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/05/2021] [Accepted: 07/14/2021] [Indexed: 12/01/2022] Open
Abstract
Systems vaccinology has seldomly been used in therapeutic HIV-1 vaccine research. Our aim was to identify early gene ‘signatures’ that predicted virus load control after analytical therapy interruption (ATI) in participants of a dendritic cell-based HIV-1 vaccine trial (DCV2). mRNA and miRNA were extracted from frozen post-vaccination PBMC samples; gene expression was determined by microarray method. In gene set enrichment analysis, responders showed an up-regulation of 14 gene sets (TNF-alpha/NFkB pathway, inflammatory response, the complement system, Il6 and Il2 JAK-STAT signaling, among others) and a down-regulation of 7 gene sets (such as E2F targets or interferon alpha response). The expression of genes regulated by three (miR-223-3p, miR-1183 and miR-8063) of the 9 differentially expressed miRNAs was significantly down-regulated in responders. The deregulation of certain gene sets related to inflammatory processes seems fundamental for viral control, and certain miRNAs may be important in fine-tuning these processes.
Collapse
Affiliation(s)
- Csaba Fehér
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, 08028 Barcelona, Spain;
- Infectious Diseases Department, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain; (L.L.); (F.D.-F.); (J.A.); (F.G.)
- Correspondence:
| | - Roque Pastor-lbáñez
- Retrovirology and Viral Immunopathology Laboratory, AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain; (R.P.-l.); (M.P.); (M.A.)
| | - Lorna Leal
- Infectious Diseases Department, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain; (L.L.); (F.D.-F.); (J.A.); (F.G.)
- Retrovirology and Viral Immunopathology Laboratory, AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain; (R.P.-l.); (M.P.); (M.A.)
| | - Montserrat Plana
- Retrovirology and Viral Immunopathology Laboratory, AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain; (R.P.-l.); (M.P.); (M.A.)
| | - Mireia Arnedo
- Retrovirology and Viral Immunopathology Laboratory, AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain; (R.P.-l.); (M.P.); (M.A.)
| | - Henk-Jan van den Ham
- Department of Viroscience, Erasmus MC, 3000CA Rotterdam, The Netherlands; (H.-J.v.d.H.); (A.C.A.); (R.A.G.)
| | - Arno C. Andeweg
- Department of Viroscience, Erasmus MC, 3000CA Rotterdam, The Netherlands; (H.-J.v.d.H.); (A.C.A.); (R.A.G.)
| | - Rob A. Gruters
- Department of Viroscience, Erasmus MC, 3000CA Rotterdam, The Netherlands; (H.-J.v.d.H.); (A.C.A.); (R.A.G.)
| | - Francisco Díez-Fuertes
- Infectious Diseases Department, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain; (L.L.); (F.D.-F.); (J.A.); (F.G.)
- National Center for Microbiology, Instituto de Salud Carlos III, 28222 Majadahonda, Spain
| | - José Alcamí
- Infectious Diseases Department, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain; (L.L.); (F.D.-F.); (J.A.); (F.G.)
- National Center for Microbiology, Instituto de Salud Carlos III, 28222 Majadahonda, Spain
| | - Patrick Aloy
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology, 08028 Barcelona, Spain;
- Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| | - Felipe García
- Infectious Diseases Department, Hospital Clinic, IDIBAPS, University of Barcelona, 08036 Barcelona, Spain; (L.L.); (F.D.-F.); (J.A.); (F.G.)
- Retrovirology and Viral Immunopathology Laboratory, AIDS Research Group, IDIBAPS, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain; (R.P.-l.); (M.P.); (M.A.)
| |
Collapse
|
17
|
Van Tilbeurgh M, Lemdani K, Beignon AS, Chapon C, Tchitchek N, Cheraitia L, Marcos Lopez E, Pascal Q, Le Grand R, Maisonnasse P, Manet C. Predictive Markers of Immunogenicity and Efficacy for Human Vaccines. Vaccines (Basel) 2021; 9:579. [PMID: 34205932 PMCID: PMC8226531 DOI: 10.3390/vaccines9060579] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
Vaccines represent one of the major advances of modern medicine. Despite the many successes of vaccination, continuous efforts to design new vaccines are needed to fight "old" pandemics, such as tuberculosis and malaria, as well as emerging pathogens, such as Zika virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Vaccination aims at reaching sterilizing immunity, however assessing vaccine efficacy is still challenging and underscores the need for a better understanding of immune protective responses. Identifying reliable predictive markers of immunogenicity can help to select and develop promising vaccine candidates during early preclinical studies and can lead to improved, personalized, vaccination strategies. A systems biology approach is increasingly being adopted to address these major challenges using multiple high-dimensional technologies combined with in silico models. Although the goal is to develop predictive models of vaccine efficacy in humans, applying this approach to animal models empowers basic and translational vaccine research. In this review, we provide an overview of vaccine immune signatures in preclinical models, as well as in target human populations. We also discuss high-throughput technologies used to probe vaccine-induced responses, along with data analysis and computational methodologies applied to the predictive modeling of vaccine efficacy.
Collapse
Affiliation(s)
- Matthieu Van Tilbeurgh
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Katia Lemdani
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Anne-Sophie Beignon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Catherine Chapon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Nicolas Tchitchek
- Unité de Recherche i3, Inserm UMR-S 959, Bâtiment CERVI, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France;
| | - Lina Cheraitia
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Ernesto Marcos Lopez
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Quentin Pascal
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Pauline Maisonnasse
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Caroline Manet
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| |
Collapse
|
18
|
Jonsson-Schmunk K, Ghose R, Croyle MA. Immunization and Drug Metabolizing Enzymes: Focus on Hepatic Cytochrome P450 3A. Expert Rev Vaccines 2021; 20:623-634. [PMID: 33666138 DOI: 10.1080/14760584.2021.1899818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Infectious disease emergencies like the 2013-2016 Ebola epidemic and the 2009 influenza and current SARS-CoV-2 pandemics illustrate that vaccines are now given to diverse populations with preexisting pathologies requiring pharmacological management. Many natural biomolecules (steroid hormones, fatty acids, vitamins) and ~60% of prescribed medications are processed by hepatic cytochrome P450 (CYP) 3A4. The objective of this work was to determine the impact of infection and vaccines on drug metabolism. METHODS The impact of an adenovirus-based vaccine expressing Ebola glycoprotein (AdEBO) and H1N1 and H3N2 influenza viruses on hepatic CYP 3A4 and associated nuclear receptors was evaluated in human hepatocytes (HC-04 cells) and in mice. RESULTS CYP3A activity was suppressed by 55% in mice 24 h after administration of mouse-adapted H1N1, while ˂10% activity remained in HC-04 cells after infection with H1N1 and H3N2 due to global suppression of cellular translation capacity, indicated by reduction (70%, H1N1, 56%, H3N2) of phosphorylated eukaryotic translation initiation factor 4e (eIF4E). AdEBO suppressed CYP3A activity in vivo (44%) and in vitro (26%) 24 hours after infection. CONCLUSION As the clinical evaluation of vaccines for SARS-CoV-2 and other global pathogens rise, studies to evaluate the impact of new vaccines and emerging pathogens on CYP3A4 and other metabolic enzymes are warranted to avoid therapeutic failures that could further compromise the public health during infectious disease emergencies.
Collapse
Affiliation(s)
- Kristina Jonsson-Schmunk
- Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas, Austin, Texas, USA
| | - Romi Ghose
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Maria A Croyle
- Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas, Austin, Texas, USA.,LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
19
|
Human Transcriptomic Response to the VSV-Vectored Ebola Vaccine. Vaccines (Basel) 2021; 9:vaccines9020067. [PMID: 33498214 PMCID: PMC7908976 DOI: 10.3390/vaccines9020067] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Ebolavirus Disease (EVD) is a severe haemorrhagic fever that occurs in epidemic outbreaks, with a high fatality rate and no specific therapies available. rVSVΔG-ZEBOV-GP (Ervebo®), a live-attenuated recombinant vesicular stomatitis virus vector expressing the glycoprotein G of Zaire Ebolavirus, is the first vaccine approved for prevention of EVD. Both innate and adaptive responses are deemed to be involved in vaccine-induced protection, yet the mechanisms are not fully elucidated. A global transcriptomic approach was used to profile the blood host-response in 51 healthy volunteers enrolled in a phase 1/2 clinical trial. Signatures of the host responses were investigated assessing the enrichment in differentially expressed genes (DEGs) of specific “blood transcription modules” (BTM). Comparison of gene-expression levels showed that vaccination produces a peak of 5469 DEGs at day one, representing 38.6% of the expressed genes. Out of 346 BTMs, 144 were significantly affected by vaccination. Innate immunity pathways were induced from day 1 to day 14. At days 2 and 3, neutrophil modules were downregulated and complement-related modules upregulated. T-cell and cell-cycle associated modules were upregulated at days 7 and 14, while at day 28, no modules remained activated. At day 14, a direct correlation was observed between ZEBOV glycoprotein-specific antibody titres and activation of seven BTMs, including two related to B-cell activation and B cell receptor signalling. Transcriptomic analysis identified an rVSVΔG-ZEBOV-GP-induced signature and demonstrated a direct correlation of blood transcriptomic changes with ZEBOV glycoprotein-specific antibody titres.
Collapse
|
20
|
Development of sustainable research excellence with a global perspective on infectious diseases: Centre de Recherches Médicales de Lambaréné (CERMEL), Gabon. Wien Klin Wochenschr 2021; 133:500-508. [PMID: 33398458 PMCID: PMC7781170 DOI: 10.1007/s00508-020-01794-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/30/2020] [Indexed: 11/17/2022]
Abstract
Medical research in sub-Saharan Africa is of high priority for societies to respond adequately to local health needs. Often enough it remains a challenge to build up capacity in infrastructure and human resources to highest international standards and to sustain this over mid-term to long-term periods due to difficulties in obtaining long-term institutional core funding, attracting highly qualified scientists for medical research and coping with ever changing structural and political environments. The Centre de Recherches Médicales de Lambaréné (CERMEL) serves as model for how to overcome such challenges and to continuously increase its impact on medical care in Central Africa and beyond. Starting off as a research annex to the Albert Schweitzer Hospital in Lambaréné, Gabon, it has since then expanded its activities to academic and regulatory clinical trials for drugs, vaccines and diagnostics in the field of malaria, tuberculosis, and a wide range of poverty related and neglected tropical infectious diseases. Advancing bioethics in medical research in Africa and steadily improving its global networks and infrastructures, CERMEL serves as a reference centre for several international consortia. In close collaboration with national authorities, CERMEL has become one of the main training hubs for medical research in Central Africa. It is hoped that CERMEL and its leitmotiv “to improve medical care for local populations” will serve as an inspiration to other institutions in sub-Saharan Africa to further increase African capacity to advance medicine.
Collapse
|
21
|
Tell JG, Coller BAG, Dubey SA, Jenal U, Lapps W, Wang L, Wolf J. Environmental Risk Assessment for rVSVΔG-ZEBOV-GP, a Genetically Modified Live Vaccine for Ebola Virus Disease. Vaccines (Basel) 2020; 8:vaccines8040779. [PMID: 33352786 PMCID: PMC7767225 DOI: 10.3390/vaccines8040779] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 01/04/2023] Open
Abstract
rVSVΔG-ZEBOV-GP is a live, attenuated, recombinant vesicular stomatitis virus (rVSV)-based vaccine for the prevention of Ebola virus disease caused by Zaire ebolavirus. As a replication-competent genetically modified organism, rVSVΔG-ZEBOV-GP underwent various environmental evaluations prior to approval, the most in-depth being the environmental risk assessment (ERA) required by the European Medicines Agency. This ERA, as well as the underlying methodology used to arrive at a sound conclusion about the environmental risks of rVSVΔG-ZEBOV-GP, are described in this review. Clinical data from vaccinated adults demonstrated only infrequent, low-level shedding and transient, low-level viremia, indicating a low person-to-person infection risk. Animal data suggest that it is highly unlikely that vaccinated individuals would infect animals with recombinant virus vaccine or that rVSVΔG-ZEBOV-GP would spread within animal populations. Preclinical studies in various hematophagous insect vectors showed that these species were unable to transmit rVSVΔG-ZEBOV-GP. Pathogenicity risk in humans and animals was found to be low, based on clinical and preclinical data. The overall risk for non-vaccinated individuals and the environment is thus negligible and can be minimized further through defined mitigation strategies. This ERA and the experience gained are relevant to developing other rVSV-based vaccines, including candidates under investigation for prevention of COVID-19.
Collapse
Affiliation(s)
- Joan G. Tell
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
- Correspondence:
| | - Beth-Ann G. Coller
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
| | - Sheri A. Dubey
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
| | - Ursula Jenal
- Jenal & Partners Biosafety Consulting, 4310 Rheinfelden, Switzerland;
| | - William Lapps
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
| | - Liman Wang
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
| | - Jayanthi Wolf
- Merck & Co., Inc., Kenilworth, NJ 07033, USA; (B.-A.G.C.); (S.A.D.); (W.L.); (L.W.); (J.W.)
| |
Collapse
|
22
|
Li J, Li S, Yang L, Cao P, Lu J. Severe fever with thrombocytopenia syndrome virus: a highly lethal bunyavirus. Crit Rev Microbiol 2020; 47:112-125. [PMID: 33245676 DOI: 10.1080/1040841x.2020.1847037] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Severe fever with thrombocytopenia syndrome virus (SFTSV) is a novel bunyavirus. Since 2007, SFTS disease has been reported in China with high fatality rate up to 30%, which drew high attention from Centre for Disease Control and Prevention and government. SFTSV is endemic in the centra l and eastern China, Korea and Japan. There also have been similar cases reported in Vietnam. The number of SFTSV infection cases has a steady growth in these years. As SFTSV could transmitted from person to person, it will expose the public to infectious risk. In 2018 annual review of the Blueprint list of priority diseases, World Health Organisation has listed SFTSV infection as prioritised diseases for research and development in emergency contexts. However, the pathogenesis of SFTSV remains largely unclear. Currently, there are no specific therapeutics or vaccines to combat infections of SFTSV. This review discusses recent findings of epidemiology, transmission pathway, pathogenesis and treatments of SFTS disease.
Collapse
Affiliation(s)
- Jing Li
- NHC Key Laboratory of Carcinogenesis, Department of Hematology, Xiangya Hospital, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Shen Li
- NHC Key Laboratory of Carcinogenesis, Department of Hematology, Xiangya Hospital, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Li Yang
- NHC Key Laboratory of Carcinogenesis, Department of Hematology, Xiangya Hospital, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Pengfei Cao
- NHC Key Laboratory of Carcinogenesis, Department of Hematology, Xiangya Hospital, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Jianhong Lu
- NHC Key Laboratory of Carcinogenesis, Department of Hematology, Xiangya Hospital, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Sciences, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| |
Collapse
|
23
|
Grazioli S, Tavaglione F, Torriani G, Wagner N, Rohr M, L’Huillier AG, Leclercq C, Perrin A, Bordessoule A, Beghetti M, Pachlopnik J, Vavassori S, Perreau M, Eberhardt C, Didierlaurent A, Kaiser L, Eckerle I, Roux-Lombard P, Blanchard-Rohner G. Immunological Assessment of Pediatric Multisystem Inflammatory Syndrome Related to Coronavirus Disease 2019. J Pediatric Infect Dis Soc 2020; 10:706-713. [PMID: 33180935 PMCID: PMC7717282 DOI: 10.1093/jpids/piaa142] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 11/10/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND Recently, cases of multisystem inflammatory syndrome in children (MIS-C) associated with coronavirus disease 2019 (COVID-19) have been reported worldwide. Negative polymerase chain reaction (RT-PCR) testing associated with positive serology in most of the cases suggests a postinfectious syndrome. Because the pathophysiology of this syndrome is still poorly understood, extensive virological and immunological investigations are needed. METHODS We report a series of 4 pediatric patients admitted to Geneva University Hospitals with persistent fever and laboratory evidence of inflammation meeting the published definition of MIS-C related to COVID-19, to whom an extensive virological and immunological workup was performed. RESULTS RT-PCRs on multiple anatomical compartments were negative, whereas anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunoglobulin A (IgA) and immunoglobulin G (IgG) were strongly positive by enzyme-linked immunosorbent assay and immunofluorescence. Both pseudoneutralization and full virus neutralization assays showed the presence of neutralizing antibodies in all children, confirming a recent infection with SARS-CoV-2. The analyses of cytokine profiles revealed an elevation in all cytokines, as reported in adults with severe COVID-19. Although differing in clinical presentation, some features of MIS-C show phenotypic overlap with hemophagocytic lymphohistiocytosis (HLH). In contrast to patients with primary HLH, our patients showed normal perforin expression and natural killer (NK) cell degranulation. The levels of soluble interleukin (IL)-2 receptor (sIL-2R) correlated with the severity of disease, reflecting recent T-cell activation. CONCLUSION Our findings suggest that MIS-C related to COVID-19 is caused by a postinfectious inflammatory syndrome associated with an elevation in all cytokines, and markers of recent T-cell activation (sIL-2R) occurring despite a strong and specific humoral response to SARS-CoV-2. Further functional and genetic analyses are essential to better understand the mechanisms of host-pathogen interactions.
Collapse
Affiliation(s)
- Serge Grazioli
- Division of Neonatal and Pediatric Intensive Care, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Fedora Tavaglione
- Division of Neonatal and Pediatric Intensive Care, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Giulia Torriani
- Geneva Centre for Emerging Viral Diseases, Geneva Switzerland
| | - Noemie Wagner
- Pediatric infectious diseases unit, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Marie Rohr
- Pediatric infectious diseases unit, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Arnaud G L’Huillier
- Geneva Centre for Emerging Viral Diseases, Geneva Switzerland,Pediatric infectious diseases unit, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Charlotte Leclercq
- Children’s Hospital of Geneva, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Anne Perrin
- Children’s Hospital of Geneva, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Alice Bordessoule
- Division of Neonatal and Pediatric Intensive Care, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Maurice Beghetti
- Pediatric Cardiology Unit, Children’s Hospital of Geneva, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | | | | | - Matthieu Perreau
- Division of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Switzerland
| | | | | | - Laurent Kaiser
- Geneva Centre for Emerging Viral Diseases, Geneva Switzerland
| | | | - Pascale Roux-Lombard
- Laboratory of Immunology and Allergology, Geneva University Hospitals and Geneva University, Geneva, Switzerland
| | - Geraldine Blanchard-Rohner
- Children’s Hospital of Geneva, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland,Center of Vaccinology, Geneva University Hospitals, Switzerland,Corresponding Author: Geraldine Blanchard Rohner, Children’s Hospital of Geneva, 6, rue Willy-Donzé, 1211 Genève 14, Switzerland,
| |
Collapse
|
24
|
To B or Not to B: Mechanisms of Protection Conferred by rVSV-EBOV-GP and the Roles of Innate and Adaptive Immunity. Microorganisms 2020; 8:microorganisms8101473. [PMID: 32992829 PMCID: PMC7600878 DOI: 10.3390/microorganisms8101473] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 12/28/2022] Open
Abstract
Zaire Ebola virus (EBOV) is a member of the Filoviridae family of negative sense, single-stranded RNA viruses. EBOV infection causes Ebola virus disease (EVD), characterized by coagulopathy, lymphopenia, and multi-organ failure, which can culminate in death. In 2019, the FDA approved the first vaccine against EBOV, a recombinant live-attenuated viral vector wherein the G protein of vesicular stomatitis virus is replaced with the glycoprotein (GP) of EBOV (rVSV-EBOV-GP, Ervebo® by Merck). This vaccine demonstrates high efficacy in nonhuman primates by providing prophylactic, rapid, and post-exposure protection. In humans, rVSV-EBOV-GP demonstrated 100% protection in several phase III clinical trials in over 10,000 individuals during the 2013–2016 West Africa epidemic. As of 2020, over 218,000 doses of rVSV-EBOV-GP have been administered to individuals with high risk of EBOV exposure. Despite licensure and robust preclinical studies, the mechanisms of rVSV-EBOV-GP-mediated protection are not fully understood. Such knowledge is crucial for understanding vaccine-mediated correlates of protection from EVD and to aid the further design and development of therapeutics against filoviruses. Here, we summarize the current literature regarding the host response to vaccination and EBOV exposure, and evidence regarding innate and adaptive immune mechanisms involved in rVSV-EBOV-GP-mediated protection, with a focus on the host transcriptional response. Current data strongly suggest a protective synergy between rapid innate and humoral immunity.
Collapse
|
25
|
Abstract
Since its discovery in 1976, Ebola virus (EBOV) has caused numerous outbreaks of fatal hemorrhagic disease in Africa. The biggest outbreak on record is the 2013-2016 epidemic in west Africa with almost 30,000 cases and over 11,000 fatalities, devastatingly affecting Guinea, Liberia, and Sierra Leone. The epidemic highlighted the need for licensed drugs or vaccines to quickly combat the disease. While at the beginning of the epidemic no licensed countermeasures were available, several experimental drugs with preclinical efficacy were accelerated into human clinical trials and used to treat patients with Ebola virus disease (EVD) toward the end of the epidemic. In the same manner, vaccines with preclinical efficacy were administered primarily to known contacts of EVD patients on clinical trial protocols using a ring-vaccination strategy. In this review, we describe the pathogenesis of EBOV and summarize the current status of EBOV vaccine development and treatment of EVD.
Collapse
Affiliation(s)
- Wakako Furuyama
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA;
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana 59840, USA;
| |
Collapse
|
26
|
Boum Y, Juan-Giner A, Hitchings M, Soumah A, Strecker T, Sadjo M, Cuthbertson H, Hayes P, Tchaton M, Jemmy JP, Clarck C, King D, Faga EM, Becker S, Halis B, Gunnstein N, Carroll M, Røttingen JA, Kondé MK, Doumbia M, Henao-Restrepo AM, Kieny MP, Cisse M, Draguez B, Grais RF. Humoral and cellular immune response induced by rVSVΔG-ZEBOV-GP vaccine among frontline workers during the 2013-2016 West Africa Ebola outbreak in Guinea. Vaccine 2020; 38:4877-4884. [PMID: 32499066 PMCID: PMC7306162 DOI: 10.1016/j.vaccine.2020.04.066] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/26/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND As part of a Phase III trial with the Ebola vaccine rVSVΔG-ZEBOV-GP in Guinea, we invited frontline workers (FLWs) to participate in a sub-study to provide additional information on the immunogenicity of the vaccine. METHODS We conducted an open-label, non-randomized, single-arm immunogenicity evaluation of one dose of rVSVΔG-ZEBOV-GP among healthy FLWs in Guinea. FLWs who refused vaccination were offered to participate as a control group. We followed participants for 84 days with a subset followed-up for 180 days. The primary endpoint was immune response, as measured by ELISA for ZEBOV-glycoprotein-specific antibodies (ELISA-GP) at 28 days. We also conducted neutralization, whole virion ELISA and enzyme-linked immunospot (ELISPOT) assay for cellular response. RESULTS A total of 1172 participants received one dose of vaccine and were followed-up for 84 days, among them 114 participants were followed-up for 180 days. Additionally, 99 participants were included in the control group and followed up for 180 days. Overall, 86.4% (95% CI 84.1-88.4) of vaccinated participants seroresponded at 28 days post-vaccination (ELISA- GP) with 65% of these seroresponding at 14 days post-vaccination. Among those who seroresponded at 28 days, 90.7% (95% CI 82.0-95.4) were still seropositive at 180 days. The proportion of seropositivity in the unvaccinated group was 0.0% (95% CI 0.0-3.8) at 28 days and 5.4% (95% CI 2.1-13.1) at 180 days post-vaccination. We found weak correlation between ELISA-GP and neutralization at baseline but significant pairwise correlation at 28 days post-vaccination. Among samples analysed for cellular response, only 1 (2.2%) exhibited responses towards the Zaire Ebola glycoprotein (Ebola GP ≥ 10) at baseline, 10 (13.5%) at day 28 post-vaccination and 27 (48.2%) at Day 180. CONCLUSIONS We found one dose of rVSVΔG-ZEBOV-GP to be highly immunogenic at 28- and 180-days post vaccination among frontline workers in Guinea. We also found a cellular response that increased with time.
Collapse
Affiliation(s)
| | | | - Matt Hitchings
- Center for Communicable Disease Dynamics and Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | - Thomas Strecker
- Institute of Virology, Philipps University, Marburg, Germany
| | - Mariama Sadjo
- Centre Hospital-Universitaire de Donka, Conakry, Guinea
| | | | - Peter Hayes
- Division of Medicine, Department of Infectious Diseases, Imperial College London, UK
| | | | - Jean-Paul Jemmy
- Médecins Sans Frontières-Operational Center Belgium, Brussels, Belgium
| | - Carolyn Clarck
- Division of Infectious Disease Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Deborah King
- Division of Medicine, Department of Infectious Diseases, Imperial College London, UK
| | | | - Stephan Becker
- Institute of Virology, Philipps University, Marburg, Germany
| | - Bassam Halis
- Public Health England, National Infection Service, Porton Down, UK
| | - Norheim Gunnstein
- Division of Infectious Disease Control, Norwegian Institute of Public Health, Oslo, Norway
| | - Miles Carroll
- Public Health England, National Infection Service, Porton Down, UK
| | - John-Arne Røttingen
- Division of Infectious Disease Control, Norwegian Institute of Public Health, Oslo, Norway; Department of Health and Society, University of Oslo, Norway; Department of Global Health and Population, Harvard TH Chan School of Public Health, Boston, MA, USA; Coalition for Epidemic Preparedness Innovations, Care of Norwegian Institute of Public Health, Oslo, Norway; Research Council of Norway, Oslo, Norway
| | - Mandy Kader Kondé
- Center of Excellence for Training, Research On Malaria & Priority Diseases In Guinea, Conakry, Guinea
| | | | | | | | - Mohamed Cisse
- Centre Hospital-Universitaire de Donka, Conakry, Guinea
| | - Bertrand Draguez
- Médecins Sans Frontières-Operational Center Belgium, Brussels, Belgium
| | | |
Collapse
|
27
|
Pejoski D, de Rham C, Martinez-Murillo P, Santoro F, Auderset F, Medaglini D, Pozzi G, Vono M, Lambert PH, Huttner A, Haks MC, Ottenhoff THM, Villard J, Siegrist CA. Rapid dose-dependent Natural Killer (NK) cell modulation and cytokine responses following human rVSV-ZEBOV Ebolavirus vaccination. NPJ Vaccines 2020; 5:32. [PMID: 32337075 PMCID: PMC7156503 DOI: 10.1038/s41541-020-0179-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/06/2020] [Indexed: 12/31/2022] Open
Abstract
The rVSV-ZEBOV Ebolavirus vaccine confers protection within days after immunization, suggesting the contribution of innate immune responses. We report modulation of rVSV-ZEBOV vaccinee blood CD56+ NK cell numbers, NKG2D or NKp30 surface receptor expression, Killer Immunoglobulin-like Receptor (KIR)+ cell percentages and NK-cell-related genes on day 1 post immunization. Inverse correlations existed between the concentration of several plasma cytokines and inhibitory KIR+ CD56dim or cytokine-responsive CD56bright NK cells. Thus, NK cells may contribute to the early protective efficacy of rVSV-ZEBOV in humans.
Collapse
Affiliation(s)
- David Pejoski
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- World Health Organization Collaborating Centre for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Casimir de Rham
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Immunology and Transplantation Unit, Geneva University Hospitals, Geneva, Switzerland
| | - Paola Martinez-Murillo
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- World Health Organization Collaborating Centre for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Francesco Santoro
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Floriane Auderset
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- World Health Organization Collaborating Centre for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Donata Medaglini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gianni Pozzi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Maria Vono
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- World Health Organization Collaborating Centre for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Paul-Henri Lambert
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- World Health Organization Collaborating Centre for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Angela Huttner
- World Health Organization Collaborating Centre for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Infectious Diseases Service, Geneva University Hospitals, Geneva, Switzerland
| | - Mariëlle C. Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Jean Villard
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Immunology and Transplantation Unit, Geneva University Hospitals, Geneva, Switzerland
| | - Claire-Anne Siegrist
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- World Health Organization Collaborating Centre for Vaccine Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
28
|
Bache BE, Grobusch MP, Agnandji ST. Safety, immunogenicity and risk-benefit analysis of rVSV-ΔG-ZEBOV-GP (V920) Ebola vaccine in Phase I-III clinical trials across regions. Future Microbiol 2020; 15:85-106. [PMID: 32030996 DOI: 10.2217/fmb-2019-0237] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
To evaluate the risk-benefits balance of the rVSV-ΔG-ZEBOV-GP vaccine. We performed a systematic review to summarize data on safety, immunogenicity and efficacy. About 17,600 adults and 234 children received 11 different doses of the V920 vaccine ranging from 3000 to 100 million and 20 million plaque-forming units, respectively, during Phase I-III clinical trials. Cases of severe but transient arthritis were reported in about six and 0.08% of vaccinees in high-income countries (HICs) and low-middle-income countries (LMICs), respectively. The 20 million plaque-forming units dose yielded GP-specific antibody titres which peaked at day 28 with a pooled geometric mean titres of 2557.7 (95% CI: 1665.5-3934.2) versus 1156.9 (95% CI: 832.5-1649.2) but with similar seroconversion rates at 96% (95% CI: 87-100) versus 100% (95% CI: 90-100) for HICs and LMICs, respectively. Data from stringent Phase I-II clinical trials in LMICs and HICs and from the ring efficacy trials yielded a good risk-benefit balance of the V920 vaccine in adults, but also in children and pregnant and lactating women and HIV-infected people.
Collapse
Affiliation(s)
- Bache Emmanuel Bache
- Centre de Recherches Médicales de Lambaréné (CERMEL), Biomedicine and Social sciences, BP 242, Lambaréné, Gabon.,Center of Tropical Medicine & Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam University Medical Centres, location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Martin P Grobusch
- Centre de Recherches Médicales de Lambaréné (CERMEL), Biomedicine and Social sciences, BP 242, Lambaréné, Gabon.,Center of Tropical Medicine & Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam University Medical Centres, location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné (CERMEL), Biomedicine and Social sciences, BP 242, Lambaréné, Gabon.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| |
Collapse
|
29
|
Fouriki A, Fougère Y, De Camaret C, Blanchard Rohner G, Grazioli S, Wagner N, Relly C, Pachlopnik Schmid J, Trück J, Kottanatu L, Perez E, Perez MH, Schaffner D, Asner SA, Hofer M. Case Report: Case Series of Children With Multisystem Inflammatory Syndrome Following SARS-CoV-2 Infection in Switzerland. Front Pediatr 2020; 8:594127. [PMID: 33469522 PMCID: PMC7813982 DOI: 10.3389/fped.2020.594127] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022] Open
Abstract
Since the beginning of the severe SARS-CoV-2 pandemic, an increasing number of countries reported cases of a systemic hyperinflammatory condition defined as multi-system inflammatory syndrome in children (MIS-C). The clinical features of MIS-C can be an overlap of Kawasaki Disease (KD), Toxic Shock Syndrome (TSS), Macrophage Activation Syndrome (MAS), or have often an acute abdominal presentation. Intravenous immunoglobulin (IVIG) is recommended as first line therapy in KD. Recent evidence suggests intravenous immunoglobulins (IVIG) resistance in some cases of SARS-CoV-2 related MIS-C, thereby questioning the benefit of immunomodulators such as IL-1 or IL-6 blocking agents. We report on a cohort of 6 Swiss children with SARS-CoV2 related MIS-C presenting with clinical features compatible with Incomplete KD and Toxic Shock Syndrome associated to a cytokine storm. Serum cytokine profile investigations showed increased IL1RA levels (8 to 22-fold) in 5 of the 6 patients (one patient had not been tested), whereas, IL-6 serum levels were increased only in the 3 patients of the 6 who were tested. With exception of one patient who had only benefited by Anakinra, all patients received at least one dose of IVIG. One patient has only received Anakinra with favorable evolution, and three patients had also a steroid treatment. In addition to all this anti-inflammatory medication two patients have also received one dose of anti-IL6. In conclusion, our case series reports on clinical and laboratory findings of most of Swiss cases with MIS-C and suggests the use of Anakinra as an alternative to steroids in these children, most of whom presented with high IL-1RA levels.
Collapse
Affiliation(s)
- Athina Fouriki
- Pediatric Immuno-Rheumatology of Western Switzerland, Department Women-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland.,Pediatric Immuno-Rheumatology, Department of Paediatrics, University Hospital, Geneva, Switzerland
| | - Yves Fougère
- Pediatric Infectious Diseases and Vaccinology Unit, Department Women-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland
| | - Caroline De Camaret
- Pediatric Immuno-Rheumatology of Western Switzerland, Department Women-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland.,Pediatric Immuno-Rheumatology, Department of Paediatrics, University Hospital, Geneva, Switzerland
| | - Géraldine Blanchard Rohner
- Pediatric Immunology and Vaccinology Unit, Faculty of Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Serge Grazioli
- Division of Neonatal and Pediatric Intensive Care, Department of Paediatrics, Gynecology and Obstetrics, Geneva University Hospital, Geneva, Switzerland
| | - Noémie Wagner
- Pediatric Infectious Diseases Unit, Department of Paediatrics, Gynecology and Obstetrics, Geneva University Hospital, Geneva, Switzerland
| | - Christa Relly
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland
| | | | - Johannes Trück
- Division of Infectious Diseases and Hospital Epidemiology, University Children's Hospital Zurich, Zurich, Switzerland.,Pediatric Immunology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Lisa Kottanatu
- Pediatric Infectious Diseases, Pediatric Institute of Southern Switzerland, Regional Hospital of Bellinzona, Bellinzona, Switzerland
| | | | - Marie-Helene Perez
- Pediatric Intensive Care Unit, Department Women-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland
| | - Damien Schaffner
- Pediatric Cardiology Unit, Department Women-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland
| | - Sandra Andrea Asner
- Pediatric Infectious Diseases and Vaccinology Unit, Department Women-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland
| | - Michael Hofer
- Pediatric Immuno-Rheumatology of Western Switzerland, Department Women-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland.,Pediatric Immuno-Rheumatology, Department of Paediatrics, University Hospital, Geneva, Switzerland
| |
Collapse
|
30
|
Gonzalez-Dias P, Lee EK, Sorgi S, de Lima DS, Urbanski AH, Silveira EL, Nakaya HI. Methods for predicting vaccine immunogenicity and reactogenicity. Hum Vaccin Immunother 2019; 16:269-276. [PMID: 31869262 PMCID: PMC7062420 DOI: 10.1080/21645515.2019.1697110] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/13/2019] [Accepted: 11/18/2019] [Indexed: 12/28/2022] Open
Abstract
Subjects receiving the same vaccine often show different levels of immune responses and some may even present adverse side effects to the vaccine. Systems vaccinology can combine omics data and machine learning techniques to obtain highly predictive signatures of vaccine immunogenicity and reactogenicity. Currently, several machine learning methods are already available to researchers with no background in bioinformatics. Here we described the four main steps to discover markers of vaccine immunogenicity and reactogenicity: (1) Preparing the data; (2) Selecting the vaccinees and relevant genes; (3) Choosing the algorithm; (4) Blind testing your model. With the increasing number of Systems Vaccinology datasets being generated, we expect that the accuracy and robustness of signatures of vaccine reactogenicity and immunogenicity will significantly improve.
Collapse
Affiliation(s)
- Patrícia Gonzalez-Dias
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Eva K. Lee
- The Center for Operations Research in Medicine and HealthCare, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sara Sorgi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Diógenes S. de Lima
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alysson H. Urbanski
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Eduardo Lv Silveira
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Helder I. Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Scientific Platform Pasteur, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
31
|
Suschak JJ, Schmaljohn CS. Vaccines against Ebola virus and Marburg virus: recent advances and promising candidates. Hum Vaccin Immunother 2019; 15:2359-2377. [PMID: 31589088 DOI: 10.1080/21645515.2019.1651140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The filoviruses Ebola virus and Marburg virus are among the most dangerous pathogens in the world. Both viruses cause viral hemorrhagic fever, with case fatality rates of up to 90%. Historically, filovirus outbreaks had been relatively small, with only a few hundred cases reported. However, the recent West African Ebola virus outbreak underscored the threat that filoviruses pose. The three year-long outbreak resulted in 28,646 Ebola virus infections and 11,323 deaths. The lack of Food and Drug Administration (FDA) licensed vaccines and antiviral drugs hindered early efforts to contain the outbreak. In response, the global scientific community has spurred the advanced development of many filovirus vaccine candidates. Novel vaccine platforms, such as viral vectors and DNA vaccines, have emerged, leading to the investigation of candidate vaccines that have demonstrated protective efficacy in small animal and nonhuman primate studies. Here, we will discuss several of these vaccine platforms with a particular focus on approaches that have advanced into clinical development.
Collapse
Affiliation(s)
- John J Suschak
- Virology Division, U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | - Connie S Schmaljohn
- Headquarters Division, U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| |
Collapse
|
32
|
Hervé C, Laupèze B, Del Giudice G, Didierlaurent AM, Tavares Da Silva F. The how's and what's of vaccine reactogenicity. NPJ Vaccines 2019; 4:39. [PMID: 31583123 PMCID: PMC6760227 DOI: 10.1038/s41541-019-0132-6] [Citation(s) in RCA: 309] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 08/02/2019] [Indexed: 12/12/2022] Open
Abstract
Reactogenicity represents the physical manifestation of the inflammatory response to vaccination, and can include injection-site pain, redness, swelling or induration at the injection site, as well as systemic symptoms, such as fever, myalgia, or headache. The experience of symptoms following vaccination can lead to needle fear, long-term negative attitudes and non-compliant behaviours, which undermine the public health impact of vaccination. This review presents current knowledge on the potential causes of reactogenicity, and how host characteristics, vaccine administration and composition factors can influence the development and perception of reactogenicity. The intent is to provide an overview of reactogenicity after vaccination to help the vaccine community, including healthcare professionals, in maintaining confidence in vaccines by promoting vaccination, setting expectations for vaccinees about what might occur after vaccination and reducing anxiety by managing the vaccination setting.
Collapse
|
33
|
Fathi A, Dahlke C, Addo MM. Recombinant vesicular stomatitis virus vector vaccines for WHO blueprint priority pathogens. Hum Vaccin Immunother 2019; 15:2269-2285. [PMID: 31368826 PMCID: PMC6816421 DOI: 10.1080/21645515.2019.1649532] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The devastating Ebola virus (EBOV) outbreak in West Africa in 2013-2016 has flagged the need for the timely development of vaccines for high-threat pathogens. To be better prepared for new epidemics, the WHO has compiled a list of priority pathogens that are likely to cause future outbreaks and for which R&D efforts are, therefore, paramount (R&D Blueprint: https://www.who.int/blueprint/priority-diseases/en/ ). To this end, the detailed characterization of vaccine platforms is needed. The vesicular stomatitis virus (VSV) has been established as a robust vaccine vector backbone for infectious diseases for well over a decade. The recent clinical trials testing the vaccine candidate VSV-EBOV against EBOV disease now have added a substantial amount of clinical data and suggest VSV to be an ideal vaccine vector candidate for outbreak pathogens. In this review, we discuss insights gained from the clinical VSV-EBOV vaccine trials as well as from animal studies investigating vaccine candidates for Blueprint pathogens.
Collapse
Affiliation(s)
- Anahita Fathi
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| | - Christine Dahlke
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| | - Marylyn M Addo
- Department of Medicine, Division of Infectious Diseases, University Medical-Center Hamburg-Eppendorf , Hamburg , Germany.,Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine , Hamburg , Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel-Riems , Germany
| |
Collapse
|
34
|
Monath TP, Fast PE, Modjarrad K, Clarke DK, Martin BK, Fusco J, Nichols R, Heppner DG, Simon JK, Dubey S, Troth SP, Wolf J, Singh V, Coller BA, Robertson JS. rVSVΔG-ZEBOV-GP (also designated V920) recombinant vesicular stomatitis virus pseudotyped with Ebola Zaire Glycoprotein: Standardized template with key considerations for a risk/benefit assessment. Vaccine X 2019; 1:100009. [PMID: 31384731 PMCID: PMC6668225 DOI: 10.1016/j.jvacx.2019.100009] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 12/07/2018] [Indexed: 12/14/2022] Open
Abstract
The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to evaluate the safety and characteristics of live, recombinant viral vector vaccines. A recent publication by the V3SWG described live, attenuated, recombinant vesicular stomatitis virus (rVSV) as a chimeric virus vaccine for HIV-1 (Clarke et al., 2016). The rVSV vector system is being explored as a platform for development of multiple vaccines. This paper reviews the molecular and biological features of the rVSV vector system, followed by a template with details on the safety and characteristics of a rVSV vaccine against Zaire ebolavirus (ZEBOV). The rVSV-ZEBOV vaccine is a live, replication competent vector in which the VSV glycoprotein (G) gene is replaced with the glycoprotein (GP) gene of ZEBOV. Multiple copies of GP are expressed and assembled into the viral envelope responsible for inducing protective immunity. The vaccine (designated V920) was originally constructed by the National Microbiology Laboratory, Public Health Agency of Canada, further developed by NewLink Genetics Corp. and Merck & Co., and is now in final stages of registration by Merck. The vaccine is attenuated by deletion of the principal virulence factor of VSV (the G protein), which also removes the primary target for anti-vector immunity. The V920 vaccine caused no toxicities after intramuscular (IM) or intracranial injection of nonhuman primates and no reproductive or developmental toxicity in a rat model. In multiple studies, cynomolgus macaques immunized IM with a wide range of virus doses rapidly developed ZEBOV-specific antibodies measured in IgG ELISA and neutralization assays and were fully protected against lethal challenge with ZEBOV virus. Over 20,000 people have received the vaccine in clinical trials; the vaccine has proven to be safe and well tolerated. During the first few days after vaccination, many vaccinees experience a mild acute-phase reaction with fever, headache, myalgia, and arthralgia of short duration; this period is associated with a low-level viremia, activation of anti-viral genes, and increased levels of chemokines and cytokines. Oligoarthritis and rash appearing in the second week occur at a low incidence, and are typically mild-moderate in severity and self-limited. V920 vaccine was used in a Phase III efficacy trial during the West African Ebola epidemic in 2015, showing 100% protection against Ebola Virus Disease, and it has subsequently been deployed for emergency control of Ebola outbreaks in central Africa. The template provided here provides a comprehensive picture of the first rVSV vector to reach the final stage of development and to provide a solution to control of an alarming human disease.
Collapse
Affiliation(s)
| | - Patricia E. Fast
- International AIDS Vaccine Initiative, New York, NY 10004, United States
| | - Kayvon Modjarrad
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, United States
| | | | | | - Joan Fusco
- NewLink Genetics Corp, Ames, IA, United States
| | | | | | | | - Sheri Dubey
- Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Sean P. Troth
- Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Jayanthi Wolf
- Merck & Co., Inc., Kenilworth, NJ 07033, United States
| | - Vidisha Singh
- Immunology and Molecular Pathogenesis, Emory University, Atlanta, GA 30322, United States
| | | | | | | |
Collapse
|
35
|
Lukashevich IS, Paessler S, de la Torre JC. Lassa virus diversity and feasibility for universal prophylactic vaccine. F1000Res 2019; 8. [PMID: 30774934 PMCID: PMC6357994 DOI: 10.12688/f1000research.16989.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2019] [Indexed: 12/14/2022] Open
Abstract
Lassa virus (LASV) is a highly prevalent mammarenavirus in West Africa and is maintained in nature in a persistently infected rodent host, Mastomys natalensis, which is widely spread in sub-Saharan Africa. LASV infection of humans can cause Lassa fever (LF), a disease associated with high morbidity and significant mortality. Recent evidence indicates an LASV expansion outside its traditional endemic areas. In 2017, the World Health Organization (WHO) included LASV in top-priority pathogens and released a Target Product Profile (TPP) for vaccine development. Likewise, in 2018, the US Food and Drug Administration added LF to a priority review voucher program to encourage the development of preventive and therapeutics measures. In this article, we review recent progress in LASV vaccine research and development with a focus on the impact of LASV genetic and biological diversity on the design and development of vaccine candidates meeting the WHO's TPP for an LASV vaccine.
Collapse
Affiliation(s)
- Igor S Lukashevich
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40292, USA
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA
| | - Juan Carlos de la Torre
- Department of Immunology and Microbiology IMM-6, The Scripps Research Institute, La Jolla, CA, 92037, USA
| |
Collapse
|
36
|
Dong F, Li D, Wen D, Li S, Zhao C, Qi Y, Jangra RK, Wu C, Xia D, Zhang X, Deng F, Chandran K, Zou Z, Yuan F, Zheng A. Single dose of a rVSV-based vaccine elicits complete protection against severe fever with thrombocytopenia syndrome virus. NPJ Vaccines 2019; 4:5. [PMID: 30701094 PMCID: PMC6347601 DOI: 10.1038/s41541-018-0096-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 12/12/2018] [Indexed: 11/09/2022] Open
Abstract
Severe fever with thrombocytopenia virus (SFTSV) is an emerging tick-borne phlebovirus that causes lethal human disease, for which there are no licensed antiviral vaccines or therapies. Herein, we developed a live attenuated recombinant vesicular stomatitis virus (rVSV)-based vaccine candidate expressing the SFTSV Gn/Gc glycoproteins (rVSV-SFTSV/AH12-GP). High titers of cross-protective, broadly neutralizing antibodies were elicited by a single dose of rVSV-SFTSV/AH12-GP in both immunocompetent and immunocompromised mice against multiple strains of SFTSV and the related but distinct phlebovirus Heartland virus (HRTV). Remarkably, complete protection against lethal challenge with SFTSV was conferred in young and old immunocompromised mice irrespective of any pre-existing vector-specific immunity. Collectively, these results suggest that a rVSV vector expressing SFTSV glycoproteins is a promising candidate vaccine against two emerging phleboviruses associated with severe human diseases.
Collapse
Affiliation(s)
- Fangfang Dong
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Dandan Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Dan Wen
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Suhua Li
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chaoyue Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yue Qi
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Rohit K. Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY USA
| | - Cuiping Wu
- Department of Infectious Disease, Yidu Central Hospital of Weifang, Weifang, China
| | - Dequan Xia
- Department of Infectious Disease, Yidu Central Hospital of Weifang, Weifang, China
| | - Xing Zhang
- University of Chinese Academy of Sciences, Beijing, China
| | - Fei Deng
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY USA
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fei Yuan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Aihua Zheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- College of Life Science, Henan Normal University, Xinxiang, China
| |
Collapse
|
37
|
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: 1.4] [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
|
38
|
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: 3.9] [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]
|
39
|
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
|
40
|
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: 100] [Impact Index Per Article: 14.3] [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
|
41
|
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: 0.6] [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
|
42
|
Denoel P, Londoño-Hayes P, Chlebus M, de Azero MR. Impact of the Innovative Medicines Initiative on vaccine development. Nat Rev Drug Discov 2018; 17:769-770. [PMID: 29853715 DOI: 10.1038/nrd.2018.72] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | | | - Magda Chlebus
- European Federation of Pharmaceutical Industries and Associations (EFPIA), Brussels, Belgium
| | | |
Collapse
|
43
|
Huttner A, Agnandji ST, Combescure C, Fernandes JF, Bache EB, Kabwende L, Ndungu FM, Brosnahan J, Monath TP, Lemaître B, Grillet S, Botto M, Engler O, Portmann J, Siegrist D, Bejon P, Silvera P, Kremsner P, Siegrist CA. Determinants of antibody persistence across doses and continents after single-dose rVSV-ZEBOV vaccination for Ebola virus disease: an observational cohort study. THE LANCET. INFECTIOUS DISEASES 2018; 18:738-748. [PMID: 29627147 DOI: 10.1016/s1473-3099(18)30165-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 02/08/2018] [Accepted: 02/14/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND The recombinant vesicular stomatitis virus (rVSV) vaccine expressing the Zaire Ebola virus (ZEBOV) glycoprotein is efficacious in the weeks following single-dose injection, but duration of immunity is unknown. We aimed to assess antibody persistence at 1 and 2 years in volunteers who received single-dose rVSV-ZEBOV in three previous trials. METHODS In this observational cohort study, we prospectively followed-up participants from the African and European phase 1 rVSV-ZEBOV trials, who were vaccinated once in 2014-15 with 300 000 (low dose) or 10-50 million (high dose) plaque-forming units (pfu) of rVSV-ZEBOV vaccine to assess ZEBOV glycoprotein (IgG) antibody persistence. The primary outcome was ZEBOV glycoprotein-specific IgG geometric mean concentrations (GMCs) measured yearly by ELISA compared with 1 month (ie, 28 days) after immunisation. We report GMCs up to 2 years (Geneva, Switzerland, including neutralising antibodies up to 6 months) and 1 year (Lambaréné, Gabon; Kilifi, Kenya) after vaccination and factors associated with higher antibody persistence beyond 6 months, according to multivariable analyses. Trials and the observational study were registered at ClinicalTrials.gov (Geneva: NCT02287480 and NCT02933931; Kilifi: NCT02296983) and the Pan-African Clinical Trials Registry (Lambaréné PACTR201411000919191). FINDINGS Of 217 vaccinees from the original studies (102 from the Geneva study, 75 from the Lambaréné study, and 40 from the Kilifi study), 197 returned and provided samples at 1 year (95 from the Geneva study, 63 from the Lambaréné, and 39 from the Kilifi study) and 90 at 2 years (all from the Geneva study). In the Geneva group, 44 (100%) of 44 participants who had been given a high dose (ie, 10-50 million pfu) of vaccine and who were seropositive at day 28 remained seropositive at 2 years, whereas 33 (89%) of 37 who had been given the low dose (ie, 300 000 pfu) remained seropositive for 2 years (p=0·042). In participants who had received a high dose, ZEBOV glycoprotein IgG GMCs decreased significantly between their peak (at 1-3 months) and month 6 after vaccination in Geneva (p<0·0001) and Lambaréné (p=0·0298) but not in Kilifi (p=0·5833) and subsequently remained stable at all sites apart from Geneva, where GMC in those given a high dose of vaccine increased significantly between 6 months and 1 year (p=0·0264). Antibody persistence was similar at 1 year and at 6 months in those who had received a low dose of vaccine, with lower titres among participants from the Geneva study at 2 years than at 1 year after vaccination (GMC ratio 0·61, 95% CI 0·49-0·77; p<0·0001). In multivariable analyses, predictors of increased IgG GMCs beyond 6 months included high-dose versus low-dose vaccination (Geneva p=0·0133; Lambaréné p=0·008) and vaccine-related arthritis (p=0·0176), but not sex, age, or baseline seropositivity (all p>0·05). Neutralising antibodies seem to be less durable, with seropositivity dropping from 64-71% at 28 days to 27-31% at 6 months in participants from the Geneva study. INTERPRETATION Antibody responses to single-dose rVSV-ZEBOV vaccination are sustained across dose ranges and settings, a key criterion in countries where booster vaccinations would be impractical. FUNDING The Wellcome Trust and Innovative Medicines Initiative 2 Joint Undertaking.
Collapse
Affiliation(s)
- Angela Huttner
- Division of Infectious Diseases, University Hospitals of Geneva, Geneva, Switzerland; Infection Control Programme, University Hospitals of Geneva, Geneva, Switzerland; Centre for Vaccinology, University Hospitals of Geneva, Geneva, Switzerland
| | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné, Hôpital Albert Schweitzer, Lambaréné, Gabon; Institut für Tropenmedizin and German Centre for Infection Research (DZIF) partner sites, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Christophe Combescure
- Division of Clinical Epidemiology, University Hospitals of Geneva, Geneva, Switzerland
| | - José F Fernandes
- Centre de Recherches Médicales de Lambaréné, Hôpital Albert Schweitzer, Lambaréné, Gabon
| | - Emmanuel Bache Bache
- Centre de Recherches Médicales de Lambaréné, Hôpital Albert Schweitzer, Lambaréné, Gabon
| | - Lumeka Kabwende
- Centre de Recherches Médicales de Lambaréné, Hôpital Albert Schweitzer, Lambaréné, Gabon
| | | | - Jessica Brosnahan
- Centre de Recherches Médicales de Lambaréné, Hôpital Albert Schweitzer, Lambaréné, Gabon; Institut für Tropenmedizin and German Centre for Infection Research (DZIF) partner sites, Universitätsklinikum Tübingen, Tübingen, Germany
| | | | - Barbara Lemaître
- Centre for Vaccinology, University Hospitals of Geneva, Geneva, Switzerland
| | - Stéphane Grillet
- Centre for Vaccinology, University Hospitals of Geneva, Geneva, Switzerland
| | - Miriam Botto
- Non-clinical Development, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Olivier Engler
- Spiez Laboratory, Federal Office for Civil Protection, Spiez, Switzerland
| | - Jasmine Portmann
- Spiez Laboratory, Federal Office for Civil Protection, Spiez, Switzerland
| | - Denise Siegrist
- Spiez Laboratory, Federal Office for Civil Protection, Spiez, Switzerland
| | - Philip Bejon
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Peter Silvera
- Non-clinical Development, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Peter Kremsner
- Centre de Recherches Médicales de Lambaréné, Hôpital Albert Schweitzer, Lambaréné, Gabon; Institut für Tropenmedizin and German Centre for Infection Research (DZIF) partner sites, Universitätsklinikum Tübingen, Tübingen, Germany
| | | | | | | | | |
Collapse
|
44
|
McElroy AK, Mühlberger E, Muñoz-Fontela C. Immune barriers of Ebola virus infection. Curr Opin Virol 2018; 28:152-160. [PMID: 29452995 PMCID: PMC5886007 DOI: 10.1016/j.coviro.2018.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/23/2018] [Accepted: 01/26/2018] [Indexed: 01/10/2023]
Abstract
Since its initial emergence in 1976 in northern Democratic Republic of Congo (DRC), Ebola virus (EBOV) has been a global health concern due to its virulence in humans, the mystery surrounding the identity of its host reservoir and the unpredictable nature of Ebola virus disease (EVD) outbreaks. Early after the first clinical descriptions of a disease resembling a 'septic-shock-like syndrome', with coagulation abnormalities and multi-system organ failure, researchers began to evaluate the role of the host immune response in EVD pathophysiology. In this review, we summarize how data gathered during the last 40 years in the laboratory as well as in the field have provided insight into EBOV immunity. From molecular mechanisms involved in EBOV recognition in infected cells, to antigen processing and adaptive immune responses, we discuss current knowledge on the main immune barriers of infection as well as outstanding research questions.
Collapse
Affiliation(s)
- Anita K McElroy
- Division of Pediatric Infectious Disease, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh of UPMC, 3501 Fifth Ave, Pittsburgh, PA 15261, USA
| | - Elke Mühlberger
- Department of Microbiology and National Emerging Infectious Diseases Laboratories, Boston University School of Medicine, 620 Albany Street, 02118 Boston, MA, USA
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Strasse 74, 20359 Hamburg, Germany; German Center for Infection Research (DZIF), Partner Site Hamburg, Germany.
| |
Collapse
|
45
|
|
46
|
Oluwagbemi O, Awe O. A comparative computational genomics of Ebola Virus Disease strains: In-silico Insight for Ebola control. INFORMATICS IN MEDICINE UNLOCKED 2018. [DOI: 10.1016/j.imu.2018.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
47
|
Agnandji ST, Fernandes JF, Bache EB, Obiang Mba RM, Brosnahan JS, Kabwende L, Pitzinger P, Staarink P, Massinga-Loembe M, Krähling V, Biedenkopf N, Fehling SK, Strecker T, Clark DJ, Staines HM, Hooper JW, Silvera P, Moorthy V, Kieny MP, Adegnika AA, Grobusch MP, Becker S, Ramharter M, Mordmüller B, Lell B, Krishna S, Kremsner PG. Safety and immunogenicity of rVSVΔG-ZEBOV-GP Ebola vaccine in adults and children in Lambaréné, Gabon: A phase I randomised trial. PLoS Med 2017; 14:e1002402. [PMID: 28985239 PMCID: PMC5630143 DOI: 10.1371/journal.pmed.1002402] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/07/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The rVSVΔG-ZEBOV-GP vaccine prevented Ebola virus disease when used at 2 × 107 plaque-forming units (PFU) in a trial in Guinea. This study provides further safety and immunogenicity data. METHODS AND FINDINGS A randomised, open-label phase I trial in Lambaréné, Gabon, studied 5 single intramuscular vaccine doses of 3 × 103, 3 × 104, 3 × 105, 3 × 106, or 2 × 107 PFU in 115 adults and a dose of 2 × 107 PFU in 20 adolescents and 20 children. The primary objective was safety and tolerability 28 days post-injection. Immunogenicity, viraemia, and shedding post-vaccination were evaluated as secondary objectives. In adults, mild-to-moderate adverse events were frequent, but there were no serious or severe adverse events related to vaccination. Before vaccination, Zaire Ebola virus (ZEBOV)-glycoprotein (GP)-specific and ZEBOV antibodies were detected in 11% and 27% of adults, respectively. In adults, 74%-100% of individuals who received a dose 3 × 104, 3 × 105, 3 × 106, or 2 × 107 PFU had a ≥4.0-fold increase in geometric mean titres (GMTs) of ZEBOV-GP-specific antibodies at day 28, reaching GMTs of 489 (95% CI: 264-908), 556 (95% CI: 280-1,101), 1,245 (95% CI: 899-1,724), and 1,503 (95% CI: 931-2,426), respectively. Twenty-two percent of adults had a ≥4-fold increase of ZEBOV antibodies, with GMTs at day 28 of 1,015 (647-1,591), 1,887 (1,154-3,085), 1,445 (1,013-2,062), and 3,958 (2,249-6,967) for the same doses, respectively. These antibodies persisted up to day 180 for doses ≥3 × 105 PFU. Adults with antibodies before vaccination had higher GMTs throughout. Neutralising antibodies were detected in more than 50% of participants at doses ≥3 × 105 PFU. As in adults, no serious or severe adverse events related to vaccine occurred in adolescents or children. At day 2, vaccine RNA titres were higher for adolescents and children than adults. At day 7, 78% of adolescents and 35% of children had recombinant vesicular stomatitis virus RNA detectable in saliva. The vaccine induced high GMTs of ZEBOV-GP-specific antibodies at day 28 in adolescents, 1,428 (95% CI: 1,025-1,989), and children, 1,620 (95% CI: 806-3,259), and in both groups antibody titres increased up to day 180. The absence of a control group, lack of stratification for baseline antibody status, and imbalances in male/female ratio are the main limitations of this study. CONCLUSIONS Our data confirm the acceptable safety and immunogenicity profile of the 2 × 107 PFU dose in adults and support consideration of lower doses for paediatric populations and those who request boosting. TRIAL REGISTRATION Pan African Clinical Trials Registry PACTR201411000919191.
Collapse
Affiliation(s)
- Selidji T. Agnandji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
| | - José F. Fernandes
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
| | | | | | - Jessica S. Brosnahan
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
| | - Lumeka Kabwende
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Paul Pitzinger
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Bernhard Nocht Hospital for Tropical Diseases, Bernhard Nocht Institute for Tropical Medicine and University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pieter Staarink
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Verena Krähling
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
- Institute for Virology, Philipps-Universität Marburg, Marburg, Germany
| | - Nadine Biedenkopf
- Institute for Virology, Philipps-Universität Marburg, Marburg, Germany
| | | | - Thomas Strecker
- Institute for Virology, Philipps-Universität Marburg, Marburg, Germany
| | - David J. Clark
- Centre for Diagnostics and Antimicrobial Resistance, Institute for Infection & Immunity, St. George’s, University of London, London, United Kingdom
| | - Henry M. Staines
- Centre for Diagnostics and Antimicrobial Resistance, Institute for Infection & Immunity, St. George’s, University of London, London, United Kingdom
| | - Jay W. Hooper
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Peter Silvera
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | | | | | - Akim A. Adegnika
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin P. Grobusch
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Stephan Becker
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
- Institute for Virology, Philipps-Universität Marburg, Marburg, Germany
| | - Michael Ramharter
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Bernhard Nocht Hospital for Tropical Diseases, Bernhard Nocht Institute for Tropical Medicine and University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Mordmüller
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
| | - Bertrand Lell
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
| | | | - Sanjeev Krishna
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- Centre for Diagnostics and Antimicrobial Resistance, Institute for Infection & Immunity, St. George’s, University of London, London, United Kingdom
- St. George’s University Hospitals NHS Foundation Trust, London, United Kingdom
- * E-mail: (SK); (PGK)
| | - Peter G. Kremsner
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universitätsklinikum Tübingen, Tübingen, Germany
- German Centre for Infection Research (DZIF) partner sites Universitätsklinikum Tübingen and Gießen-Marburg-Langen, Germany
- * E-mail: (SK); (PGK)
| |
Collapse
|
48
|
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: 101] [Impact Index Per Article: 12.6] [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
|
49
|
Medaglini D, Siegrist CA. Immunomonitoring of human responses to the rVSV-ZEBOV Ebola vaccine. Curr Opin Virol 2017; 23:88-94. [PMID: 28460340 DOI: 10.1016/j.coviro.2017.03.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 03/05/2017] [Accepted: 03/15/2017] [Indexed: 11/27/2022]
Abstract
The rVSV-ZEBOV vaccine is currently the only Ebola vaccine with demonstrated clinical efficacy in a ring-vaccination clinical trial. It has been shown to be reactogenic but immunogenic and safe in several Phase I clinical studies. However, its mechanisms of protection are unknown and available immunogenicity data are mostly limited to classical serological analysis; it is now of paramount importance to apply cutting-edge technologies, including transcriptomic and metabolomic analyses, and to perform integrative analyses with standard serology and clinical data to comprehensively profile the rVSV-ZEBOV immune signature.
Collapse
Affiliation(s)
- Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Claire-Anne Siegrist
- World Health Organization Collaborating Center for Vaccine Immunology, Departments of Pathology-Immunology, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|