51
|
Venkatraman N, Ndiaye BP, Bowyer G, Wade D, Sridhar S, Wright D, Powlson J, Ndiaye I, Dièye S, Thompson C, Bakhoum M, Morter R, Capone S, Del Sorbo M, Jamieson S, Rampling T, Datoo M, Roberts R, Poulton I, Griffiths O, Ballou WR, Roman F, Lewis DJM, Lawrie A, Imoukhuede E, Gilbert SC, Dieye TN, Ewer KJ, Mboup S, Hill AVS. Safety and Immunogenicity of a Heterologous Prime-Boost Ebola Virus Vaccine Regimen in Healthy Adults in the United Kingdom and Senegal. J Infect Dis 2020; 219:1187-1197. [PMID: 30407513 PMCID: PMC6452431 DOI: 10.1093/infdis/jiy639] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/04/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The 2014 West African outbreak of Ebola virus disease highlighted the urgent need to develop an effective Ebola vaccine. METHODS We undertook 2 phase 1 studies assessing safety and immunogenicity of the viral vector modified vaccinia Ankara virus vectored Ebola Zaire vaccine (MVA-EBO-Z), manufactured rapidly on a new duck cell line either alone or in a heterologous prime-boost regimen with recombinant chimpanzee adenovirus type 3 vectored Ebola Zaire vaccine (ChAd3-EBO-Z) followed by MVA-EBO-Z. Adult volunteers in the United Kingdom (n = 38) and Senegal (n = 40) were vaccinated and an accelerated 1-week prime-boost regimen was assessed in Senegal. Safety was assessed by active and passive collection of local and systemic adverse events. RESULTS The standard and accelerated heterologous prime-boost regimens were well-tolerated and elicited potent cellular and humoral immunogenicity in the United Kingdom and Senegal, but vaccine-induced antibody responses were significantly lower in Senegal. Cellular immune responses measured by flow cytometry were significantly greater in African vaccinees receiving ChAd3 and MVA vaccines in the same rather than the contralateral limb. CONCLUSIONS MVA biomanufactured on an immortalized duck cell line shows potential for very large-scale manufacturing with lower cost of goods. This first trial of MVA-EBO-Z in humans encourages further testing in phase 2 studies, with the 1-week prime-boost interval regimen appearing to be particularly suitable for outbreak control. CLINICAL TRIALS REGISTRATION NCT02451891; NCT02485912.
Collapse
Affiliation(s)
| | | | | | - Djibril Wade
- Centre Hospitalier Universitaire le Dantec, Dakar, Senegal
| | | | - Daniel Wright
- Jenner Institute, University of Oxford, United Kingdom
| | | | | | - Siry Dièye
- Centre Hospitalier Universitaire le Dantec, Dakar, Senegal
| | | | - Momar Bakhoum
- Centre Hospitalier Universitaire le Dantec, Dakar, Senegal
| | | | | | | | | | | | - Mehreen Datoo
- Jenner Institute, University of Oxford, United Kingdom
| | | | - Ian Poulton
- Jenner Institute, University of Oxford, United Kingdom
| | | | | | | | - David J M Lewis
- National Institute for Health Research/Imperial Clinical Research Facility, Hammersmith Hospital, London, United Kingdom
| | - Alison Lawrie
- Jenner Institute, University of Oxford, United Kingdom
| | | | | | | | - Katie J Ewer
- Jenner Institute, University of Oxford, United Kingdom
| | | | | |
Collapse
|
52
|
Ebola virus disease: An emerging and re-emerging viral threat. J Autoimmun 2019; 106:102375. [PMID: 31806422 DOI: 10.1016/j.jaut.2019.102375] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/19/2019] [Accepted: 11/21/2019] [Indexed: 12/21/2022]
Abstract
The genus Ebolavirus from the family Filoviridae is composed of five species including Sudan ebolavirus, Reston ebolavirus, Bundibugyo ebolavirus, Taï Forest ebolavirus, and Ebola virus (previously known as Zaire ebolavirus). These viruses have a large non-segmented, negative-strand RNA of approximately 19 kb that encodes for glycoproteins (i.e., GP, sGP, ssGP), nucleoproteins, virion proteins (i.e., VP 24, 30,40) and an RNA dependent RNA polymerase. These viruses have become a global health concern because of mortality, their rapid dissemination, new outbreaks in West-Africa, and the emergence of a new condition known as "Post-Ebola virus disease syndrome" that resembles inflammatory and autoimmune conditions such as rheumatoid arthritis, systemic lupus erythematosus and spondyloarthritis with uveitis. However, there are many gaps in the understanding of the mechanisms that may induce the development of such autoimmune-like syndromes. Some of these mechanisms may include a high formation of neutrophil extracellular traps, an uncontrolled "cytokine storm", and the possible formation of auto-antibodies. The likely appearance of autoimmune phenomena in Ebola survivors suppose a new challenge in the management and control of this disease and opens a new field of research in a special subgroup of patients. Herein, the molecular biology, pathogenesis, clinical manifestations, and treatment of Ebola virus disease are reviewed and some strategies for control of disease are discussed.
Collapse
|
53
|
Poetsch JH, Dahlke C, Zinser ME, Kasonta R, Lunemann S, Rechtien A, Ly ML, Stubbe HC, Krähling V, Biedenkopf N, Eickmann M, Fehling SK, Olearo F, Strecker T, Sharma P, Lang KS, Lohse AW, Schmiedel S, Becker S, Addo MM. Detectable Vesicular Stomatitis Virus (VSV)-Specific Humoral and Cellular Immune Responses Following VSV-Ebola Virus Vaccination in Humans. J Infect Dis 2019; 219:556-561. [PMID: 30452666 PMCID: PMC6350948 DOI: 10.1093/infdis/jiy565] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 11/13/2018] [Indexed: 01/16/2023] Open
Abstract
In response to the Ebola virus (EBOV) crisis of 2013–2016, a recombinant vesicular stomatitis virus (VSV)–based EBOV vaccine was clinically tested (NCT02283099). A single-dose regimen of VSV-EBOV revealed a safe and immunogenic profile and demonstrated clinical efficacy. While EBOV-specific immune responses to this candidate vaccine have previously been investigated, limited human data on immunity to the VSV vector are available. Within the scope of a phase 1 study, we performed a comprehensive longitudinal analysis of adaptive immune responses to internal VSV proteins following VSV-EBOV immunization. While no preexisting immunity to the vector was observed, more than one-third of subjects developed VSV-specific cytotoxic T-lymphocyte responses and antibodies.
Collapse
Affiliation(s)
- Joseph H Poetsch
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| | - Christine Dahlke
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| | - Madeleine E Zinser
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| | - Rahel Kasonta
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems
| | - Sebastian Lunemann
- Leibniz Institute for Experimental Virology, Heinrich Pette Institute, Hamburg
| | - Anne Rechtien
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf.,Leibniz Institute for Experimental Virology, Heinrich Pette Institute, Hamburg
| | - My L Ly
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| | - Hans C Stubbe
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, Department of Medicine II, Ludwig Maximilian University, Munich
| | - Verena Krähling
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | - Nadine Biedenkopf
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | - Markus Eickmann
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | - Sarah K Fehling
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | - Flaminia Olearo
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg
| | - Thomas Strecker
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | - Piyush Sharma
- Institute of Immunology, University Duisburg-Essen, Essen, Germany
| | - Karl S Lang
- Institute of Immunology, University Duisburg-Essen, Essen, Germany
| | - Ansgar W Lohse
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems
| | - Stefan Schmiedel
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| | - Stephan Becker
- Institute for Virology, Philipps University Marburg.,German Center for Infection Research, partner site Gießen-Marburg-Langen
| | | | - Marylyn M Addo
- First Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg.,German Center for Infection Research, partner site Hamburg-Lübeck-Borstel-Riems.,Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf
| |
Collapse
|
54
|
Tian X, Chen D, Wang H, Xu S, Zhu L, Wu X, Wu Z. The induction and characterization of monoclonal antibodies specific to GP of Ebola virus. J Med Virol 2019; 92:996-1006. [PMID: 31663613 DOI: 10.1002/jmv.25615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 10/24/2019] [Indexed: 01/18/2023]
Abstract
The Ebola virus is highly infectious and characterized by hemorrhagic fever, headache, and so on with a high mortality rate. Currently, there are neither therapeutic drugs or vaccines against the Ebola virus nor fast diagnostic methods for the detection of Ebola virus infection. This study reported the induction and isolation of two monoclonal antibodies that specifically recognized the glycoprotein (GP) and secreted glycoprotein (sGP) of the Ebola virus. Plasmids encoding either GP or sGP were constructed and immunized BALB/c mice, accordingly purified sGP was boosted. The antisera were analyzed for binding activity against sGP protein in enzyme-linked immunosorbent assay (ELISA) and neutralization activity in a pseudotyped virus neutralization assay. A number of reactive clones were isolated and two monoclonal antibodies T231 and T242 were identified to react with both GP and sGP. Western blot and ELISA assays showed that the monoclonal antibodies could react with GP and sGP, respectively. Moreover, they could recognize Ebola pseudovirus by cellular immunochemistry assay. We labeled the monoclonal antibody T231 with biotin and analyzed the competitiveness of the two antibodies by the ELISA test. The results showed that the binding epitopes of the two monoclonal antibodies to sGP were partially overlapped. In summary, two GP-specific mAbs were identified, which will be used to detect the Ebola virus or investigate GP.
Collapse
Affiliation(s)
- Xiaoyan Tian
- Department of General Surgery, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China.,Center for Public Health Research, Nanjing University, Nanjing, China
| | - Deyan Chen
- Center for Public Health Research, Nanjing University, Nanjing, China
| | - Huanru Wang
- Center for Public Health Research, Nanjing University, Nanjing, China
| | - Shijie Xu
- Center for Public Health Research, Nanjing University, Nanjing, China
| | - Linjing Zhu
- Y-Clone Medical Science Co Ltd, Nanjing, China
| | - Xilin Wu
- Center for Public Health Research, Nanjing University, Nanjing, China.,Y-Clone Medical Science Co Ltd, Nanjing, China
| | - Zhiwei Wu
- Center for Public Health Research, Nanjing University, Nanjing, China.,State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
55
|
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: 3.4] [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
|
56
|
Gélinas JF, Azizi H, Kiesslich S, Lanthier S, Perdersen J, Chahal PS, Ansorge S, Kobinger G, Gilbert R, Kamen AA. Production of rVSV-ZEBOV in serum-free suspension culture of HEK 293SF cells. Vaccine 2019; 37:6624-6632. [DOI: 10.1016/j.vaccine.2019.09.044] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/28/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022]
|
57
|
Olukitibi TA, Ao Z, Mahmoudi M, Kobinger GA, Yao X. Dendritic Cells/Macrophages-Targeting Feature of Ebola Glycoprotein and its Potential as Immunological Facilitator for Antiviral Vaccine Approach. Microorganisms 2019; 7:E402. [PMID: 31569539 PMCID: PMC6843631 DOI: 10.3390/microorganisms7100402] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/19/2019] [Accepted: 09/26/2019] [Indexed: 01/06/2023] Open
Abstract
In the prevention of epidemic and pandemic viral infection, the use of the antiviral vaccine has been the most successful biotechnological and biomedical approach. In recent times, vaccine development studies have focused on recruiting and targeting immunogens to dendritic cells (DCs) and macrophages to induce innate and adaptive immune responses. Interestingly, Ebola virus (EBOV) glycoprotein (GP) has a strong binding affinity with DCs and macrophages. Shreds of evidence have also shown that the interaction between EBOV GP with DCs and macrophages leads to massive recruitment of DCs and macrophages capable of regulating innate and adaptive immune responses. Therefore, studies for the development of vaccine can utilize the affinity between EBOV GP and DCs/macrophages as a novel immunological approach to induce both innate and acquired immune responses. In this review, we will discuss the unique features of EBOV GP to target the DC, and its potential to elicit strong immune responses while targeting DCs/macrophages. This review hopes to suggest and stimulate thoughts of developing a stronger and effective DC-targeting vaccine for diverse virus infection using EBOV GP.
Collapse
Affiliation(s)
- Titus Abiola Olukitibi
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Zhujun Ao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Mona Mahmoudi
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| | - Gary A Kobinger
- Centre de Recherche en Infectiologie de l' Université Laval/Centre Hospitalier de l' Université Laval (CHUL), Québec, QC G1V 4G2, Canada.
| | - Xiaojian Yao
- Laboratory of Molecular Human Retrovirology, Department of Medical Microbiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada.
| |
Collapse
|
58
|
Guo Q, Chan JFW, Poon VKM, Wu S, Chan CCS, Hou L, Yip CCY, Ren C, Cai JP, Zhao M, Zhang AJ, Song X, Chan KH, Wang B, Kok KH, Wen Y, Yuen KY, Chen W. Immunization With a Novel Human Type 5 Adenovirus-Vectored Vaccine Expressing the Premembrane and Envelope Proteins of Zika Virus Provides Consistent and Sterilizing Protection in Multiple Immunocompetent and Immunocompromised Animal Models. J Infect Dis 2019; 218:365-377. [PMID: 29617816 DOI: 10.1093/infdis/jiy187] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/28/2018] [Indexed: 12/30/2022] Open
Abstract
Background Zika virus (ZIKV) infection may be associated with severe complications and disseminated via both vector-borne and nonvector-borne routes. Adenovirus-vectored vaccines represent a favorable controlling measure for the ZIKV epidemic because they have been shown to be safe, immunogenic, and rapidly generable for other emerging viral infections. Evaluations of 2 previously reported adenovirus-vectored ZIKV vaccines were performed using nonlethal animal models and/or nonepidemic ZIKV strain. Methods We constructed 2 novel human adenovirus 5 (Ad5)-vectored vaccines containing the ZIKV premembrane-envelope (Ad5-Sig-prM-Env) and envelope (Ad5-Env) proteins, respectively, and evaluated them in multiple nonlethal and lethal animal models using epidemic ZIKV strains. Results Both vaccines elicited robust humoral and cellular immune responses in immunocompetent BALB/c mice. Dexamethasone-immunosuppressed mice vaccinated with either vaccine demonstrated robust and durable antibody responses and significantly lower blood and tissue viral loads than controls (P < .05). Similar findings were also observed in interferon-α/β receptor-deficient A129 mice. In both of these immunocompromised animal models, Ad5-Sig-prM-Env-vaccinated mice had significantly (P < .05) higher titers of anti-ZIKV-specific neutralizing antibody titers and lower (undetectable) viral loads than Ad5-Env-vaccinated mice. The close correlation between the neutralizing antibody titer and viral load helped to explain the better protective effect of Ad5-Sig-prM-Env than Ad5-Env. Anamnestic response was absent in Ad5-Sig-prM-Env-vaccinated A129 mice. Conclusions Ad5-Sig-prM-Env provided sterilizing protection against ZIKV infection in mice.
Collapse
Affiliation(s)
- Qiang Guo
- Beijing Institute of Biotechnology, China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Vincent Kwok-Man Poon
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, China
| | - Chris Chung-Sing Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, China
| | - Cyril Chik-Yan Yip
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | | | - Jian-Piao Cai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | | | - Anna Jinxia Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | | | - Kwok-Hung Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Busen Wang
- Beijing Institute of Biotechnology, China
| | - Kin-Hang Kok
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yanbo Wen
- Beijing Institute of Biotechnology, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Wei Chen
- Beijing Institute of Biotechnology, China
| |
Collapse
|
59
|
Silva AC, Moreira JN, Lobo JMS, Almeida H. Advances in Vaccines. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2019; 171:155-188. [PMID: 31446443 PMCID: PMC7120466 DOI: 10.1007/10_2019_107] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Vaccines represent one of the most important advances in science and medicine, helping people around the world in preventing the spread of infectious diseases. However, there are still gaps in vaccination programs in many countries. Out of 11.2 million children born in EU region, more than 500,000 infants did not receive the complete three-dose series of diphtheria, pertussis, and tetanus vaccine before the first birthday. Data shows that there were more than 30,000 measles cases in the European region in recent years, and measles cases are rising in the USA. There are about 20 million children in the world still not getting adequate coverage of basic vaccines. Emerging infectious diseases such as malaria, Ebola virus disease, and Zika virus disease also threaten public health around the world. This chapter provides an overview of recent advances in vaccine development and technologies, manufacturing, characterization of various vaccines, challenges, and strategies in vaccine clinical development. It also provides an overview of recently approved major vaccines for human use.
Collapse
Affiliation(s)
- Ana Catarina Silva
- grid.5808.50000 0001 1503 7226UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - João Nuno Moreira
- grid.8051.c0000 0000 9511 4342Center for Neurosciences and Cell Biology (CNC) and Faculty of Pharmacy (FFUC), University of Coimbra, Coimbra, Portugal
| | - José Manuel Sousa Lobo
- grid.5808.50000 0001 1503 7226UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Hugo Almeida
- grid.5808.50000 0001 1503 7226UCIBIO, REQUIMTE, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
60
|
Abstract
Over 40 years since the discovery of Ebola virus, the anti-Ebola virus vaccine efforts of the past 2 decades have culminated in over 12 different vaccine candidates that have been placed into a number of clinical trial phases, past and present. Of these 12 vaccines, four candidates are up to or in phase II clinical trials, and only one has completed the phase III clinical trial stage. While remarkable progress toward a national regulatory agency-approved vaccine for Ebola virus has been made, there remain unanswered questions on issues such as, but not limited to, vaccine protective immunity and duration of that immunity, and the appropriate vaccination strategy for those at risk of Ebola virus infection.
Collapse
Affiliation(s)
- Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MO, USA
| | - Chad E Mire
- Galveston National Laboratory, and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
61
|
Muñoz-Fontela C, McElroy AK. Ebola Virus Disease in Humans: Pathophysiology and Immunity. Curr Top Microbiol Immunol 2019; 411:141-169. [PMID: 28653186 PMCID: PMC7122202 DOI: 10.1007/82_2017_11] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Viruses of the Ebolavirus genus cause sporadic epidemics of severe and systemic febrile disease that are fueled by human-to-human transmission. Despite the notoriety of ebolaviruses, particularly Ebola virus (EBOV), as prominent viral hemorrhagic fever agents, and the international concern regarding Ebola virus disease (EVD) outbreaks, very little is known about the pathophysiology of EVD in humans and, in particular, about the human immune correlates of survival and immune memory. This lack of basic knowledge about physiological characteristics of EVD is probably attributable to the dearth of clinical and laboratory data gathered from past outbreaks. The unprecedented magnitude of the EVD epidemic that occurred in West Africa from 2013 to 2016 has allowed, for the first time, evaluation of clinical, epidemiological, and immunological parameters in a significant number of patients using state-of-the-art laboratory equipment. This review will summarize the data from the literature regarding human pathophysiologic and immunologic responses to filoviral infection.
Collapse
Affiliation(s)
- César Muñoz-Fontela
- Laboratory of Emerging Viruses, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistraße 52, 20251, Hamburg, Germany.
| | - Anita K McElroy
- Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Drive NE, Atlanta, GA, 30322, USA.
| |
Collapse
|
62
|
Acoustic levitation as a screening method for excipient selection in the development of dry powder vaccines. Int J Pharm 2019; 563:71-78. [DOI: 10.1016/j.ijpharm.2019.03.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/08/2019] [Accepted: 03/12/2019] [Indexed: 11/24/2022]
|
63
|
Zhang D, Jiang N, Chen Q. Vaccination with recombinant adenoviruses expressing Toxoplasma gondii MIC3, ROP9, and SAG2 provide protective immunity against acute toxoplasmosis in mice. Vaccine 2019; 37:1118-1125. [DOI: 10.1016/j.vaccine.2018.12.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/10/2018] [Accepted: 12/16/2018] [Indexed: 12/31/2022]
|
64
|
Rahim MN, Wee EG, He S, Audet J, Tierney K, Moyo N, Hannoun Z, Crook A, Baines A, Korber B, Qiu X, Hanke T. Complete protection of the BALB/c and C57BL/6J mice against Ebola and Marburg virus lethal challenges by pan-filovirus T-cell epigraph vaccine. PLoS Pathog 2019; 15:e1007564. [PMID: 30817809 PMCID: PMC6394903 DOI: 10.1371/journal.ppat.1007564] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/04/2019] [Indexed: 01/31/2023] Open
Abstract
There are a number of vaccine candidates under development against a small number of the most common outbreak filoviruses all employing the virus glycoprotein (GP) as the vaccine immunogen. However, antibodies induced by such GP vaccines are typically autologous and limited to the other members of the same species. In contrast, T-cell vaccines offer a possibility to design a single pan-filovirus vaccine protecting against all known and even likely existing, but as yet unencountered members of the family. Here, we used a cross-filovirus immunogen based on conserved regions of the filovirus nucleoprotein, matrix and polymerase to construct simian adenovirus- and poxvirus MVA-vectored vaccines, and in a proof-of-concept study demonstrated a protection of the BALB/c and C57BL/6J mice against high, lethal challenges with Ebola and Marburg viruses, two distant members of the family, by vaccine-elicited T cells in the absence of GP antibodies.
Collapse
Affiliation(s)
- Md Niaz Rahim
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Edmund G. Wee
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Shihua He
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Jonathan Audet
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Kevin Tierney
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Nathifa Moyo
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Zara Hannoun
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Alison Crook
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrea Baines
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Bette Korber
- Los Alamo National Laboratory, Theoretical Biology and Biophysics, Los Alamos, New Mexico, United States of America
- The New Mexico Consortium, Los Alamos, New Mexico, United States of America
| | - Xiangguo Qiu
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
65
|
Yang X, Wang X, Song Y, Zhou P, Li D, Zhang C, Jin X, Huang Z, Zhou D. Chimpanzee adenoviral vector prime-boost regimen elicits potent immune responses against Ebola virus in mice and rhesus macaques. Emerg Microbes Infect 2019; 8:1086-1097. [PMID: 31339465 PMCID: PMC6711196 DOI: 10.1080/22221751.2019.1644968] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 07/11/2019] [Indexed: 12/19/2022]
Abstract
In the last few decades, Ebola virus (EBOV) has emerged periodically and infected people in Africa, resulting in an extremely high mortality rate. With no available prophylaxis or cure so far, a highly effective Ebola vaccine is urgently needed. In this study, we developed a novel chimpanzee adenovirus-based prime-boost vaccine by exploiting two recombinant replication-deficient chimpanzee adenoviral vectors, AdC7 and AdC68, which express glycoproteins (GP) of the EBOV strain identified in the 2014 outbreak. Our results indicated that a single immunization using AdC7 or AdC68 could stimulate potent EBOV-specific antibody responses, whereas the AdC7 prime-AdC68 boost regimen induced much stronger and sustained humoral and cellular immune responses in both mice and rhesus monkeys, compared with AdC7 or AdC68 single vaccination or the AdC68 prime-AdC7 boost regimen. This prime-boost vaccine could also protect mice from the simulated infection with EBOV-like particle (EBOVLP) in biosafety level 2 (BSL-2) laboratories, and antibodies from the prime-boost immunized rhesus macaques could passively provide protection against EBOVLP infection. Altogether, our results show that the AdC7 prime-AdC68 boost vaccine is a promising candidate for further development to combat EBOV infections.
Collapse
Affiliation(s)
- Xi Yang
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Xiang Wang
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Yufeng Song
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People’s Republic of China
| | - Ping Zhou
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Dapeng Li
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Chao Zhang
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Xia Jin
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Zhong Huang
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
| | - Dongming Zhou
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
- Vaccine Research Center, CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People’s Republic of China
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People’s Republic of China
| |
Collapse
|
66
|
Wang B, Li J, Wu S, Chen Y, Zhang Z, Zhai Y, Guo Q, Zhang J, Song X, Zhao Z, Hou L, Chen W. Seroepidemiological investigation of HAdV-4 infection among healthy adults in China and in Sierra Leone, West Africa. Emerg Microbes Infect 2018; 7:200. [PMID: 30514848 PMCID: PMC6279822 DOI: 10.1038/s41426-018-0206-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/31/2018] [Accepted: 11/11/2018] [Indexed: 12/31/2022]
Abstract
An apparent increase in the frequency of human adenovirus type 4 (HAdV-4) infections among general populations has been observed over the past 10 years. However, available epidemiological data that may reflect previous viral circulation and assist in predicting potential outbreaks are sparse, particularly in mainland China and Africa. In this study, a convenient neutralization assay for use in the surveillance of historical HAdV-4 infections was established based on a recombinant luciferase-expressing virus. Subsequently, the neutralizing antibodies (nAbs) of 1013 healthy adult serum samples from China and Sierra Leone were evaluated. Our results showed that over 50% of the participants from China and nearly 70% of donors from Sierra Leone had detectable nAbs against HAdV-4 despite the few infection cases officially reported in these regions. Furthermore, the prevalence of nAbs to HAdV-4 is lower than that to HAdV-5, and both varied by geographic location. In addition, the seropositive rates of both HAdV-4 and HAdV-5 nAbs increased with age. However, the nAbs stimulated by HAdV-4 remained stable at low (≤200) levels among the different age groups, whereas moderate (201–1000) or high (>1000) nAb levels were produced by HAdV-5 and tended to decrease with age. These results elucidate the human humoral immune response against HAdV-4 and revealed that this virus may be an underestimated causative agent of respiratory disease among adults in China and West Africa, demonstrating the importance of HAdV-4 surveillance and providing useful insights for the future development of HAdV-4-based vaccines.
Collapse
Affiliation(s)
- Busen Wang
- Beijing Institute of Biotechnology, 20 East Street, Beijing, 100071, China
| | - Jianhua Li
- Beijing Institute of Biotechnology, 20 East Street, Beijing, 100071, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, 20 East Street, Beijing, 100071, China
| | - Yi Chen
- Beijing Institute of Biotechnology, 20 East Street, Beijing, 100071, China
| | - Zhe Zhang
- Beijing Institute of Biotechnology, 20 East Street, Beijing, 100071, China
| | - Yanfang Zhai
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, 5 Yiheyuan Road, Beijing, 100871, China
| | - Qiang Guo
- Beijing Institute of Biotechnology, 20 East Street, Beijing, 100071, China
| | - Jinlong Zhang
- Beijing Institute of Biotechnology, 20 East Street, Beijing, 100071, China
| | - Xiaohong Song
- Beijing Institute of Biotechnology, 20 East Street, Beijing, 100071, China
| | - Zhenghao Zhao
- Beijing Institute of Biotechnology, 20 East Street, Beijing, 100071, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, 20 East Street, Beijing, 100071, China.
| | - Wei Chen
- Beijing Institute of Biotechnology, 20 East Street, Beijing, 100071, China.
| |
Collapse
|
67
|
Li Y, Wang L, Zhu T, Wu S, Feng L, Cheng P, Liu J, Wang J. Establishing China's National Standard for the Recombinant Adenovirus Type 5 Vector–Based Ebola Vaccine (Ad5-EBOV) Virus Titer. HUM GENE THER CL DEV 2018; 29:226-232. [PMID: 30381976 DOI: 10.1089/humc.2018.129] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Yuhua Li
- National Institute for Food and Drug Control, Beijing, P.R. China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Ling Wang
- National Institute for Food and Drug Control, Beijing, P.R. China
| | - Tao Zhu
- Tianjin CanSino Biologics, Inc. Tianjin, P.R. China
| | - Shipo Wu
- Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Liqiang Feng
- Guangzhou Institutes of Biomedicine and Heath, Chinese Academy of Sciences, Guangzhou, P.R. China
| | - Ping Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jingjing Liu
- National Institute for Food and Drug Control, Beijing, P.R. China
| | - Junzhi Wang
- National Institute for Food and Drug Control, Beijing, P.R. China
| |
Collapse
|
68
|
Lin CL, Huang HM, Hsieh CL, Fan CK, Lee YL. Jagged1-expressing adenovirus-infected dendritic cells induce expansion of Foxp3 + regulatory T cells and alleviate T helper type 2-mediated allergic asthma in mice. Immunology 2018; 156:199-212. [PMID: 30418664 DOI: 10.1111/imm.13021] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 10/24/2018] [Accepted: 11/02/2018] [Indexed: 12/28/2022] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells that play a key role in directing T-cell responses. Regulatory T (Treg) cells possess an immunosuppressive ability to inhibit effector T-cell responses, and Notch ligand Jagged1 (Jag1) is implicated in Treg cell differentiation. In this study, we evaluated whether bone marrow-derived DCs genetically engineered to express Jag1 (Jag1-DCs) would affect the maturation and function of DCs in vitro and further investigated the immunoregulatory ability of Jag1-DCs to manipulate T helper type 2 (Th2) -mediated allergic asthma in mice. We produced Jag1-DCs by adenoviral transduction. Overexpression of Jag1 by ovalbumin (OVA) -stimulated Jag1-DCs exhibited increased expression of programmed cell death ligand 1 (PD-L1) and OX40L molecules. Subsequently, co-culture of these OVA-pulsed Jag1-DCs with allogeneic or syngeneic CD4+ T cells promoted the generation of Foxp3+ Treg cells, and blocking PD-L1 using specific antibodies partially reduced Treg cell expansion. Furthermore, adoptive transfer of OVA-pulsed Jag1-DCs to mice with OVA-induced asthma reduced allergen-specific immunoglobulin E production, airway hyperresponsiveness, airway inflammation, and secretion of Th2-type cytokines (interleukin-4, interleukin-5, and interleukin-13). Notably, an increased number of Foxp3+ Treg cells associated with enhanced levels of transforming growth factor-β production was observed in Jag1-DC-treated mice. These data indicate that transgenic expression of Jag1 by DCs promotes induction of Foxp3+ Treg cells, which ameliorated Th2-mediated allergic asthma in mice. Our study supports an attractive strategy to artificially generate immunoregulatory DCs and provides a novel approach for manipulating Th2 cell-driven deleterious immune diseases.
Collapse
Affiliation(s)
- Chu-Lun Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Huei-Mei Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Ling Hsieh
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chia-Kwung Fan
- Department of Molecular Parasitology and Tropical Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
69
|
van Winkel CAJ, Moreno A, Curiel DT. Capsid-Incorporation Strategy To Display Antigens for an Alternative Adenoviral Vector Vaccine Approach. Mol Pharm 2018; 15:5446-5453. [PMID: 30359030 DOI: 10.1021/acs.molpharmaceut.8b00591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The adenovirus (Ad) is widely used as a vaccine because of its ability to induce a cellular and humoral immune response. In addition, human clinical trials have validated the safety and efficacy of Ad as a vaccine vector. The traditional approach for employing the adenovirus as vaccine is to configure the antigen genes into the expression cassette of the Ad genome. An alternative method for inducing an immune response is the "capsid-incorporation" strategy. This strategy is based upon the incorporation of proteins or peptides into the capsid proteins. This review will focus on the established uses of this approach as well as highlighting the new developments regarding the capsid-incorporation strategy.
Collapse
Affiliation(s)
- Claudia A J van Winkel
- Cancer Biology Division, Department of Radiation Oncology , Washington University School of Medicine , St. Louis , Missouri 63110 , United States.,Department of Chemical and Pharmaceutical Biology , University of Groningen , Groningen 9700 AB , The Netherlands
| | - Alberto Moreno
- Emory Vaccine Center and Yerkes National Primate Research Center , Emory University , Atlanta , Georgia 30322 , United States.,Division of Infectious Diseases, Department of Medicine , Emory University , Atlanta , Georgia 30322 , United States
| | - David T Curiel
- Cancer Biology Division, Department of Radiation Oncology , Washington University School of Medicine , St. Louis , Missouri 63110 , United States
| |
Collapse
|
70
|
Identification of novel HLA-A11-restricted T-cell epitopes in the Ebola virus nucleoprotein. Microbes Infect 2018; 21:56-62. [PMID: 29775667 DOI: 10.1016/j.micinf.2018.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/20/2018] [Accepted: 04/26/2018] [Indexed: 12/17/2022]
Abstract
The Ebola virus (EBOV) is a very contagious virus that is highly fatal in humans and animals. The largest epidemic was in West Africa in 2014, in which over 11,000 people died. However, to date, there are no licensed vaccines against it. Studies show that CD4+ and CD8+ T-cell responses, especially cytotoxic T-lymphocyte (CTL) responses, play key roles in protecting individuals from EBOV infection. Since HLA-restricted epitope vaccines are likely to be effective and safe immunization strategies for infectious diseases, the present study screened for CTL epitopes in the EBOV-nucleoprotein that are restricted by HLA-A11 (a common allele in Chinese people). Predictive computer analysis of the amino-acid sequence of EBOV-nucleoprotein identified ten putative HLA-A11-restricted epitopes. ELISPOT assay of immunized HLA-A11/DR1 transgenic mice showed that five (GR-9, VR-9, EK-9, PK-9, and RK-9) induced effective CTL responses. Additional epitope analyses will aid the design of epitope vaccines against EBOV.
Collapse
|
71
|
Rauch S, Jasny E, Schmidt KE, Petsch B. New Vaccine Technologies to Combat Outbreak Situations. Front Immunol 2018; 9:1963. [PMID: 30283434 PMCID: PMC6156540 DOI: 10.3389/fimmu.2018.01963] [Citation(s) in RCA: 360] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/09/2018] [Indexed: 01/07/2023] Open
Abstract
Ever since the development of the first vaccine more than 200 years ago, vaccinations have greatly decreased the burden of infectious diseases worldwide, famously leading to the eradication of small pox and allowing the restriction of diseases such as polio, tetanus, diphtheria, and measles. A multitude of research efforts focuses on the improvement of established and the discovery of new vaccines such as the HPV (human papilloma virus) vaccine in 2006. However, radical changes in the density, age distribution and traveling habits of the population worldwide as well as the changing climate favor the emergence of old and new pathogens that bear the risk of becoming pandemic threats. In recent years, the rapid spread of severe infections such as HIV, SARS, Ebola, and Zika have highlighted the dire need for global preparedness for pandemics, which necessitates the extremely rapid development and comprehensive distribution of vaccines against potentially previously unknown pathogens. What is more, the emergence of antibiotic resistant bacteria calls for new approaches to prevent infections. Given these changes, established methods for the identification of new vaccine candidates are no longer sufficient to ensure global protection. Hence, new vaccine technologies able to achieve rapid development as well as large scale production are of pivotal importance. This review will discuss viral vector and nucleic acid-based vaccines (DNA and mRNA vaccines) as new approaches that might be able to tackle these challenges to global health.
Collapse
|
72
|
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
|
73
|
Gross L, Lhomme E, Pasin C, Richert L, Thiebaut R. Ebola vaccine development: Systematic review of pre-clinical and clinical studies, and meta-analysis of determinants of antibody response variability after vaccination. Int J Infect Dis 2018; 74:83-96. [PMID: 29981944 DOI: 10.1016/j.ijid.2018.06.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/20/2018] [Accepted: 06/28/2018] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVES For Ebola vaccine development, antibody response is a major endpoint although its determinants are not well known. We aimed to review Ebola vaccine studies and to assess factors associated with antibody response variability in humans. METHODS We searched PubMed and Scopus for preventive Ebola vaccine studies in humans or non-human primates (NHP), published up to February 2018. For each vaccination group with Ebola Zaire antibody titre measurements after vaccination, data about antibody response and its potential determinants were extracted. A random-effects meta-regression was conducted including human groups with at least 8 individuals. RESULTS We reviewed 49 studies (202 vaccination groups including 74 human groups) with various vaccine platforms and antigen inserts. Mean antibody titre was slightly higher in NHP (3.10, 95% confidence interval [293; 327]) than in humans (2.75 [257; 293]). Vaccine platform (p<0·001) and viral strain used for antibody detection (p<0·001) were associated with antibody response in humans, but adjusted heterogeneity remained at 95%. CONCLUSIONS Various platforms have been evaluated in humans, including Ad26, Ad5, ChimpAd3, DNA, MVA, and VSV. In addition to platforms, viral strain used for antibody detection influences antibody response. However, variability remained mostly unexplained. Therefore, comparison of vaccine immunogenicity needs randomised controlled trials.
Collapse
Affiliation(s)
- Lise Gross
- SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France
| | - Edouard Lhomme
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France; Pôle de Santé Publique, CHU de Bordeaux, Bordeaux, F-33000, France
| | - Chloé Pasin
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France
| | - Laura Richert
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France; Pôle de Santé Publique, CHU de Bordeaux, Bordeaux, F-33000, France
| | - Rodolphe Thiebaut
- INSERM, Bordeaux Population Health Research Centre, UMR 1219, Univ. Bordeaux, ISPED, F-33000, Bordeaux, France; SISTM Team (Statistics in System Biology and Translational Medicine), INRIA Research Centre, Bordeaux, F-33000, France; Vaccine Research Institute (VRI), Créteil, F-94000, France; Pôle de Santé Publique, CHU de Bordeaux, Bordeaux, F-33000, France.
| |
Collapse
|
74
|
Kumar D, Gauthami S, Uma M, Nagalekshmi K, Rao PP, Basu A, Ella KM, Hegde NR. Immunogenicity of a Candidate Ebola Hemorrhagic Fever Vaccine in Mice Based on Controlled In Vitro Expression of Ebolavirus Glycoprotein. Viral Immunol 2018; 31:500-512. [PMID: 30095362 DOI: 10.1089/vim.2017.0122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Ebolavirus (EBOV) is the etiology of Ebola hemorrhagic fever (EHF). A major EHF outbreak in 2014-2015 in West Africa claimed >11,000 lives. A licensed vaccine is not available for EHF, although several vaccines have undergone clinical trials. We developed a human adenovirus (Ad) serotype 5-based candidate EHF vaccine based on controlled expression of the EBOV (Makona strain) glycoprotein (GP) as the immunogen. Two clones, AdGP72 and AdGP75, and a control Ad515 vector, were generated and tested for protein expression in vitro and immunogenicity in mice. Eight groups of mice were immunized with three doses of buffer, Ad515, AdGP72, and AdGP75, by two different dose regimens. Three different antigens (AdGP75-infected Vero E6 cell extract and two baculovirus expressed EBOV GP antigens, namely, GP alone or GP with EBOV VP40) were used to evaluate the immune response. Expression studies indicated that full-length GP was cleaved into its component subunits when expressed in mammalian cells through the Ad vectors. Moreover, in coimmunoprecipitation studies, EBOV GP was found to be associated with VP40 when expressed in baculoviruses. The candidate vaccines were immunogenic in mice, as evaluated by enzyme-linked immunosorbent assay using mammalian- or baculovirus-derived antigens. Further characterization and development of the candidate vaccines are warranted.
Collapse
Affiliation(s)
| | | | | | | | | | - Atanu Basu
- 2 National Institute of Virology , Pune, India
| | | | | |
Collapse
|
75
|
Dhama K, Karthik K, Khandia R, Chakraborty S, Munjal A, Latheef SK, Kumar D, Ramakrishnan MA, Malik YS, Singh R, Malik SVS, Singh RK, Chaicumpa W. Advances in Designing and Developing Vaccines, Drugs, and Therapies to Counter Ebola Virus. Front Immunol 2018; 9:1803. [PMID: 30147687 PMCID: PMC6095993 DOI: 10.3389/fimmu.2018.01803] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 07/23/2018] [Indexed: 01/10/2023] Open
Abstract
Ebola virus (EBOV), a member of the family Filoviridae, is responsible for causing Ebola virus disease (EVD) (formerly named Ebola hemorrhagic fever). This is a severe, often fatal illness with mortality rates varying from 50 to 90% in humans. Although the virus and associated disease has been recognized since 1976, it was only when the recent outbreak of EBOV in 2014-2016 highlighted the danger and global impact of this virus, necessitating the need for coming up with the effective vaccines and drugs to counter its pandemic threat. Albeit no commercial vaccine is available so far against EBOV, a few vaccine candidates are under evaluation and clinical trials to assess their prophylactic efficacy. These include recombinant viral vector (recombinant vesicular stomatitis virus vector, chimpanzee adenovirus type 3-vector, and modified vaccinia Ankara virus), Ebola virus-like particles, virus-like replicon particles, DNA, and plant-based vaccines. Due to improvement in the field of genomics and proteomics, epitope-targeted vaccines have gained top priority. Correspondingly, several therapies have also been developed, including immunoglobulins against specific viral structures small cell-penetrating antibody fragments that target intracellular EBOV proteins. Small interfering RNAs and oligomer-mediated inhibition have also been verified for EVD treatment. Other treatment options include viral entry inhibitors, transfusion of convalescent blood/serum, neutralizing antibodies, and gene expression inhibitors. Repurposed drugs, which have proven safety profiles, can be adapted after high-throughput screening for efficacy and potency for EVD treatment. Herbal and other natural products are also being explored for EVD treatment. Further studies to better understand the pathogenesis and antigenic structures of the virus can help in developing an effective vaccine and identifying appropriate antiviral targets. This review presents the recent advances in designing and developing vaccines, drugs, and therapies to counter the EBOV threat.
Collapse
Affiliation(s)
- Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | - Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, Agartala, India
| | - Ashok Munjal
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | - Shyma K. Latheef
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Deepak Kumar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | | | - Yashpal Singh Malik
- Division of Biological Standardization, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Rajendra Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Satya Veer Singh Malik
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Raj Kumar Singh
- ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine SIriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
76
|
Feng Y, Li C, Hu P, Wang Q, Zheng X, Zhao Y, Shi Y, Yang S, Yi C, Feng Y, Wu C, Qu L, Xu W, Li Y, Sun C, Gao FG, Xia X, Feng L, Chen L. An adenovirus serotype 2-vectored ebolavirus vaccine generates robust antibody and cell-mediated immune responses in mice and rhesus macaques. Emerg Microbes Infect 2018; 7:101. [PMID: 29872043 PMCID: PMC5988821 DOI: 10.1038/s41426-018-0102-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 05/03/2018] [Accepted: 05/06/2018] [Indexed: 01/21/2023]
Abstract
Ebolavirus vaccines based on several adenoviral vectors have been investigated in preclinical studies and clinical trials. The use of adenovirus serotype 2 as a vector for ebolavirus vaccine has not been reported. Herein, we generated rAd2-ZGP, a recombinant replication-incompetent adenovirus serotype 2 expressing codon-optimized Zaire ebolavirus glycoprotein, and evaluated its immunogenicity in mice and rhesus macaques. rAd2-ZGP induced significant antibody and cell-mediated immune responses at 2 weeks after a single immunization. The glycoprotein (GP)-specific immune responses could be further enhanced with a booster immunization. Compared to protein antigens, Zaire ebolavirus GP and Zaire ebolavirus-like particles, rAd2-ZGP could induce stronger cross-reactive antibody and cell-mediated immune responses to heterologous Sudan ebolavirus in mice and rhesus macaques. In rAd2-ZGP-immunized macaques, GP-specific CD8+ T cells could secret IFN-γ and IL-2, indicating a Th1-biased response. In adenovirus serotype 5 seropositive macaques, rAd2-ZGP could induce robust antibody and cell-mediated immune responses, suggesting that the efficacy of rAd2-ZGP is not affected by pre-existing immunity to adenovirus serotype 5. These results demonstrated that rAd2-ZGP can be considered an alternative ebolavirus vaccine for use in adenovirus serotype 5 seropositive subjects or as a sequential booster vaccine after the subjects have been immunized with a recombinant adenovirus serotype 5-based vaccine.
Collapse
Affiliation(s)
- Yupeng Feng
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chufang Li
- The Guangzhou 8th People's Hospital, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510060, China
| | - Peiyu Hu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.,The Guangzhou 8th People's Hospital, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510060, China
| | - Qian Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuehua Zheng
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Yi Shi
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Changhua Yi
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Ying Feng
- The Guangzhou 8th People's Hospital, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510060, China
| | - Chunxiu Wu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Linbing Qu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Wei Xu
- The Guangzhou 8th People's Hospital, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510060, China
| | - Yao Li
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Caijun Sun
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Fu Geroge Gao
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Liqiang Feng
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Ling Chen
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,The Guangzhou 8th People's Hospital, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510060, China.
| |
Collapse
|
77
|
Distinct Immunogenicity and Efficacy of Poxvirus-Based Vaccine Candidates against Ebola Virus Expressing GP and VP40 Proteins. J Virol 2018. [PMID: 29514907 DOI: 10.1128/jvi.00363-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Zaire and Sudan ebolavirus species cause a severe disease in humans and nonhuman primates (NHPs) characterized by a high mortality rate. There are no licensed therapies or vaccines against Ebola virus disease (EVD), and the recent 2013 to 2016 outbreak in West Africa highlighted the need for EVD-specific medical countermeasures. Here, we generated and characterized head-to-head the immunogenicity and efficacy of five vaccine candidates against Zaire ebolavirus (EBOV) and Sudan ebolavirus (SUDV) based on the highly attenuated poxvirus vector modified vaccinia virus Ankara (MVA) expressing either the virus glycoprotein (GP) or GP together with the virus protein 40 (VP40) forming virus-like particles (VLPs). In a human monocytic cell line, the different MVA vectors (termed MVA-EBOVs and MVA-SUDVs) triggered robust innate immune responses, with production of beta interferon (IFN-β), proinflammatory cytokines, and chemokines. Additionally, several innate immune cells, such as dendritic cells, neutrophils, and natural killer cells, were differentially recruited in the peritoneal cavity of mice inoculated with MVA-EBOVs. After immunization of mice with a homologous prime/boost protocol (MVA/MVA), total IgG antibodies against GP or VP40 from Zaire and Sudan ebolavirus were differentially induced by these vectors, which were mainly of the IgG1 and IgG3 isotypes. Remarkably, an MVA-EBOV construct coexpressing GP and VP40 protected chimeric mice challenged with EBOV to a greater extent than a vector expressing GP alone. These results support the consideration of MVA-EBOVs and MVA-SUDVs expressing GP and VP40 and producing VLPs as best-in-class potential vaccine candidates against EBOV and SUDV.IMPORTANCE EBOV and SUDV cause a severe hemorrhagic fever affecting humans and NHPs. Since their discovery in 1976, they have caused several sporadic epidemics, with the recent outbreak in West Africa from 2013 to 2016 being the largest and most severe, with more than 11,000 deaths being reported. Although some vaccines are in advanced clinical phases, less expensive, safer, and more effective licensed vaccines are desirable. We generated and characterized head-to-head the immunogenicity and efficacy of five novel vaccines against EBOV and SUDV based on the poxvirus MVA expressing GP or GP and VP40. The expression of GP and VP40 leads to the formation of VLPs. These MVA-EBOV and MVA-SUDV recombinants triggered robust innate and humoral immune responses in mice. Furthermore, MVA-EBOV recombinants expressing GP and VP40 induced high protection against EBOV in a mouse challenge model. Thus, MVA expressing GP and VP40 and producing VLPs is a promising vaccine candidate against EBOV and SUDV.
Collapse
|
78
|
Wong G, Mendoza EJ, Plummer FA, Gao GF, Kobinger GP, Qiu X. From bench to almost bedside: the long road to a licensed Ebola virus vaccine. Expert Opin Biol Ther 2018; 18:159-173. [PMID: 29148858 PMCID: PMC5841470 DOI: 10.1080/14712598.2018.1404572] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION The Ebola virus (EBOV) disease epidemic during 2014-16 in West Africa has accelerated the clinical development of several vaccine candidates that have demonstrated efficacy in the gold standard nonhuman primate (NHP) model, namely cynomolgus macaques. AREAS COVERED This review discusses the pre-clinical research and if available, clinical evaluation of the currently available EBOV vaccine candidates, while emphasizing the translatability of pre-clinical data generated in the NHP model to clinical data in humans. EXPERT OPINION Despite the existence of many successful EBOV vaccine candidates in the pre-clinical stages, only two platforms became the focus of Phase 2/3 efficacy trials in Liberia, Sierra Leone, and Guinea near the peak of the epidemic: the Vesicular stomatitis virus (VSV)-vectored vaccine and the chimpanzee adenovirus type 3 (ChAd3)-vectored vaccine. The results of three distinct clinical trials involving these candidates may soon pave the way for a licensed, safe and efficacious EBOV vaccine to help combat future epidemics.
Collapse
Affiliation(s)
- Gary Wong
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People’s Hospital, Shenzhen, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, Winnipeg, MB, Canada
| | - Emelissa J. Mendoza
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | | | - George F. Gao
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen Key Laboratory of Pathogen and Immunity, Shenzhen Third People’s Hospital, Shenzhen, China
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, China
| | - Gary P. Kobinger
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
- Département de microbiologie-infectiologie et d’immunologie, Universite Laval, Quebec, QC, Canada
| | - Xiangguo Qiu
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology, Winnipeg, MB, Canada
| |
Collapse
|
79
|
|
80
|
Narayanan N, Lacy CR, Cruz JE, Nahass M, Karp J, Barone JA, Hermes-DeSantis ER. Disaster Preparedness: Biological Threats and Treatment Options. Pharmacotherapy 2018; 38:217-234. [PMID: 29236288 DOI: 10.1002/phar.2068] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Biological disasters can be natural, accidental, or intentional. Biological threats have made a lasting impact on civilization. This review focuses on agents of clinical significance, bioterrorism, and national security, specifically Category A agents (anthrax, botulism, plague, tularemia, and smallpox), as well as briefly discusses other naturally emerging infections of public health significance, Ebola virus (also a Category A agent) and Zika virus. The role of pharmacists in disaster preparedness and disaster response is multifaceted and important. Their expertise includes clinical knowledge, which can aid in drug information consultation, patient-specific treatment decision making, and development of local treatment plans. To fulfill this role, pharmacists must have a comprehensive understanding of medical countermeasures for these significant biological threats across all health care settings. New and reemerging infectious disease threats will continue to challenge the world. Pharmacists will be at the forefront of preparedness and response, sharing knowledge and clinical expertise with responders, official decision makers, and the general public.
Collapse
Affiliation(s)
- Navaneeth Narayanan
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Department of Pharmacy, Robert Wood Johnson University Hospital, New Brunswick, New Jersey.,Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Clifton R Lacy
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey.,Rutgers School of Communication and Information, New Brunswick, New Jersey
| | - Joseph E Cruz
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Department of Pharmacy, Englewood Hospital and Medical Center, Englewood, New Jersey
| | - Meghan Nahass
- Alpert Medical School, Brown University, Providence, Rhode Island
| | - Jonathan Karp
- University of Vermont (at the time of writing), Burlington, Vermont
| | - Joseph A Barone
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Department of Pharmacy, Robert Wood Johnson University Hospital, New Brunswick, New Jersey
| | - Evelyn R Hermes-DeSantis
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Department of Pharmacy, Robert Wood Johnson University Hospital, New Brunswick, New Jersey
| |
Collapse
|
81
|
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: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
82
|
Lambe T, Bowyer G, Ewer KJ. A review of Phase I trials of Ebola virus vaccines: what can we learn from the race to develop novel vaccines? Philos Trans R Soc Lond B Biol Sci 2017; 372:rstb.2016.0295. [PMID: 28396468 PMCID: PMC5394635 DOI: 10.1098/rstb.2016.0295] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2016] [Indexed: 11/23/2022] Open
Abstract
Sporadic outbreaks of Ebola virus infection have been documented since the mid-Seventies and viral exposure can lead to lethal haemorrhagic fever with case fatalities as high as 90%. There is now a comprehensive body of data from both ongoing and completed clinical trials assessing various vaccine strategies, which were rapidly advanced through clinical trials in response to the 2013–2016 Ebola virus disease (EVD) public health emergency. Careful consideration of immunogenicity post vaccination is essential but has been somewhat stifled because of the wide array of immunological assays and outputs that have been used in the numerous clinical trials. We discuss here the different aspects of the immune assays currently used in the Phase I clinical trials for Ebola virus vaccines, and draw comparisons across the immune outputs where possible; various trials have examined both cellular and humoral immunity in European and African cohorts. Assessment of the safety data, the immunological outputs and the ease of field deployment for the various vaccine modalities will help both the scientific community and policy-makers prioritize and potentially license vaccine candidates. If this can be achieved, the next outbreak of Ebola virus, or other emerging pathogen, can be more readily contained and will not have such widespread and devastating consequences. This article is part of the themed issue ‘The 2013–2016 West African Ebola epidemic: data, decision-making and disease control’.
Collapse
Affiliation(s)
- Teresa Lambe
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Georgina Bowyer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
83
|
Wang L, Liu J, Kong Y, Hou L, Li Y. Immunogenicity of Recombinant Adenovirus Type 5 Vector-Based Ebola Vaccine Expressing Glycoprotein from the 2014 Epidemic Strain in Mice. Hum Gene Ther 2017; 29:87-95. [PMID: 28795602 DOI: 10.1089/hum.2017.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The 2014 Ebola outbreak in West Africa highlighted the worldwide public health threat posed by this virus and the urgent need for an Ebola vaccine. A novel recombinant adenovirus type 5 vector-based Ebola vaccine (Ad5-EBOV), based on the 2014 Zaire Guinea epidemic strain, was developed in China. A good safety profile and robust immune response elicited by Ad5-EBOV were confirmed in phase 1 and phase 2 clinical trials. Nonetheless, clinical studies of this Ebola vaccine are still at an early stage and there are still no solid efficacy data for humans. For efficacy evaluation and quality control of Ad5-EBOV, the cellular and humoral immune responses in BALB/c mice vaccinated with Ad5-EBOV were examined at various time points. ELISpot and flow cytometric analysis showed that EBOV glycoprotein (GP)-specific T cell responses were detectable early in the first week after infection and by week 4 had increased to maximum levels, which lasted through week 6. During week 1, high titers of EBOV GP-specific antibodies were found (geometric mean [GM], 1783). These titers peaked at week 10 (GM, 26,214) and lasted to 6 months (GM, 1,351). The titer of neutralizing antibodies based on pseudovirus assays also increased over time to peak at 1:16 in one mouse and 1:8 in nine mice during week 6, before decreasing to zero by week 12. These results suggest that BALB/c mice can be used to evaluate the effectiveness of Ad5-EBOV, and that the cellular immune response and humoral immune response can be used as indicators to evaluate vaccine effectiveness. Rapid determination of such methods and indicators is critical for the evaluation of Ebola vaccine efficacy, and can provide effective quality control for Ad5-EBOV.
Collapse
Affiliation(s)
- Ling Wang
- 1 National Institutes for Food and Drug Control , Beijing, China
| | - Jingjing Liu
- 1 National Institutes for Food and Drug Control , Beijing, China
| | - Yan Kong
- 1 National Institutes for Food and Drug Control , Beijing, China
| | - Lihua Hou
- 2 Beijing Institute of Biotechnology , Beijing, China
| | - Yuhua Li
- 1 National Institutes for Food and Drug Control , Beijing, China
| |
Collapse
|
84
|
Ewer K, Sebastian S, Spencer AJ, Gilbert S, Hill AVS, Lambe T. Chimpanzee adenoviral vectors as vaccines for outbreak pathogens. Hum Vaccin Immunother 2017; 13:3020-3032. [PMID: 29083948 PMCID: PMC5718829 DOI: 10.1080/21645515.2017.1383575] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/15/2017] [Accepted: 09/19/2017] [Indexed: 12/27/2022] Open
Abstract
The 2014-15 Ebola outbreak in West Africa highlighted the potential for large disease outbreaks caused by emerging pathogens and has generated considerable focus on preparedness for future epidemics. Here we discuss drivers, strategies and practical considerations for developing vaccines against outbreak pathogens. Chimpanzee adenoviral (ChAd) vectors have been developed as vaccine candidates for multiple infectious diseases and prostate cancer. ChAd vectors are safe and induce antigen-specific cellular and humoral immunity in all age groups, as well as circumventing the problem of pre-existing immunity encountered with human Ad vectors. For these reasons, such viral vectors provide an attractive platform for stockpiling vaccines for emergency deployment in response to a threatened outbreak of an emerging pathogen. Work is already underway to develop vaccines against a number of other outbreak pathogens and we will also review progress on these approaches here, particularly for Lassa fever, Nipah and MERS.
Collapse
Affiliation(s)
- Katie Ewer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Headington, Oxford, UK
| | - Sarah Sebastian
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Headington, Oxford, UK
| | - Alexandra J. Spencer
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Headington, Oxford, UK
| | - Sarah Gilbert
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Headington, Oxford, UK
| | - Adrian V. S. Hill
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Headington, Oxford, UK
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Headington, Oxford, UK
| |
Collapse
|
85
|
Shi W, Li J, Zhou H, Gao GF. Pathogen genomic surveillance elucidates the origins, transmission and evolution of emerging viral agents in China. SCIENCE CHINA. LIFE SCIENCES 2017; 60:1317-1330. [PMID: 29270793 PMCID: PMC7088571 DOI: 10.1007/s11427-017-9211-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/01/2017] [Indexed: 02/06/2023]
Abstract
In the past twenty years, numerous novel zoonotic viral agents with pandemic potential have emerged in China, such as the severe acute respiratory syndrome (SARS) coronavirus and, more recently, the avian-origin influenza A/H7N9 virus, which have caused outbreaks among humans with high morbidity and mortality. In addition, several emerging and re-emerging viral pathogens have also been imported into China from travelers, e.g. the Middle East respiratory syndrome (MERS) coronavirus and Zika virus (ZIKV). Herein, we review these emerging viral pathogens in China and focus on how surveillance by pathogen genomics has been employed to discover and annotate novel pathogenic agents, identify natural reservoirs, monitor the transmission events and delineate their evolution and adaption to the human host. We also highlight the application of genomic sequencing in the recent Ebola epidemics in Western Africa. In summary, genomic sequencing has become a standard research tool in the field of emerging infectious diseases which has been proven invaluable in containing these viral infections and reducing burden of disease in humans and animals. Genomic surveillance of pathogenic agents will serve as a key epidemiological and research tool in the modern era of precision infectious diseases and in the future studies of virosphere.
Collapse
Affiliation(s)
- Weifeng Shi
- Key Laboratory of Etiology and Epidemiology of Emerging Infectious Diseases in Universities of Shandong (Taishan Medical College), Taishan Medical College, Taian, 271000, China.
| | - Juan Li
- Key Laboratory of Etiology and Epidemiology of Emerging Infectious Diseases in Universities of Shandong (Taishan Medical College), Taishan Medical College, Taian, 271000, China
| | - Hong Zhou
- Key Laboratory of Etiology and Epidemiology of Emerging Infectious Diseases in Universities of Shandong (Taishan Medical College), Taishan Medical College, Taian, 271000, China
| | - George F Gao
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC), Beijing, 102206, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
86
|
Wu L, Zhang Z, Gao H, Li Y, Hou L, Yao H, Wu S, Liu J, Wang L, Zhai Y, Ou H, Lin M, Wu X, Liu J, Lang G, Xin Q, Wu G, Luo L, Liu P, Shentu J, Wu N, Sheng J, Qiu Y, Chen W, Li L. Open-label phase I clinical trial of Ad5-EBOV in Africans in China. Hum Vaccin Immunother 2017; 13. [PMID: 28708962 DOI: 10.1002/smll.201701815] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/31/2017] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND To determine the safety and immunogenicity of a novel recombinant adenovirus type 5 vector based Ebola virus disease vaccine (Ad5-EBOV) in Africans in China. METHODS A phase 1, dose-escalation, open-label trial was conducted. 61 healthy Africans were sequentially enrolled, with 31 participants receiving one shot intramuscular injection and 30 participants receiving a double-shot regimen. Primary and secondary end points related to safety and immunogenicity were assessed within 28 d after vaccination. This study was registered with ClinicalTrials.gov (NCT02401373). RESULTS Ad5-EBOV is well tolerated and no adverse reaction of grade 3 or above was observed. 53 (86.89%) participants reported at least one adverse reaction within 28 d of vaccination. The most common reaction was fever and the mild pain at injection site, and there were no significant difference between these 2 groups. Ebola glycoprotein-specific antibodies appeared in all 61 participants and antibodies titers peaked after 28 d of vaccination. The geometric mean titres (GMTs) were similar between these 2 groups (1919.01 vs 1684.70 P = 0.5562). The glycoprotein-specific T-cell responses rapidly peaked after 14 d of vaccination and then decreased, however, the percentage of subjects with responses were much higher in the high-dose group (60.00% vs 9.68%, P = 0.0014). Pre-existing Ad5 neutralizing antibodies could significantly dampen the specific humoral immune response and cellular response to the vaccine. CONCLUSION The application of Ad5-EBOV demonstrated safe in Africans in China and a specific GP antibody and T-cell response could occur 14 d after the first immunization. This acceptable safety profile provides a reliable basis to proceed with trials in Africa.
Collapse
MESH Headings
- Adult
- Africa/epidemiology
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- China
- Ebola Vaccines/administration & dosage
- Ebola Vaccines/adverse effects
- Ebola Vaccines/immunology
- Ebolavirus/immunology
- Female
- Fever/ethnology
- Healthy Volunteers
- Hemorrhagic Fever, Ebola/epidemiology
- Hemorrhagic Fever, Ebola/ethnology
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/prevention & control
- Humans
- Immunity, Cellular
- Immunity, Humoral
- Immunogenicity, Vaccine
- Injections, Intramuscular
- Male
- Membrane Glycoproteins/immunology
- Middle Aged
- T-Lymphocytes/immunology
- Vaccination
- Young Adult
Collapse
Affiliation(s)
- Lihua Wu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Zhe Zhang
- c Beijing Institute of Biotechnology , Haidian District, Beijing , China
| | - Hainv Gao
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
- d Zhejiang University International Hospital , Xiacheng District, Hangzhou , Zhejiang , China
| | - Yuhua Li
- e National Institutes for Food and Drug Control , Chongwen District, Beijing , China
| | - Lihua Hou
- c Beijing Institute of Biotechnology , Haidian District, Beijing , China
| | - Hangping Yao
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Shipo Wu
- c Beijing Institute of Biotechnology , Haidian District, Beijing , China
| | - Jian Liu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Ling Wang
- e National Institutes for Food and Drug Control , Chongwen District, Beijing , China
| | - You Zhai
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Huilin Ou
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Meihua Lin
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Xiaoxin Wu
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
- d Zhejiang University International Hospital , Xiacheng District, Hangzhou , Zhejiang , China
| | - Jingjing Liu
- e National Institutes for Food and Drug Control , Chongwen District, Beijing , China
| | - Guanjing Lang
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Qian Xin
- f The General Hospital of People's Liberation Army , Beijing , China
| | - Guolan Wu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Li Luo
- g Department of Epidemiology and Biostatistics , School of Public Health, Southeast University , Nanjing , Jiangsu , China
| | - Pei Liu
- g Department of Epidemiology and Biostatistics , School of Public Health, Southeast University , Nanjing , Jiangsu , China
| | - Jianzhong Shentu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Nanping Wu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Jifang Sheng
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Yunqing Qiu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Wei Chen
- c Beijing Institute of Biotechnology , Haidian District, Beijing , China
| | - Lanjuan Li
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
- d Zhejiang University International Hospital , Xiacheng District, Hangzhou , Zhejiang , China
| |
Collapse
|
87
|
Vujadinovic M, Wunderlich K, Callendret B, Koning M, Vermeulen M, Sanders B, van der Helm E, Gecgel A, Spek D, de Boer K, Stalknecht M, Serroyen J, Grazia Pau M, Schuitemaker H, Zahn R, Custers J, Vellinga J. Adenoviral Type 35 and 26 Vectors with a Bidirectional Expression Cassette in the E1 Region Show an Improved Genetic Stability Profile and Potent Transgene-Specific Immune Response. Hum Gene Ther 2017; 29:337-351. [PMID: 28816084 DOI: 10.1089/hum.2017.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Genetic vaccines based on replication-incompetent adenoviral (AdV) vectors are currently in clinical development. Monovalent AdV vectors express one antigen from an expression cassette placed in most cases in the E1 region. For many vaccines, inclusion of several antigens is necessary in order to raise protective immunity and/or target more than one pathogen or pathogen strain. On the basis of the current technology, a mix of several monovalent vectors can be employed. However, a mix of the standard monovalent AdV vectors may not be optimal with respect to manufacturing costs and the final dose per vector in humans. Alternatively, a variety of bivalent recombinant AdV vector approaches is described in the literature. It remains unclear whether all strategies are equally suitable for clinical development while preserving all the beneficial properties of the monovalent AdV (e.g., immunogenic potency). Therefore, a thorough assessment of different bivalent AdV strategies was performed in a head-to-head fashion compared with the monovalent benchmark. The vectors were tested for rescue efficiency, genetic stability, transgene expression, and potency to induce transgene-specific immune responses. We report that the vector expressing multiple antigens from a bidirectional expression cassette in E1 shows a better genetic stability profile and a potent transgene-specific immune response compared with the other tested bivalent vectors.
Collapse
Affiliation(s)
- Marija Vujadinovic
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Kerstin Wunderlich
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Benoit Callendret
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Marina Koning
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Mark Vermeulen
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Barbara Sanders
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Esmeralda van der Helm
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Adile Gecgel
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Dirk Spek
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Karin de Boer
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Masha Stalknecht
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jan Serroyen
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Maria Grazia Pau
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Hanneke Schuitemaker
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Roland Zahn
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jerome Custers
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| | - Jort Vellinga
- Janssen Vaccines and Prevention, Janssen Pharmaceutical Companies of Johnson & Johnson , Leiden, the Netherlands
| |
Collapse
|
88
|
Venkatraman N, Silman D, Folegatti PM, Hill AVS. Vaccines against Ebola virus. Vaccine 2017; 36:5454-5459. [PMID: 28780120 DOI: 10.1016/j.vaccine.2017.07.054] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 06/15/2017] [Accepted: 07/17/2017] [Indexed: 11/29/2022]
Abstract
We have just witnessed the largest and most devastating outbreak of Ebola virus disease, which highlighted the urgent need for development of an efficacious vaccine that could be used to curtail future outbreaks. Prior to 2014, there had been limited impetus worldwide to develop a vaccine since the virus was first discovered in 1976. Though too many lives were lost during this outbreak, it resulted in the significantly accelerated clinical development of a number of candidate vaccines through an extraordinary collaborative global effort coordinated by the World Health Organisation (WHO) and involving a number of companies, trial centres, funders, global stakeholders and agencies. We have acquired substantial safety and immunogenicity data on a number of vaccines in Caucasian and African populations. The rapid pace of events led to the initiation of the landmark efficacy trial testing the rVSV-vectored vaccine, which showed high level efficacy in an outbreak setting when deployed using an innovative ring vaccination strategy. Though the Public Health Emergency of International Concern (PHEIC) declared by the WHO has now been lifted, the global scientific community faces numerous challenges ahead to ensure that there is a licensed, deployable vaccine available for use in future outbreaks for at least the Zaire and Sudan strains of Ebola virus. There remain several unanswered questions on the durability of protection, mechanistic immunological correlates and preferred deployment strategies. This review outlines a brief history of the development of Ebola vaccines, the significant progress made since the scale of the outbreak became apparent, some lessons learnt and how they could shape future development of vaccines and the management of similar outbreaks.
Collapse
Affiliation(s)
- Navin Venkatraman
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom.
| | - Daniel Silman
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Pedro M Folegatti
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Adrian V S Hill
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| |
Collapse
|
89
|
Poland GA, Ovsyannikova IG, Kennedy RB. Personalized vaccinology: A review. Vaccine 2017; 36:5350-5357. [PMID: 28774561 PMCID: PMC5792371 DOI: 10.1016/j.vaccine.2017.07.062] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/19/2017] [Accepted: 07/12/2017] [Indexed: 12/18/2022]
Abstract
At the current time, the field of vaccinology remains empirical in many respects. Vaccine development, vaccine immunogenicity, and vaccine efficacy have, for the most part, historically been driven by an empiric “isolate-inactivate-inject” paradigm. In turn, a population-level public health paradigm of “the same dose for everyone for every disease” model has been the normative thinking in regard to prevention of vaccine-preventable infectious diseases. In addition, up until recently, no vaccines had been designed specifically to overcome the immunosenescence of aging, consistent with a post-WWII mentality of developing vaccines and vaccine programs for children. It is now recognized that the current lack of knowledge concerning how immune responses to vaccines are generated is a critical barrier to understanding poor vaccine responses in the elderly and in immunoimmaturity, discovery of new correlates of vaccine immunogenicity (vaccine response biomarkers), and a directed approach to new vaccine development. The new fields of vaccinomics and adversomics provide models that permit global profiling of the innate, humoral, and cellular immune responses integrated at a systems biology level. This has advanced the science beyond that of reductionist scientific approaches by revealing novel interactions between and within the immune system and other biological systems (beyond transcriptional level), which are critical to developing “downstream” adaptive humoral and cellular responses to infectious pathogens and vaccines. Others have applied systems level approaches to the study of antibody responses (a.k.a. “systems serology”), [1] high-dimensional cell subset immunophenotyping through CyTOF, [2,3] and vaccine induced metabolic changes [4]. In turn, this knowledge is being utilized to better understand the following: identifying who is at risk for which infections; the level of risk that exists regarding poor immunogenicity and/or serious adverse events; and the type or dose of vaccine needed to fully protect an individual. In toto, such approaches allow for a personalized approach to the practice of vaccinology, analogous to the substantial inroads that individualized medicine is playing in other fields of human health and medicine. Herein we briefly review the field of vaccinomics, adversomics, and personalized vaccinology.
Collapse
Affiliation(s)
- G A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA.
| | - I G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA
| | - R B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
90
|
Wu L, Zhang Z, Gao H, Li Y, Hou L, Yao H, Wu S, Liu J, Wang L, Zhai Y, Ou H, Lin M, Wu X, Liu J, Lang G, Xin Q, Wu G, Luo L, Liu P, Shentu J, Wu N, Sheng J, Qiu Y, Chen W, Li L. Open-label phase I clinical trial of Ad5-EBOV in Africans in China. Hum Vaccin Immunother 2017; 13:2078-2085. [PMID: 28708962 DOI: 10.1080/21645515.2017.1342021] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND To determine the safety and immunogenicity of a novel recombinant adenovirus type 5 vector based Ebola virus disease vaccine (Ad5-EBOV) in Africans in China. METHODS A phase 1, dose-escalation, open-label trial was conducted. 61 healthy Africans were sequentially enrolled, with 31 participants receiving one shot intramuscular injection and 30 participants receiving a double-shot regimen. Primary and secondary end points related to safety and immunogenicity were assessed within 28 d after vaccination. This study was registered with ClinicalTrials.gov (NCT02401373). RESULTS Ad5-EBOV is well tolerated and no adverse reaction of grade 3 or above was observed. 53 (86.89%) participants reported at least one adverse reaction within 28 d of vaccination. The most common reaction was fever and the mild pain at injection site, and there were no significant difference between these 2 groups. Ebola glycoprotein-specific antibodies appeared in all 61 participants and antibodies titers peaked after 28 d of vaccination. The geometric mean titres (GMTs) were similar between these 2 groups (1919.01 vs 1684.70 P = 0.5562). The glycoprotein-specific T-cell responses rapidly peaked after 14 d of vaccination and then decreased, however, the percentage of subjects with responses were much higher in the high-dose group (60.00% vs 9.68%, P = 0.0014). Pre-existing Ad5 neutralizing antibodies could significantly dampen the specific humoral immune response and cellular response to the vaccine. CONCLUSION The application of Ad5-EBOV demonstrated safe in Africans in China and a specific GP antibody and T-cell response could occur 14 d after the first immunization. This acceptable safety profile provides a reliable basis to proceed with trials in Africa.
Collapse
Affiliation(s)
- Lihua Wu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Zhe Zhang
- c Beijing Institute of Biotechnology , Haidian District, Beijing , China
| | - Hainv Gao
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China.,d Zhejiang University International Hospital , Xiacheng District, Hangzhou , Zhejiang , China
| | - Yuhua Li
- e National Institutes for Food and Drug Control , Chongwen District, Beijing , China
| | - Lihua Hou
- c Beijing Institute of Biotechnology , Haidian District, Beijing , China
| | - Hangping Yao
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Shipo Wu
- c Beijing Institute of Biotechnology , Haidian District, Beijing , China
| | - Jian Liu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Ling Wang
- e National Institutes for Food and Drug Control , Chongwen District, Beijing , China
| | - You Zhai
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Huilin Ou
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Meihua Lin
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Xiaoxin Wu
- b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China.,d Zhejiang University International Hospital , Xiacheng District, Hangzhou , Zhejiang , China
| | - Jingjing Liu
- e National Institutes for Food and Drug Control , Chongwen District, Beijing , China
| | - Guanjing Lang
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Qian Xin
- f The General Hospital of People's Liberation Army , Beijing , China
| | - Guolan Wu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Li Luo
- g Department of Epidemiology and Biostatistics , School of Public Health, Southeast University , Nanjing , Jiangsu , China
| | - Pei Liu
- g Department of Epidemiology and Biostatistics , School of Public Health, Southeast University , Nanjing , Jiangsu , China
| | - Jianzhong Shentu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Nanping Wu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Jifang Sheng
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Yunqing Qiu
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China
| | - Wei Chen
- c Beijing Institute of Biotechnology , Haidian District, Beijing , China
| | - Lanjuan Li
- a The First Affiliated Hospital, College of Medicine, Zhejiang University , Xiacheng District, Hangzhou , Zhejiang , China.,b The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases , Xiacheng District, Hangzhou , Zhejiang , China.,d Zhejiang University International Hospital , Xiacheng District, Hangzhou , Zhejiang , China
| |
Collapse
|
91
|
Huttner A, Combescure C, Grillet S, Haks MC, Quinten E, Modoux C, Agnandji ST, Brosnahan J, Dayer JA, Harandi AM, Kaiser L, Medaglini D, Monath T, Roux-Lombard P, Kremsner PG, Ottenhoff THM, Siegrist CA. A dose-dependent plasma signature of the safety and immunogenicity of the rVSV-Ebola vaccine in Europe and Africa. Sci Transl Med 2017; 9:9/385/eaaj1701. [PMID: 28404856 DOI: 10.1126/scitranslmed.aaj1701] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/05/2016] [Accepted: 03/15/2017] [Indexed: 12/17/2022]
Abstract
The 2014-2015 Ebola epidemic affected several African countries, claiming more than 11,000 lives and leaving thousands with ongoing sequelae. Safe and effective vaccines could prevent or limit future outbreaks. The recombinant vesicular stomatitis virus-vectored Zaire Ebola (rVSV-ZEBOV) vaccine has shown marked immunogenicity and efficacy in humans but is reactogenic at higher doses. To understand its effects, we examined plasma samples from 115 healthy volunteers from Geneva who received low-dose (LD) or high-dose (HD) vaccine or placebo. Fifteen plasma chemokines/cytokines were assessed at baseline and on days 1, 2 to 3, and 7 after injection. Significant increases in monocyte-mediated MCP-1/CCL2, MIP-1β/CCL4, IL-6, TNF-α, IL-1Ra, and IL-10 occurred on day 1. A signature explaining 68% of cytokine/chemokine vaccine-response variability was identified. Its score was higher in HD versus LD vaccinees and was associated positively with vaccine viremia and negatively with cytopenia. It was higher in vaccinees with injection-site pain, fever, myalgia, chills, and headache; higher scores reflected increasing severity. In contrast, HD vaccinees who subsequently developed arthritis had lower day 1 scores than other HD vaccinees. Vaccine dose did not influence the signature despite its influence on specific outcomes. The Geneva-derived signature associated strongly (ρ = 0.97) with that of a cohort of 75 vaccinees from a parallel trial in Lambaréné, Gabon. Its score in Geneva HD vaccinees with subsequent arthritis was significantly lower than that in Lambaréné HD vaccinees, none of whom experienced arthritis. This signature, which reveals monocytes' critical role in rVSV-ZEBOV immunogenicity and safety across doses and continents, should prove useful in assessments of other vaccines.
Collapse
Affiliation(s)
- Angela Huttner
- Infection Control Program, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland.,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
| | - Christophe Combescure
- Division of Clinical Epidemiology, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Stéphane Grillet
- World Health Organization Collaborating Center for Vaccine Immunology, Faculty of Medicine, Geneva, Switzerland
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Edwin Quinten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Christine Modoux
- Division of Immunology and Allergy, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Selidji Todagbe Agnandji
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, and German Center for Infection Research, Tübingen, Germany
| | | | - Julie-Anne Dayer
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Ali M Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Laurent Kaiser
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy.,Sclavo Vaccines Association, Siena, Italy
| | - Tom Monath
- NewLink Genetics Corp., 94 Jackson Road, Devens, MA 01439, USA
| | | | - Pascale Roux-Lombard
- Division of Immunology and Allergy, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Peter G Kremsner
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon.,Institut für Tropenmedizin, Universitätsklinikum Tübingen, and German Center for Infection Research, Tübingen, Germany
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Claire-Anne Siegrist
- Center for Vaccinology, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland. .,World Health Organization Collaborating Center for Vaccine Immunology, Faculty of Medicine, Geneva, Switzerland
| |
Collapse
|
92
|
Lee CS, Bishop ES, Zhang R, Yu X, Farina EM, Yan S, Zhao C, Zeng Z, Shu Y, Wu X, Lei J, Li Y, Zhang W, Yang C, Wu K, Wu Y, Ho S, Athiviraham A, Lee MJ, Wolf JM, Reid RR, He TC. Adenovirus-Mediated Gene Delivery: Potential Applications for Gene and Cell-Based Therapies in the New Era of Personalized Medicine. Genes Dis 2017; 4:43-63. [PMID: 28944281 PMCID: PMC5609467 DOI: 10.1016/j.gendis.2017.04.001] [Citation(s) in RCA: 411] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 04/19/2017] [Indexed: 12/12/2022] Open
Abstract
With rapid advances in understanding molecular pathogenesis of human diseases in the era of genome sciences and systems biology, it is anticipated that increasing numbers of therapeutic genes or targets will become available for targeted therapies. Despite numerous setbacks, efficacious gene and/or cell-based therapies still hold the great promise to revolutionize the clinical management of human diseases. It is wildly recognized that poor gene delivery is the limiting factor for most in vivo gene therapies. There has been a long-lasting interest in using viral vectors, especially adenoviral vectors, to deliver therapeutic genes for the past two decades. Among all currently available viral vectors, adenovirus is the most efficient gene delivery system in a broad range of cell and tissue types. The applications of adenoviral vectors in gene delivery have greatly increased in number and efficiency since their initial development. In fact, among over 2,000 gene therapy clinical trials approved worldwide since 1989, a significant portion of the trials have utilized adenoviral vectors. This review aims to provide a comprehensive overview on the characteristics of adenoviral vectors, including adenoviral biology, approaches to engineering adenoviral vectors, and their applications in clinical and pre-clinical studies with an emphasis in the areas of cancer treatment, vaccination and regenerative medicine. Current challenges and future directions regarding the use of adenoviral vectors are also discussed. It is expected that the continued improvements in adenoviral vectors should provide great opportunities for cell and gene therapies to live up to its enormous potential in personalized medicine.
Collapse
Affiliation(s)
- Cody S. Lee
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Elliot S. Bishop
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Ruyi Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Xinyi Yu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Evan M. Farina
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Shujuan Yan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Chen Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Yi Shu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Xingye Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Jiayan Lei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Yasha Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Wenwen Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Laboratory Medicine and Clinical Diagnostics, The Affiliated Yantai Hospital, Binzhou Medical University, Yantai 264100, China
| | - Chao Yang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Ke Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Ying Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Immunology and Microbiology, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Sherwin Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R. Reid
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
93
|
Li JX, Hou LH, Meng FY, Wu SP, Hu YM, Liang Q, Chu K, Zhang Z, Xu JJ, Tang R, Wang WJ, Liu P, Hu JL, Luo L, Jiang R, Zhu FC, Chen W. Immunity duration of a recombinant adenovirus type-5 vector-based Ebola vaccine and a homologous prime-boost immunisation in healthy adults in China: final report of a randomised, double-blind, placebo-controlled, phase 1 trial. LANCET GLOBAL HEALTH 2017; 5:e324-e334. [DOI: 10.1016/s2214-109x(16)30367-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 11/24/2016] [Accepted: 11/30/2016] [Indexed: 12/21/2022]
|
94
|
Huang Y. China's Response to the 2014 Ebola Outbreak in West Africa. GLOBAL CHALLENGES (HOBOKEN, NJ) 2017; 1:1600001. [PMID: 31565261 PMCID: PMC6607196 DOI: 10.1002/gch2.201600001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/01/2016] [Indexed: 05/15/2023]
Abstract
Beginning in March 2014, West Africa has endured the largest outbreak of Ebola viral disease (EVD) in history. The crisis highlighted the role of China in addressing public health emergencies of international concern (PHEIC). Through bilateral and multilateral channels, China kicked off its largest ever humanitarian mission in addressing a PHEIC. The unprecedented generosity served the domestic needs to prevent EVD from spreading into China, but it was also consistent with China's foreign policy objective to pursue soft power in Africa. While its total funding to EVD control in West Africa was no match of top donors like the United States, it becomes much more impressive when adjusted for gross domestic product (GDP) per capita. As Beijing becomes more sensitive to disease outbreaks overseas and as the scope of its humanitarian engagement grows and diversifies, the space for China's cooperation with international actors over global health governance is expected to further expand.
Collapse
Affiliation(s)
- Yanzhong Huang
- Center for Global Health Studies, School of Diplomacy and International RelationsSeton Hall UniversitySouth OrangeNJ07079USA
| |
Collapse
|
95
|
Dai Q, Liang Q, Hu Y, Meng F, Li J, Hou L, Zhou H, Chu K, Hu X, Tang R, Wang W, Hu J, Huang H, Li Z, Yang S, Zhu F. The early-onset febrile reaction following vaccination and associated factors: An exploratory sub-study based on the Ebola vaccine clinical trial. Hum Vaccin Immunother 2017; 13:1-6. [PMID: 28362208 DOI: 10.1080/21645515.2017.1288328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
A phase-1 clinical trial aimed to assess the safety and immunogenicity of the type-5 adenovirus vector based Ebola vaccine (Ad5-EBOV) was conducted in China. To provide more evidence for the safety evaluation and dose-selection, an exploratory sub-study using a wireless automatic temperature measuring platform was done based on the phase-1 clinical trial. The main aim of the sub-study was to obtain more information about the occurrence of fever and detect the potential associated factors, second was to assess the feasibility of the temperature measuring platform in vaccine clinical trials. Temperature data of 3 treatment groups all presented a rising tendency during the first 6 hours after vaccination, the incidence of elevated temperature and possible associated factors were analyzed. For the incidence of elevated temperature, no marked dose-response relationship was found in 6 hours with wireless thermometers; the information from mercury thermometers showed that the grade-1 fever proportion peaked at 6 hours and there was no difference between groups, while grade-2 fever proportion peaked at 24 hours and was significantly higher in high-dose group than those in the other 2 groups. Significant differences were found between sex groups (males vs. female, incidence rate ratios (IRR) = 2.93 and 7.62 for any-grade, grade-2 fever respectively, P<0.001); a decline in grade-2 fever incidence was found with the increasing age groups (IRR = 0.78, P = 0.003) and body mass index (BMI, IRR = 0.67, P<0.001) .Our findings show that the dose-dependent manner between fever and the dose of Ad5-EBOV in this study might emerge after 6 hours, and which is slight and transient. Wireless thermometers secured on the skin surface are not suitable for a long time (longer than 6 hours) measurement, new methods for temperature monitoring, like ear temperature measurement, should be tested in the further research.
Collapse
Affiliation(s)
- Qigang Dai
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Qi Liang
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Yuemei Hu
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Fanyue Meng
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Jingxin Li
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Lihua Hou
- b Beijing Institute of Biotechnology , Beijing , China
| | | | - Kai Chu
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Xiaokui Hu
- d Taixing Center for Disease Control and Prevention , Taizhou , China
| | - Rong Tang
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Wenjuan Wang
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Jialei Hu
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Haodi Huang
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Zhen Li
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Shuqi Yang
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| | - Fengcai Zhu
- a Jiangsu Provincial Center for Disease Control and Prevention , Nanjing , China
| |
Collapse
|
96
|
Grobusch MP, Goorhuis A. Safety and immunogenicity of a recombinant adenovirus vector-based Ebola vaccine. Lancet 2017; 389:578-580. [PMID: 28017405 DOI: 10.1016/s0140-6736(16)32619-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 12/13/2016] [Indexed: 10/20/2022]
Affiliation(s)
- Martin P Grobusch
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, University of Amsterdam, 1100 DE Amsterdam, The Netherlands; Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany; Lion Heart Medical Research Unit, Yele, Sierra Leone.
| | - Abraham Goorhuis
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Academic Medical Center, University of Amsterdam, 1100 DE Amsterdam, The Netherlands; Lion Heart Medical Research Unit, Yele, Sierra Leone
| |
Collapse
|
97
|
Zhu FC, Wurie AH, Hou LH, Liang Q, Li YH, Russell JBW, Wu SP, Li JX, Hu YM, Guo Q, Xu WB, Wurie AR, Wang WJ, Zhang Z, Yin WJ, Ghazzawi M, Zhang X, Duan L, Wang JZ, Chen W. Safety and immunogenicity of a recombinant adenovirus type-5 vector-based Ebola vaccine in healthy adults in Sierra Leone: a single-centre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2017; 389:621-628. [PMID: 28017399 DOI: 10.1016/s0140-6736(16)32617-4] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/19/2016] [Accepted: 11/15/2016] [Indexed: 11/18/2022]
Abstract
BACKGROUND A recombinant adenovirus type-5 vector-based vaccine expressing the glycoprotein of Ebola Zaire Makona variant showed good safety and immunogenicity in a phase 1 trial of healthy Chinese adults. We aimed to assess the safety and immunogenicity of this vaccine in healthy adults in Sierra Leone and to determine the optimal dose. METHODS We did a single-centre, randomised, double-blind, placebo-controlled, phase 2 clinical trial at Sierra Leone-China Friendship Hospital, Freetown, Sierra Leone. We recruited healthy adults aged 18-50 years who were HIV negative, had no history of Ebola virus infection, and had no previous immunisation with other Ebola vaccine candidates. Participants were sequentially enrolled and randomly assigned (2:1:1), by computer-generated block randomisation (block size of eight), to receive the high-dose vaccine (1·6 × 1011 viral particles), low-dose vaccine (8·0 × 1010 viral particles), or placebo (containing only vaccine excipients, with no viral particles). Participants, investigators, and study staff (except two study pharmacists) were masked from treatment allocation. The primary safety outcome was occurrence of solicited adverse reactions within 7 days of vaccination, analysed by intention to treat. The primary immunogenicity outcome was glycoprotein-specific antibody responses at days 14, 28, and 168 after vaccination, analysed in all vaccinated participants who had blood samples drawn for antibody tests. The trial is registered with the Pan African Clinical Trials Registry, number PACTR201509001259869, and is completed. FINDINGS During Oct 10-28, 2015, 500 participants were enrolled and randomly assigned to receive the high-dose vaccine (n=250), low-dose vaccine (n=125), or placebo (n=125). 132 (53%) participants in the high-dose group, 60 (48%) in the low-dose group, and 54 (43%) in the placebo group reported at least one solicited adverse reaction within 7 days of vaccination. Most adverse reactions were mild and self-limiting. Solicited injection-site adverse reactions were significantly more frequent in vaccine recipients (65 [26%] in high-dose group and 31 [25%] in low-dose group) than in those receiving placebo (17 [14%]; p=0·0169). Glycoprotein-specific antibody responses were detected from day 14 onwards (geometric mean titre 1251·0 [95% CI 976·6-1602·5] in low-dose group and 1728·4 [1459·4-2047·0] in high-dose group) and peaked at day 28 (1471·8 [1151·0-1881·8] and 2043·1 [1762·4-2368·4]), but declined quickly in the following months (223·3 [148·2-336·4] and 254·2 [185·0-349·5] at day 168). Geometric mean titres in the placebo group remained around 6·0-6·8 throughout the study period. Three serious adverse events (malaria, gastroenteritis, and one fatal asthma episode) were reported in the high-dose vaccine group, but none was deemed related to the vaccine. INTERPRETATION The recombinant adenovirus type-5 vector-based Ebola vaccine was safe and highly immunogenic in healthy Sierra Leonean adults, and 8·0 × 1010 viral particles was the optimal dose. FUNDING Chinese Ministry of Science and Technology and the National Health and Family Planning Commission, Beijing Institute of Biotechnology, and Tianjin CanSino Biotechnology.
Collapse
Affiliation(s)
- Feng-Cai Zhu
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Alie H Wurie
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Li-Hua Hou
- Beijing Institute of Biotechnology, Beijing, China
| | - Qi Liang
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Yu-Hua Li
- National Institute for Food and Drug Control, Beijing, China
| | | | - Shi-Po Wu
- Beijing Institute of Biotechnology, Beijing, China
| | - Jing-Xin Li
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Yue-Mei Hu
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Qiang Guo
- Beijing Institute of Biotechnology, Beijing, China
| | - Wen-Bo Xu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Abdul R Wurie
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Wen-Juan Wang
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Zhe Zhang
- Beijing Institute of Biotechnology, Beijing, China
| | - Wen-Jiao Yin
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Manal Ghazzawi
- Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Xu Zhang
- Tianjin CanSino Biotechnology, Tianjin, China
| | - Lei Duan
- Tianjin CanSino Biotechnology, Tianjin, China
| | - Jun-Zhi Wang
- National Institute for Food and Drug Control, Beijing, China.
| | - Wei Chen
- Beijing Institute of Biotechnology, Beijing, China.
| |
Collapse
|
98
|
Dolzhikova IV, Zubkova OV, Tukhvatulin AI, Dzharullaeva AS, Tukhvatulina NM, Shcheblyakov DV, Shmarov MM, Tokarskaya EA, Simakova YV, Egorova DA, Scherbinin DN, Tutykhina IL, Lysenko AA, Kostarnoy AV, Gancheva PG, Ozharovskaya TA, Belugin BV, Kolobukhina LV, Pantyukhov VB, Syromyatnikova SI, Shatokhina IV, Sizikova TV, Rumyantseva IG, Andrus AF, Boyarskaya NV, Voytyuk AN, Babira VF, Volchikhina SV, Kutaev DA, Bel'skih AN, Zhdanov KV, Zakharenko SM, Borisevich SV, Logunov DY, Naroditsky BS, Gintsburg AL. Safety and immunogenicity of GamEvac-Combi, a heterologous VSV- and Ad5-vectored Ebola vaccine: An open phase I/II trial in healthy adults in Russia. Hum Vaccin Immunother 2017; 13:613-620. [PMID: 28152326 PMCID: PMC5360131 DOI: 10.1080/21645515.2016.1238535] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Ebola hemorrhagic fever, also known as Ebola virus disease or EVD, is one of the most dangerous viral diseases in humans and animals. In this open-label, dose-escalation clinical trial, we assessed the safety, side effects, and immunogenicity of a novel, heterologous prime-boost vaccine against Ebola, which was administered in 2 doses to 84 healthy adults of both sexes between 18 and 55 years. The vaccine consists of live-attenuated recombinant vesicular stomatitis virus (VSV) and adenovirus serotype-5 (Ad5) expressing Ebola envelope glycoprotein. The most common adverse event was pain at the injection site, although no serious adverse events were reported. The vaccine did not significantly impact blood, urine, and immune indices. Seroconversion rate was 100 %. Antigen-specific IgG geometric mean titer at day 42 was 3,277 (95 % confidence interval 2,401–4,473) in volunteers immunized at full dose. Neutralizing antibodies were detected in 93.1 % of volunteers immunized at full dose, with geometric mean titer 20. Antigen-specific response in peripheral blood mononuclear cells was also detected in 100 % of participants, as well as in CD4+ and CD8+ T cells in 82.8 % and 58.6 % of participants vaccinated at full dose, respectively. The data indicate that the vaccine is safe and induces strong humoral and cellular immune response in up to 100 % of healthy adult volunteers, and provide a rationale for testing efficacy in Phase III trials. Indeed, the strong immune response to the vaccine may elicit long-term protection. This trial was registered with grls.rosminzdrav.ru (No. 495*), and with zakupki.gov.ru (No. 0373100043215000055).
Collapse
Affiliation(s)
- I V Dolzhikova
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - O V Zubkova
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - A I Tukhvatulin
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - A S Dzharullaeva
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - N M Tukhvatulina
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - D V Shcheblyakov
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - M M Shmarov
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - E A Tokarskaya
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - Y V Simakova
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - D A Egorova
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - D N Scherbinin
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - I L Tutykhina
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - A A Lysenko
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - A V Kostarnoy
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - P G Gancheva
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - T A Ozharovskaya
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - B V Belugin
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - L V Kolobukhina
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - V B Pantyukhov
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - S I Syromyatnikova
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - I V Shatokhina
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - T V Sizikova
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - I G Rumyantseva
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - A F Andrus
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - N V Boyarskaya
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - A N Voytyuk
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - V F Babira
- d No. 7 Main Military Clinical Hospital named after academician N. N. Burdenko , Ministry of Defense , Posad-7, Russia
| | - S V Volchikhina
- d No. 7 Main Military Clinical Hospital named after academician N. N. Burdenko , Ministry of Defense , Posad-7, Russia
| | - D A Kutaev
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - A N Bel'skih
- b Military Medical Academy named after S. M. Kirov , Ministry of Defense , Saint Petersburg , Russia
| | - K V Zhdanov
- b Military Medical Academy named after S. M. Kirov , Ministry of Defense , Saint Petersburg , Russia
| | - S M Zakharenko
- b Military Medical Academy named after S. M. Kirov , Ministry of Defense , Saint Petersburg , Russia
| | - S V Borisevich
- c 48 Central Research Institute , Ministry of Defense , Sergiev Posad-6, Russia
| | - D Y Logunov
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - B S Naroditsky
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| | - A L Gintsburg
- a Federal Research Centre of Epidemiology and Microbiology named after Honorary Academician N. F. Gamaleya , Ministry of Health , Moscow, Russia
| |
Collapse
|
99
|
Rojek A, Carson G, Kato Y, Horby PW, Leblebicioglu H. Viral Hemorrhagic Fevers. Infect Dis (Lond) 2017. [DOI: 10.1016/b978-0-7020-6285-8.00132-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
100
|
Higgs ES, Dubey SA, Coller BAG, Simon JK, Bollinger L, Sorenson RA, Wilson B, Nason MC, Hensley LE. Accelerating Vaccine Development During the 2013-2016 West African Ebola Virus Disease Outbreak. Curr Top Microbiol Immunol 2017; 411:229-261. [PMID: 28918539 DOI: 10.1007/82_2017_53] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Ebola virus disease outbreak that began in Western Africa in December 2013 was unprecedented in both scope and spread, and the global response was slower and less coherent than was optimal given the scale and pace of the epidemic. Past experience with limited localized outbreaks, lack of licensed medical countermeasures, reluctance by first responders to direct scarce resources to clinical research, community resistance to outside interventions, and lack of local infrastructure were among the factors delaying clinical research during the outbreak. Despite these hurdles, the global health community succeeded in accelerating Ebola virus vaccine development, in a 5-month interval initiating phase I trials in humans in September 2014 and initiating phase II/III trails in February 2015. Each of the three Ebola virus disease-affected countries, Sierra Leone, Guinea, and Liberia, conducted a phase II/III Ebola virus vaccine trial. Only one of these trials evaluating recombinant vesicular stomatitis virus expressing Ebola virus glycoprotein demonstrated vaccine efficacy using an innovative mobile ring vaccination trial design based on a ring vaccination strategy responsible for eradicating smallpox that reached areas of new outbreaks. Thoughtful and intensive community engagement in each country enabled the critical community partnership and acceptance of the phase II/III in each country. Due to the delayed clinical trial initiation, relative to the epidemiologic peak of the outbreak in the three countries, vaccine interventions may or may not have played a major role in bringing the epidemic under control. Having demonstrated that clinical trials can be performed during a large outbreak, the global research community can now build on the experience to implement trials more rapidly and efficiently in future outbreaks. Incorporating clinical research needs into planning for future health emergencies and understanding what kind of trial designs is needed for reliable results in an epidemic of limited duration should improve global response to future infectious disease outbreaks.
Collapse
Affiliation(s)
- Elizabeth S Higgs
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | | | | | | | - Laura Bollinger
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Robert A Sorenson
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Martha C Nason
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lisa E Hensley
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| |
Collapse
|