1
|
O’Donnell KL, Henderson CW, Anhalt H, Fusco J, Erasmus JH, Lambe T, Marzi A. Vaccine Platform Comparison: Protective Efficacy against Lethal Marburg Virus Challenge in the Hamster Model. Int J Mol Sci 2024; 25:8516. [PMID: 39126087 PMCID: PMC11313278 DOI: 10.3390/ijms25158516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/31/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024] Open
Abstract
Marburg virus (MARV), a filovirus, was first identified in 1967 in Marburg, Germany, and Belgrade, former Yugoslavia. Since then, MARV has caused sporadic outbreaks of human disease with high case fatality rates in parts of Africa, with the largest outbreak occurring in 2004/05 in Angola. From 2021 to 2023, MARV outbreaks occurred in Guinea, Ghana, New Guinea, and Tanzania, emphasizing the expansion of its endemic area into new geographical regions. There are currently no approved vaccines or therapeutics targeting MARV, but several vaccine candidates have shown promise in preclinical studies. We compared three vaccine platforms simultaneously by vaccinating hamsters with either a single dose of an adenovirus-based (ChAdOx-1 MARV) vaccine, an alphavirus replicon-based RNA (LION-MARV) vaccine, or a recombinant vesicular stomatitis virus-based (VSV-MARV) vaccine, all expressing the MARV glycoprotein as the antigen. Lethal challenge with hamster-adapted MARV 4 weeks after vaccination resulted in uniform protection of the VSV-MARV and LION-MARV groups and 83% of the ChAdOx-1 MARV group. Assessment of the antigen-specific humoral response and its functionality revealed vaccine-platform-dependent differences, particularly in the Fc effector functions.
Collapse
Affiliation(s)
- Kyle L. O’Donnell
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Corey W. Henderson
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Hanna Anhalt
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| | - Joan Fusco
- Public Health Vaccines Inc., Cambridge, MA 02412, USA
| | | | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 7BN, UK
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT 59840, USA
| |
Collapse
|
2
|
Bissett C, Belij-Rammerstorfer S, Ulaszewska M, Smith H, Kailath R, Morris S, Powers C, Sebastian S, Sharpe HR, Allen ER, Wang Z, Cunliffe RF, Sallah HJ, Spencer AJ, Gilbert S, Tregoning JS, Lambe T. Systemic prime mucosal boost significantly increases protective efficacy of bivalent RSV influenza viral vectored vaccine. NPJ Vaccines 2024; 9:118. [PMID: 38926455 PMCID: PMC11208422 DOI: 10.1038/s41541-024-00912-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Although licensed vaccines against influenza virus have been successful in reducing pathogen-mediated disease, they have been less effective at preventing viral infection of the airways and current seasonal updates to influenza vaccines do not always successfully accommodate viral drift. Most licensed influenza and recently licensed RSV vaccines are administered via the intramuscular route. Alternative immunisation strategies, such as intranasal vaccinations, and "prime-pull" regimens, may deliver a more sterilising form of protection against respiratory viruses. A bivalent ChAdOx1-based vaccine (ChAdOx1-NP + M1-RSVF) encoding conserved nucleoprotein and matrix 1 proteins from influenza A virus and a modified pre-fusion stabilised RSV A F protein, was designed, developed and tested in preclinical animal models. The aim was to induce broad, cross-protective tissue-resident T cells against heterotypic influenza viruses and neutralising antibodies against RSV in the respiratory mucosa and systemically. When administered via an intramuscular prime-intranasal boost (IM-IN) regimen in mice, superior protection was generated against challenge with either RSV A, Influenza A H3N2 or H1N1. These results support further clinical development of a pan influenza & RSV vaccine administered in a prime-pull regimen.
Collapse
Affiliation(s)
- Cameron Bissett
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | | | - Marta Ulaszewska
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Holly Smith
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Reshma Kailath
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Susan Morris
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Claire Powers
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah Sebastian
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Hannah R Sharpe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Elizabeth R Allen
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ziyin Wang
- Department of Infectious Disease, Imperial College London, London, UK
| | - Robert F Cunliffe
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Alexandra J Spencer
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Sarah Gilbert
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| |
Collapse
|
3
|
Neckermann P, Mohr M, Billmeier M, Karlas A, Boilesen DR, Thirion C, Holst PJ, Jordan I, Sandig V, Asbach B, Wagner R. Transgene expression knock-down in recombinant Modified Vaccinia virus Ankara vectors improves genetic stability and sustained transgene maintenance across multiple passages. Front Immunol 2024; 15:1338492. [PMID: 38380318 PMCID: PMC10877035 DOI: 10.3389/fimmu.2024.1338492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/19/2024] [Indexed: 02/22/2024] Open
Abstract
Modified vaccinia virus Ankara is a versatile vaccine vector, well suited for transgene delivery, with an excellent safety profile. However, certain transgenes render recombinant MVA (rMVA) genetically unstable, leading to the accumulation of mutated rMVA with impaired transgene expression. This represents a major challenge for upscaling and manufacturing of rMVA vaccines. To prevent transgene-mediated negative selection, the continuous avian cell line AGE1.CR pIX (CR pIX) was modified to suppress transgene expression during rMVA generation and amplification. This was achieved by constitutively expressing a tetracycline repressor (TetR) together with a rat-derived shRNA in engineered CR pIX PRO suppressor cells targeting an operator element (tetO) and 3' untranslated sequence motif on a chimeric poxviral promoter and the transgene mRNA, respectively. This cell line was instrumental in generating two rMVA (isolate CR19) expressing a Macaca fascicularis papillomavirus type 3 (MfPV3) E1E2E6E7 artificially-fused polyprotein following recombination-mediated integration of the coding sequences into the DelIII (CR19 M-DelIII) or TK locus (CR19 M-TK), respectively. Characterization of rMVA on parental CR pIX or engineered CR pIX PRO suppressor cells revealed enhanced replication kinetics, higher virus titers and a focus morphology equaling wild-type MVA, when transgene expression was suppressed. Serially passaging both rMVA ten times on parental CR pIX cells and tracking E1E2E6E7 expression by flow cytometry revealed a rapid loss of transgene product after only few passages. PCR analysis and next-generation sequencing demonstrated that rMVA accumulated mutations within the E1E2E6E7 open reading frame (CR19 M-TK) or deletions of the whole transgene cassette (CR19 M-DelIII). In contrast, CR pIX PRO suppressor cells preserved robust transgene expression for up to 10 passages, however, rMVAs were more stable when E1E2E6E7 was integrated into the TK as compared to the DelIII locus. In conclusion, sustained knock-down of transgene expression in CR pIX PRO suppressor cells facilitates the generation, propagation and large-scale manufacturing of rMVA with transgenes hampering viral replication.
Collapse
Affiliation(s)
- Patrick Neckermann
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Madlen Mohr
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Martina Billmeier
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | | | - Ditte R. Boilesen
- Department of Immunology and Microbiology, Center for Medical Parasitology, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
- InProTher APS, Copenhagen, Denmark
| | | | - Peter J. Holst
- Department of Immunology and Microbiology, Center for Medical Parasitology, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
- InProTher APS, Copenhagen, Denmark
| | | | | | - Benedikt Asbach
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
| | - Ralf Wagner
- Institute of Medical Microbiology and Hygiene, Molecular Microbiology (Virology), University of Regensburg, Regensburg, Germany
- Institue of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
4
|
Ni T, Mendonça L, Zhu Y, Howe A, Radecke J, Shah PM, Sheng Y, Krebs AS, Duyvesteyn HM, Allen E, Lambe T, Bisset C, Spencer A, Morris S, Stuart DI, Gilbert S, Zhang P. ChAdOx1 COVID vaccines express RBD open prefusion SARS-CoV-2 spikes on the cell surface. iScience 2023; 26:107882. [PMID: 37766989 PMCID: PMC10520439 DOI: 10.1016/j.isci.2023.107882] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/18/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been proven to be an effective means of decreasing COVID-19 mortality, hospitalization rates, and transmission. One of the vaccines deployed worldwide is ChAdOx1 nCoV-19, which uses an adenovirus vector to drive the expression of the original SARS-CoV-2 spike on the surface of transduced cells. Using cryo-electron tomography and subtomogram averaging, we determined the native structures of the vaccine product expressed on cell surfaces in situ. We show that ChAdOx1-vectored vaccines expressing the Beta SARS-CoV-2 variant produce abundant native prefusion spikes predominantly in one-RBD-up conformation. Furthermore, the ChAdOx1-vectored HexaPro-stabilized spike yields higher cell surface expression, enhanced RBD exposure, and reduced shedding of S1 compared to the wild type. We demonstrate in situ structure determination as a powerful means for studying antigen design options in future vaccine development against emerging novel SARS-CoV-2 variants and broadly against other infectious viruses.
Collapse
Affiliation(s)
- Tao Ni
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Luiza Mendonça
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Yanan Zhu
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Andrew Howe
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - Julika Radecke
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - Pranav M. Shah
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Yuewen Sheng
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
| | - Anna-Sophia Krebs
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Helen M.E. Duyvesteyn
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Elizabeth Allen
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Teresa Lambe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- NIHR Oxford Biomedical Research Centre, Oxford OX3 7BN, UK
| | - Cameron Bisset
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Alexandra Spencer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Susan Morris
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, OX3 7TY, UK
| | - David I. Stuart
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford OX3 7BN, UK
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, OX3 7TY, UK
| | - Sarah Gilbert
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- NIHR Oxford Biomedical Research Centre, Oxford OX3 7BN, UK
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, OX3 7TY, UK
| | - Peijun Zhang
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot OX11 0DE, UK
- Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford OX3 7BN, UK
| |
Collapse
|
5
|
Partial Disturbance of Microprocessor Function in Human Stem Cells Carrying a Heterozygous Mutation in the DGCR8 Gene. Genes (Basel) 2022; 13:genes13111925. [DOI: 10.3390/genes13111925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 11/04/2022] Open
Abstract
Maturation of microRNAs (miRNAs) begins by the “Microprocessor” complex, containing the Drosha endonuclease and its partner protein, "DiGeorge Syndrome Critical Region 8" (DGCR8). Although the main function of the two proteins is to coordinate the first step of precursor miRNAs formation, several studies revealed their miRNA-independent functions in other RNA-related pathways (e.g., in snoRNA decay) or, for the DGCR8, the role in tissue development. To investigate the specific roles of DGCR8 in various cellular pathways, we previously established a human embryonic stem-cell (hESC) line carrying a monoallelic DGCR8 mutation by using the CRISPR-Cas9 system. In this study, we genetically characterized single-cell originated progenies of the cell line and showed that DGCR8 heterozygous mutation results in only a modest effect on the mRNA level but a significant decrease at the protein level. Self-renewal and trilineage differentiation capacity of these hESCs were not affected by the mutation. However, partial disturbance of the Microprocessor function could be revealed in pri-miRNA processing along the human chromosome 19 miRNA cluster in several clones. With all these studies, we can demonstrate that the mutant hESC line is a good model to study not only miRNA-related but also other “noncanonical” functions of the DGCR8 protein.
Collapse
|
6
|
Padron-Regalado E, Ulaszewska M, Douglas AD, Hill AVS, Spencer AJ. STING-pathway modulation to enhance the immunogenicity of adenoviral-vectored vaccines. Sci Rep 2022; 12:14464. [PMID: 36002507 PMCID: PMC9401198 DOI: 10.1038/s41598-022-18750-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/18/2022] [Indexed: 11/09/2022] Open
Abstract
Traditional chemical adjuvants remain a practical means of enhancing the immunogenicity of vaccines. Nevertheless, it is recognized that increasing the immunogenicity of viral vectors is challenging. Recently, STING ligands have been shown to enhance the efficacy of different vaccine platforms, but their affectivity on viral-vectored vaccination has not been fully assessed. In this study we used a multi-pronged approach to shed light on the immunological properties and potential mechanisms of action of this type of adjuvant and focused our study on replication-deficient human adenovirus serotype 5 (AdHu5). When the STING ligand 2'3'-cGAMP was mixed with AdHu5, the adjuvant enhanced anti-vector immune responses while decreasing the transgene-specific CD8+ T cell response. Studies employing STING-knockout mice and a 2'3'-cGAMP inactive analogue confirmed the aforementioned effects were STING dependent. In vitro assays demonstrated 2'3'-cGAMP induced the production of IFN-β which in turn negatively affected AdHu5 transgene expression and CD8+ T cell immunogenicity. In an effort to overcome the negative impact of early 2'3'-cGAMP signaling on AdHu5 transgene immunogenicity, we generated a bicistronic vector encoding the 2'3'-cGAMP together with a model antigen. Intracellular production of 2'3'-cGAMP after AdHu5 infection was able to enhance transgene-specific CD8+ T cell immunogenicity, although not to a level that would warrant progression of this adjuvant to clinical assessment. This work highlights the importance of timing of 2'3'-cGAMP administration when assessing its adjuvant capacity with different vaccine modalities.
Collapse
Affiliation(s)
- Eriko Padron-Regalado
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Marta Ulaszewska
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexander D Douglas
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Adrian V S Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Alexandra J Spencer
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
7
|
Joe CCD, Jiang J, Linke T, Li Y, Fedosyuk S, Gupta G, Berg A, Segireddy RR, Mainwaring D, Joshi A, Cashen P, Rees B, Chopra N, Nestola P, Humphreys J, Davies S, Smith N, Bruce S, Verbart D, Bormans D, Knevelman C, Woodyer M, Davies L, Cooper L, Kapanidou M, Bleckwenn N, Pappas D, Lambe T, Smith DC, Green CM, Venkat R, Ritchie AJ, Gilbert SC, Turner R, Douglas AD. Manufacturing a chimpanzee adenovirus-vectored SARS-CoV-2 vaccine to meet global needs. Biotechnol Bioeng 2022; 119:48-58. [PMID: 34585736 PMCID: PMC8653296 DOI: 10.1002/bit.27945] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/14/2021] [Accepted: 09/21/2021] [Indexed: 12/16/2022]
Abstract
Manufacturing has been the key factor limiting rollout of vaccination during the COVID-19 pandemic, requiring rapid development and large-scale implementation of novel manufacturing technologies. ChAdOx1 nCoV-19 (AZD1222, Vaxzevria) is an efficacious vaccine against SARS-CoV-2, based upon an adenovirus vector. We describe the development of a process for the production of this vaccine and others based upon the same platform, including novel features to facilitate very large-scale production. We discuss the process economics and the "distributed manufacturing" approach we have taken to provide the vaccine at globally-relevant scale and with international security of supply. Together, these approaches have enabled the largest viral vector manufacturing campaign to date, providing a substantial proportion of global COVID-19 vaccine supply at low cost.
Collapse
Affiliation(s)
- Carina C. D. Joe
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Jinlin Jiang
- Biopharmaceuticals DevelopmentR&D, AstraZenecaGaithersburgMarylandUSA
| | - Thomas Linke
- Biopharmaceuticals DevelopmentR&D, AstraZenecaGaithersburgMarylandUSA
| | - Yuanyuan Li
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Sofiya Fedosyuk
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Gaurav Gupta
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Adam Berg
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Nicole Bleckwenn
- Biopharmaceuticals DevelopmentR&D, AstraZenecaGaithersburgMarylandUSA
| | - Daniel Pappas
- Biopharmaceuticals DevelopmentR&D, AstraZenecaGaithersburgMarylandUSA
| | - Teresa Lambe
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | | | - Catherine M. Green
- Clinical Biomanufacturing Facility, Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Raghavan Venkat
- Biopharmaceuticals DevelopmentR&D, AstraZenecaGaithersburgMarylandUSA
| | - Adam J. Ritchie
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Sarah C. Gilbert
- Nuffield Department of Medicine, Jenner InstituteUniversity of OxfordOxfordUK
| | - Richard Turner
- Purification Process Sciences, Biopharmaceuticals DevelopmentR&D, AstraZenecaCambridgeUK
| | | |
Collapse
|
8
|
Vatzia E, Allen ER, Manjegowda T, Morris S, McNee A, Martini V, Kaliath R, Ulaszewska M, Boyd A, Paudyal B, Carr VB, Chrun T, Maze E, MacLoughlin R, van Diemen PM, Everett HE, Lambe T, Gilbert SC, Tchilian E. Respiratory and Intramuscular Immunization With ChAdOx2-NPM1-NA Induces Distinct Immune Responses in H1N1pdm09 Pre-Exposed Pigs. Front Immunol 2021; 12:763912. [PMID: 34804053 PMCID: PMC8595216 DOI: 10.3389/fimmu.2021.763912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/13/2021] [Indexed: 01/12/2023] Open
Abstract
There is a critical need to develop superior influenza vaccines that provide broader protection. Influenza vaccines are traditionally tested in naive animals, although humans are exposed to influenza in the first years of their lives, but the impact of prior influenza exposure on vaccine immune responses has not been well studied. Pigs are an important natural host for influenza, are a source of pandemic viruses, and are an excellent model for human influenza. Here, we investigated the immunogenicity of the ChAdOx2 viral vectored vaccine, expressing influenza nucleoprotein, matrix protein 1, and neuraminidase in H1N1pdm09 pre-exposed pigs. We evaluated the importance of the route of administration by comparing intranasal, aerosol, and intramuscular immunizations. Aerosol delivery boosted the local lung T-cell and antibody responses, while intramuscular immunization boosted peripheral blood immunity. These results will inform how best to deliver vaccines in order to harness optimal protective immunity.
Collapse
Affiliation(s)
- Eleni Vatzia
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Elizabeth R Allen
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Tanuja Manjegowda
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Susan Morris
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Adam McNee
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Veronica Martini
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Reshma Kaliath
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Marta Ulaszewska
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Amy Boyd
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Basudev Paudyal
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Veronica B Carr
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Tiphany Chrun
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Emmanuel Maze
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | | | | | - Helen E Everett
- Animal and Plant Health Agency (APHA)-Weybridge, Addlestone, United Kingdom
| | - Teresa Lambe
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sarah C Gilbert
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Elma Tchilian
- Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| |
Collapse
|
9
|
Rojas JM, Sevilla N, Martín V. A New Look at Vaccine Strategies Against PPRV Focused on Adenoviral Candidates. Front Vet Sci 2021; 8:729879. [PMID: 34568477 PMCID: PMC8455998 DOI: 10.3389/fvets.2021.729879] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/09/2021] [Indexed: 11/28/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) is a virus that mainly infects goats and sheep causing significant economic loss in Africa and Asia, but also posing a serious threat to Europe, as recent outbreaks in Georgia (2016) and Bulgaria (2018) have been reported. In order to carry out the eradication of PPRV, an objective set for 2030 by the Office International des Epizooties (OIE) and the Food and Agriculture Organization of the United Nations (FAO), close collaboration between governments, pharmaceutical companies, farmers and researchers, among others, is needed. Today, more than ever, as seen in the response to the SARS-CoV2 pandemic that we are currently experiencing, these goals are feasible. We summarize in this review the current vaccination approaches against PPRV in the field, discussing their advantages and shortfalls, as well as the development and generation of new vaccination strategies, focusing on the potential use of adenovirus as vaccine platform against PPRV and more broadly against other ruminant pathogens.
Collapse
Affiliation(s)
| | | | - Verónica Martín
- Centro de Investigación en Sanidad Animal (CISA-INIA-CSIC), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
10
|
Fischer RJ, Purushotham JN, van Doremalen N, Sebastian S, Meade-White K, Cordova K, Letko M, Jeremiah Matson M, Feldmann F, Haddock E, LaCasse R, Saturday G, Lambe T, Gilbert SC, Munster VJ. ChAdOx1-vectored Lassa fever vaccine elicits a robust cellular and humoral immune response and protects guinea pigs against lethal Lassa virus challenge. NPJ Vaccines 2021; 6:32. [PMID: 33654106 PMCID: PMC7925663 DOI: 10.1038/s41541-021-00291-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 01/27/2021] [Indexed: 01/31/2023] Open
Abstract
Lassa virus (LASV) infects hundreds of thousands of individuals each year, highlighting the need for the accelerated development of preventive, diagnostic, and therapeutic interventions. To date, no vaccine has been licensed for LASV. ChAdOx1-Lassa-GPC is a chimpanzee adenovirus-vectored vaccine encoding the Josiah strain LASV glycoprotein precursor (GPC) gene. In the following study, we show that ChAdOx1-Lassa-GPC is immunogenic, inducing robust T-cell and antibody responses in mice. Furthermore, a single dose of ChAdOx1-Lassa-GPC fully protects Hartley guinea pigs against morbidity and mortality following lethal challenge with a guinea pig-adapted LASV (strain Josiah). By contrast, control vaccinated animals reached euthanasia criteria 10-12 days after infection. Limited amounts of LASV RNA were detected in the tissues of vaccinated animals. Viable LASV was detected in only one animal receiving a single dose of the vaccine. A prime-boost regimen of ChAdOx1-Lassa-GPC in guinea pigs significantly increased antigen-specific antibody titers and cleared viable LASV from the tissues. These data support further development of ChAdOx1-Lassa-GPC and testing in non-human primate models of infection.
Collapse
Affiliation(s)
- Robert J. Fischer
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA
| | - Jyothi N. Purushotham
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA ,grid.4991.50000 0004 1936 8948The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Neeltje van Doremalen
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA
| | - Sarah Sebastian
- grid.4991.50000 0004 1936 8948The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK ,Present Address: Vaccitech Limited, Oxford, UK
| | - Kimberly Meade-White
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA
| | - Kathleen Cordova
- grid.419681.30000 0001 2164 9667Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Michael Letko
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA ,grid.30064.310000 0001 2157 6568Paul G. Allen School of Global Animal Health, Washington State University, Pullman, WA USA
| | - M. Jeremiah Matson
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA ,grid.36425.360000 0001 2216 9681Marshall University Joan C. Edwards School of Medicine, Huntington, WV USA
| | - Friederike Feldmann
- grid.419681.30000 0001 2164 9667Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Elaine Haddock
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA
| | - Rachel LaCasse
- grid.419681.30000 0001 2164 9667Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Greg Saturday
- grid.419681.30000 0001 2164 9667Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT USA
| | - Teresa Lambe
- grid.4991.50000 0004 1936 8948The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah C. Gilbert
- grid.4991.50000 0004 1936 8948The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Vincent J. Munster
- grid.419681.30000 0001 2164 9667Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT USA
| |
Collapse
|
11
|
van Doremalen N, Haddock E, Feldmann F, Meade-White K, Bushmaker T, Fischer RJ, Okumura A, Hanley PW, Saturday G, Edwards NJ, Clark MHA, Lambe T, Gilbert SC, Munster VJ. A single dose of ChAdOx1 MERS provides protective immunity in rhesus macaques. SCIENCE ADVANCES 2020; 6:eaba8399. [PMID: 32577525 PMCID: PMC7286676 DOI: 10.1126/sciadv.aba8399] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/16/2020] [Indexed: 05/21/2023]
Abstract
Developing a vaccine to protect against the lethal effects of the many strains of coronavirus is critical given the current global pandemic. For Middle East respiratory syndrome coronavirus (MERS-CoV), we show that rhesus macaques seroconverted rapidly after a single intramuscular vaccination with ChAdOx1 MERS. The vaccine protected against respiratory injury and pneumonia and reduced viral load in lung tissue by several orders of magnitude. MERS-CoV replication in type I and II pneumocytes of ChAdOx1 MERS-vaccinated animals was absent. A prime-boost regimen of ChAdOx1 MERS boosted antibody titers, and viral replication was completely absent from the respiratory tract tissue of these rhesus macaques. We also found that antibodies elicited by ChAdOx1 MERS in rhesus macaques neutralized six different MERS-CoV strains. Transgenic human dipeptidyl peptidase 4 mice vaccinated with ChAdOx1 MERS were completely protected against disease and lethality for all different MERS-CoV strains. The data support further clinical development of ChAdOx1 MERS.
Collapse
Affiliation(s)
- Neeltje van Doremalen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
- Corresponding author. (N.v.D.); (S.C.G.); (V.J.M.)
| | - Elaine Haddock
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Trenton Bushmaker
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Robert J. Fischer
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Atsushi Okumura
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Patrick W. Hanley
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | | | - Madeleine H. A. Clark
- The Jenner Institute, University of Oxford, Oxford, UK
- Transmission Biology Group, The Pirbright Institute, Pirbright, Woking, UK
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Oxford, UK
| | - Sarah C. Gilbert
- The Jenner Institute, University of Oxford, Oxford, UK
- Corresponding author. (N.v.D.); (S.C.G.); (V.J.M.)
| | - Vincent J. Munster
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
- Corresponding author. (N.v.D.); (S.C.G.); (V.J.M.)
| |
Collapse
|
12
|
Modification of Adenovirus vaccine vector-induced immune responses by expression of a signalling molecule. Sci Rep 2020; 10:5716. [PMID: 32235848 PMCID: PMC7109070 DOI: 10.1038/s41598-020-61730-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/05/2020] [Indexed: 11/09/2022] Open
Abstract
Adenoviral vectors are being developed as vaccines against infectious agents and tumour-associated antigens, because of their ability to induce cellular immunity. However, the protection afforded in animal models has not easily translated into primates and clinical trials, underlying the need for improving adenoviral vaccines-induced immunogenicity. A Toll-like receptor signalling molecule, TRAM, was assessed for its ability to modify the immune responses induced by an adenovirus-based vaccine. Different adenovirus vectors either expressing TRAM alone or co-expressing TRAM along with a model antigen were constructed. The modification of T-cell and antibody responses induced by TRAM was assessed in vivo in mice and in primates. Co-expression of TRAM and an antigen from adenoviruses increased the transgene-specific CD8+ T cell responses in mice. Similar effects were seen when a TRAM expressing virus was co-administered with the antigen-expressing adenovirus. However, in primate studies, co-administration of a TRAM expressing adenovirus with a vaccine expressing the ME-TRAP malaria antigen had no significant effect on the immune responses. While these results support the idea that modification of innate immune signalling by genetic vectors modifies immunogenicity, they also emphasise the difficulty in generalising results from rodents into primates, where the regulatory pathway may be different to that in mice.
Collapse
|
13
|
Bliss CM, Parsons AJ, Nachbagauer R, Hamilton JR, Cappuccini F, Ulaszewska M, Webber JP, Clayton A, Hill AV, Coughlan L. Targeting Antigen to the Surface of EVs Improves the In Vivo Immunogenicity of Human and Non-human Adenoviral Vaccines in Mice. Mol Ther Methods Clin Dev 2020; 16:108-125. [PMID: 31934599 PMCID: PMC6953706 DOI: 10.1016/j.omtm.2019.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/12/2019] [Indexed: 12/25/2022]
Abstract
Adenoviral (Ad) vectors represent promising vaccine platforms for infectious disease. To overcome pre-existing immunity to commonly used human adenovirus serotype 5 (Ad5), vectors based on rare species or non-human Ads are being developed. However, these vectors often exhibit reduced potency compared with Ad5, necessitating the use of innovative approaches to augment the immunogenicity of the encoded antigen (Ag). To achieve this, we engineered model Ag, enhanced green fluorescent protein (EGFP), for targeting to the surface of host-derived extracellular vesicles (EVs), namely exosomes. Exosomes are nano-sized EVs that play important roles in cell-to-cell communication and in regulating immune responses. Directed targeting of Ag to the surface of EVs/exosomes is achieved by "exosome display," through fusion of Ag to the C1C2 domain of lactadherin, a protein highly enriched in exosomes. Herein, we engineered chimpanzee adenovirus ChAdOx1 and Ad5-based vaccines encoding EGFP, or EGFP targeted to EVs (EGFP_C1C2), and compared vaccine immunogenicity in mice. We determined that exosome display substantially increases Ag-specific humoral immunity following intramuscular and intranasal vaccination, improving the immunological potency of both ChAdOx1 and Ad5. We propose that this Ag-engineering approach could increase the immunogenicity of diverse Ad vectors that exhibit desirable manufacturing characteristics, but currently lack the potency of Ad5.
Collapse
Affiliation(s)
- Carly M. Bliss
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Andrea J. Parsons
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Raffael Nachbagauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jennifer R. Hamilton
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Federica Cappuccini
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, ORCRB Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Marta Ulaszewska
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, ORCRB Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Jason P. Webber
- Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 2XN, UK
| | - Aled Clayton
- Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 2XN, UK
| | - Adrian V.S. Hill
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, ORCRB Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | - Lynda Coughlan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, ORCRB Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| |
Collapse
|
14
|
van Doremalen N, Lambe T, Sebastian S, Bushmaker T, Fischer R, Feldmann F, Haddock E, Letko M, Avanzato VA, Rissanen I, LaCasse R, Scott D, Bowden TA, Gilbert S, Munster V. A single-dose ChAdOx1-vectored vaccine provides complete protection against Nipah Bangladesh and Malaysia in Syrian golden hamsters. PLoS Negl Trop Dis 2019; 13:e0007462. [PMID: 31170144 PMCID: PMC6581282 DOI: 10.1371/journal.pntd.0007462] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/18/2019] [Accepted: 05/13/2019] [Indexed: 11/23/2022] Open
Abstract
Nipah virus (NiV) is a highly pathogenic re-emerging virus that causes outbreaks in South East Asia. Currently, no approved and licensed vaccine or antivirals exist. Here, we investigated the efficacy of ChAdOx1 NiVB, a simian adenovirus-based vaccine encoding NiV glycoprotein (G) Bangladesh, in Syrian hamsters. Prime-only as well as prime-boost vaccination resulted in uniform protection against a lethal challenge with NiV Bangladesh: all animals survived challenge and we were unable to find infectious virus either in oral swabs, lung or brain tissue. Furthermore, no pathological lung damage was observed. A single-dose of ChAdOx1 NiVB also prevented disease and lethality from heterologous challenge with NiV Malaysia. While we were unable to detect infectious virus in swabs or tissue of animals challenged with the heterologous strain, a very limited amount of viral RNA could be found in lung tissue by in situ hybridization. A single dose of ChAdOx1 NiVB also provided partial protection against Hendra virus and passive transfer of antibodies elicited by ChAdOx1 NiVB vaccination partially protected Syrian hamsters against NiV Bangladesh. From these data, we conclude that ChAdOx1 NiVB is a suitable candidate for further NiV vaccine pre-clinical development.
Collapse
Affiliation(s)
- Neeltje van Doremalen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, United States of America
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sarah Sebastian
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Trenton Bushmaker
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, United States of America
| | - Robert Fischer
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, United States of America
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States of America
| | - Elaine Haddock
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, United States of America
| | - Michael Letko
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, United States of America
| | - Victoria A. Avanzato
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, United States of America
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ilona Rissanen
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Rachel LaCasse
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States of America
| | - Dana Scott
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, United States of America
| | - Thomas A. Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Sarah Gilbert
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Vincent Munster
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, United States of America
| |
Collapse
|
15
|
Fedosyuk S, Merritt T, Peralta-Alvarez MP, Morris SJ, Lam A, Laroudie N, Kangokar A, Wright D, Warimwe GM, Angell-Manning P, Ritchie AJ, Gilbert SC, Xenopoulos A, Boumlic A, Douglas AD. Simian adenovirus vector production for early-phase clinical trials: A simple method applicable to multiple serotypes and using entirely disposable product-contact components. Vaccine 2019; 37:6951-6961. [PMID: 31047679 PMCID: PMC6949866 DOI: 10.1016/j.vaccine.2019.04.056] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/14/2019] [Accepted: 04/19/2019] [Indexed: 12/21/2022]
Abstract
A variety of Good Manufacturing Practice (GMP) compliant processes have been reported for production of non-replicating adenovirus vectors, but important challenges remain. Most clinical development of adenovirus vectors now uses simian adenoviruses or rare human serotypes, whereas reported manufacturing processes mainly use serotypes such as AdHu5 which are of questionable relevance for clinical vaccine development. Many clinically relevant vaccine transgenes interfere with adenovirus replication, whereas most reported process development uses selected antigens or even model transgenes such as fluorescent proteins which cause little such interference. Processes are typically developed for a single adenovirus serotype - transgene combination, requiring extensive further optimization for each new vaccine. There is a need for rapid production platforms for small GMP batches of non-replicating adenovirus vectors for early-phase vaccine trials, particularly in preparation for response to emerging pathogen outbreaks. Such platforms must be robust to variation in the transgene, and ideally also capable of producing adenoviruses of more than one serotype. It is also highly desirable for such processes to be readily implemented in new facilities using commercially available single-use materials, avoiding the need for development of bespoke tools or cleaning validation, and for them to be readily scalable for later-stage studies. Here we report the development of such a process, using single-use stirred-tank bioreactors, a transgene-repressing HEK293 cell - promoter combination, and fully single-use filtration and ion exchange components. We demonstrate applicability of the process to candidate vaccines against rabies, malaria and Rift Valley fever, each based on a different adenovirus serotype. We compare performance of a range of commercially available ion exchange media, including what we believe to be the first published use of a novel media for adenovirus purification (NatriFlo® HD-Q, Merck). We demonstrate the need for minimal process individualization for each vaccine, and that the product fulfils regulatory quality expectations. Cell-specific yields are at the upper end of those previously reported in the literature, and volumetric yields are in the range 1 × 1013 - 5 × 1013 purified virus particles per litre of culture, such that a 2-4 L process is comfortably adequate to produce vaccine for early-phase trials. The process is readily transferable to any GMP facility with the capability for mammalian cell culture and aseptic filling of sterile products.
Collapse
Affiliation(s)
- Sofiya Fedosyuk
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Thomas Merritt
- Clinical Biomanufacturing Facility, University of Oxford, Roosevelt Drive, Oxford OX3 7JT, UK
| | | | - Susan J Morris
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Ada Lam
- Millipore (UK) Ltd. Bedfont Cross, Stanwell Road, TW14 8NX Feltham, UK
| | - Nicolas Laroudie
- Millipore SAS, 39 Route Industrielle de la Hardt, Molsheim 67120, France
| | | | - Daniel Wright
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - George M Warimwe
- Centre for Tropical Medicine and Global Health, University of Oxford, Roosevelt Drive, Oxford OX3 7FZ, UK; KEMRI-Wellcome Trust Research Programme, P.O. 230-80108 Kilifi, Kenya
| | - Phillip Angell-Manning
- Clinical Biomanufacturing Facility, University of Oxford, Roosevelt Drive, Oxford OX3 7JT, UK
| | - Adam J Ritchie
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Sarah C Gilbert
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Alex Xenopoulos
- EMD Millipore Corporation, 80 Ashby Road, Bedford, MA 01730, USA
| | - Anissa Boumlic
- Millipore SAS, 39 Route Industrielle de la Hardt, Molsheim 67120, France
| | - Alexander D Douglas
- Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK.
| |
Collapse
|
16
|
Fougeroux C, Turner L, Bojesen AM, Lavstsen T, Holst PJ. Modified MHC Class II-Associated Invariant Chain Induces Increased Antibody Responses against Plasmodium falciparum Antigens after Adenoviral Vaccination. THE JOURNAL OF IMMUNOLOGY 2019; 202:2320-2331. [PMID: 30833346 DOI: 10.4049/jimmunol.1801210] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 02/01/2019] [Indexed: 01/04/2023]
Abstract
Adenoviral vectors can induce T and B cell immune responses to Ags encoded in the recombinant vector. The MHC class II invariant chain (Ii) has been used as an adjuvant to enhance T cell responses to tethered Ag encoded in adenoviral vectors. In this study, we modified the Ii adjuvant by insertion of a furin recognition site (Ii-fur) to obtain a secreted version of the Ii. To test the capacity of this adjuvant to enhance immune responses, we recombined vectors to encode Plasmodium falciparum virulence factors: two cysteine-rich interdomain regions (CIDR) α1 (IT4var19 and PFCLINvar30 var genes), expressed as a dimeric Ag. These domains are members of a highly polymorphic protein family involved in the vascular sequestration and immune evasion of parasites in malaria. The Ii-fur molecule directed secretion of both Ags in African green monkey cells and functioned as an adjuvant for MHC class I and II presentation in T cell hybridomas. In mice, the Ii-fur adjuvant induced a similar T cell response, as previously demonstrated with Ii, accelerated and enhanced the specific Ab response against both CIDR Ags, with an increased binding capacity to the cognate endothelial protein C receptor, and enhanced the breadth of the response toward different CIDRs. We also demonstrate that the endosomal sorting signal, secretion, and the C-terminal part of Ii were needed for the full adjuvant effect for Ab responses. We conclude that engineered secretion of Ii adjuvant-tethered Ags establishes a single adjuvant and delivery vehicle platform for potent T and B cell-dependent immunity.
Collapse
Affiliation(s)
- Cyrielle Fougeroux
- Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; and
| | - Louise Turner
- Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; and
| | - Anders Miki Bojesen
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark
| | - Thomas Lavstsen
- Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; and
| | - Peter Johannes Holst
- Center for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark; and
| |
Collapse
|
17
|
Adenovirus based virus-like-vaccines targeting endogenous retroviruses can eliminate growing colorectal cancers in mice. Oncotarget 2019; 10:1458-1472. [PMID: 30858929 PMCID: PMC6402721 DOI: 10.18632/oncotarget.26680] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 02/01/2019] [Indexed: 12/31/2022] Open
Abstract
Endogenous retroviruses (ERVs) that make up 8% of the human genome have been associated with the development and progression of cancer. The murine model system of the melanoma associated retrovirus (MelARV), which is expressed in different murine cancer cell lines, can be used to study mechanisms and therapeutic approaches against ERVs in cancer. We designed a vaccine strategy (Ad5-MelARV) of adenoviruses encoding the MelARV proteins Gag and Env that assemble in vivo into virus-like particles displaying the cancer-associated MelARV Env to the immune system. The novel vaccine was designed to induce both humoral as well as cellular immune responses in order to attack ERV expressing tumor cells. Despite a lack of antibody induction, we found that T cell responses were strong enough to prevent colorectal CT26 tumor growth and progression in BALB/c mice after a single vaccination before or after tumor challenge. A combination with the checkpoint inhibitor anti-PD-1 further increased the efficacy of the vaccination leading to complete tumor regression. Furthermore, immune responses in vaccinated mice were not restricted to only one cancer cell line but vaccinated animals were also protected from a rechallenge with the distinct breast cancer cell line 4T1. Thus, the developed vaccine strategy could represent a novel tool to successfully target diverse ERV-bearing tumors in cancer patients.
Collapse
|
18
|
Halbroth BR, Sebastian S, Poyntz HC, Bregu M, Cottingham MG, Hill AVS, Spencer AJ. Development of a Molecular Adjuvant to Enhance Antigen-Specific CD8 + T Cell Responses. Sci Rep 2018; 8:15020. [PMID: 30301933 PMCID: PMC6177389 DOI: 10.1038/s41598-018-33375-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 09/27/2018] [Indexed: 12/26/2022] Open
Abstract
Despite promising progress in malaria vaccine development, an efficacious subunit vaccine against P. falciparum remains to be licensed and deployed. This study aimed to improve on the immunogenicity of the leading liver-stage vaccine candidate (ChAd63-MVA ME-TRAP), known to confer protection by eliciting high levels of antigen-specific CD8+ T cells. We previously showed fusion of ME-TRAP to the human MHC class II invariant chain (Ii) could enhance CD8+ T cell responses in non-human primates, but did not progress to clinical testing due to potential risk of auto-immunity by vaccination of humans with a self-antigen. Initial immunogenicity analyses of ME-TRAP fused to subdomains of the Ii showed that the Ii transmembrane domain alone can enhance CD8+ T cell responses. Subsequently, truncated Ii sequences with low homology to human Ii were developed and shown to enhance CD8+ T cell responses. By systematically mutating the TM domain sequence, multimerization of the Ii chain was shown to be important for immune enhancement. We subsequently identified several proteins from a variety of microbial pathogens with similar characteristics, that also enhance the CD8+ T cell response and could therefore be used in viral vector vaccines when potent cell mediated immunity is required.
Collapse
Affiliation(s)
- Benedict R Halbroth
- The Jenner Institute, University of Oxford, ORCRB, Roosevelt Drive, Oxford, United Kingdom.
| | - Sarah Sebastian
- The Jenner Institute, University of Oxford, ORCRB, Roosevelt Drive, Oxford, United Kingdom
| | - Hazel C Poyntz
- The Jenner Institute, University of Oxford, ORCRB, Roosevelt Drive, Oxford, United Kingdom
| | - Migena Bregu
- The Jenner Institute, University of Oxford, ORCRB, Roosevelt Drive, Oxford, United Kingdom
| | - Matthew G Cottingham
- The Jenner Institute, University of Oxford, ORCRB, Roosevelt Drive, Oxford, United Kingdom
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, ORCRB, Roosevelt Drive, Oxford, United Kingdom
| | - Alexandra J Spencer
- The Jenner Institute, University of Oxford, ORCRB, Roosevelt Drive, Oxford, United Kingdom.
| |
Collapse
|
19
|
Wang C, Dulal P, Zhou X, Xiang Z, Goharriz H, Banyard A, Green N, Brunner L, Ventura R, Collin N, Draper SJ, Hill AVS, Ashfield R, Fooks AR, Ertl HC, Douglas AD. A simian-adenovirus-vectored rabies vaccine suitable for thermostabilisation and clinical development for low-cost single-dose pre-exposure prophylaxis. PLoS Negl Trop Dis 2018; 12:e0006870. [PMID: 30372438 PMCID: PMC6224154 DOI: 10.1371/journal.pntd.0006870] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/08/2018] [Accepted: 09/24/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Estimates of current global rabies mortality range from 26,000 to 59,000 deaths per annum. Although pre-exposure prophylaxis using inactivated rabies virus vaccines (IRVs) is effective, it requires two to three doses and is regarded as being too expensive and impractical for inclusion in routine childhood immunization programmes. METHODOLOGY/ PRINCIPAL FINDINGS Here we report the development of a simian-adenovirus-vectored rabies vaccine intended to enable cost-effective population-wide pre-exposure prophylaxis against rabies. ChAdOx2 RabG uses the chimpanzee adenovirus serotype 68 (AdC68) backbone previously shown to achieve pre-exposure protection against rabies in non-human primates. ChAdOx2 differs from AdC68 in that it contains the human adenovirus serotype 5 (AdHu5) E4 orf6/7 region in place of the AdC68 equivalents, enhancing ease of manufacturing in cell lines which provide AdHu5 E1 proteins in trans. We show that immunogenicity of ChAdOx2 RabG in mice is comparable to that of AdC68 RabG and other adenovirus serotypes expressing rabies virus glycoprotein. High titers of rabies virus neutralizing antibody (VNA) are elicited after a single dose. The relationship between levels of VNA activity and rabies virus glycoprotein monomer-binding antibody differs after immunization with adenovirus-vectored vaccines and IRV vaccines, suggesting routes to further enhancement of the efficacy of the adenovirus-vectored candidates. We also demonstrate that ChAdOx2 RabG can be thermostabilised using a low-cost method suitable for clinical bio-manufacture and ambient-temperature distribution in tropical climates. Finally, we show that a dose-sparing effect can be achieved by formulating ChAdOx2 RabG with a simple chemical adjuvant. This approach could lower the cost of ChAdOx2 RabG and other adenovirus-vectored vaccines. CONCLUSIONS/ SIGNIFICANCE ChAdOx2 RabG may prove to be a useful tool to reduce the human rabies death toll. We have secured funding for Good Manufacturing Practice- compliant bio-manufacture and Phase I clinical trial of this candidate.
Collapse
Affiliation(s)
- Chuan Wang
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | - Pawan Dulal
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | - Xiangyang Zhou
- Wistar Institute of Anatomy & Biology, Philadelphia, Pennsylvania, United States of America
| | - Zhiquan Xiang
- Wistar Institute of Anatomy & Biology, Philadelphia, Pennsylvania, United States of America
| | - Hooman Goharriz
- Animal and Plant Health Agency (APHA), Wildlife Zoonoses and Vector-borne Diseases Research Group, New Haw, Surrey, United Kingdom
| | - Ashley Banyard
- Animal and Plant Health Agency (APHA), Wildlife Zoonoses and Vector-borne Diseases Research Group, New Haw, Surrey, United Kingdom
| | - Nicky Green
- Clinical Biomanufacturing Facility, University of Oxford, Oxford, United Kingdom
| | - Livia Brunner
- Vaccine Formulation Laboratory, University of Lausanne, Epalinges, Switzerland
| | - Roland Ventura
- Vaccine Formulation Laboratory, University of Lausanne, Epalinges, Switzerland
| | - Nicolas Collin
- Vaccine Formulation Laboratory, University of Lausanne, Epalinges, Switzerland
| | - Simon J. Draper
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | - Adrian V. S. Hill
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | - Rebecca Ashfield
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | - Anthony R. Fooks
- Animal and Plant Health Agency (APHA), Wildlife Zoonoses and Vector-borne Diseases Research Group, New Haw, Surrey, United Kingdom
| | - Hildegund C. Ertl
- Wistar Institute of Anatomy & Biology, Philadelphia, Pennsylvania, United States of America
| | - Alexander D. Douglas
- Jenner Institute, University of Oxford, Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| |
Collapse
|
20
|
Virus-Like-Vaccines against HIV. Vaccines (Basel) 2018; 6:vaccines6010010. [PMID: 29439476 PMCID: PMC5874651 DOI: 10.3390/vaccines6010010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/02/2018] [Accepted: 02/10/2018] [Indexed: 12/27/2022] Open
Abstract
Protection against chronic infections has necessitated the development of ever-more potent vaccination tools. HIV seems to be the most challenging foe, with a remarkable, poorly immunogenic and fragile surface glycoprotein and the ability to overpower the cell immune system. Virus-like-particle (VLP) vaccines have emerged as potent inducers of antibody and helper T cell responses, while replication-deficient viral vectors have yielded potent cytotoxic T cell responses. Here, we review the emerging concept of merging these two technologies into virus-like-vaccines (VLVs) for the targeting of HIV. Such vaccines are immunologically perceived as viruses, as they infect cells and produce VLPs in situ, but they only resemble viruses, as the replication defective vectors and VLPs cannot propagate an infection. The inherent safety of such a platform, despite robust particle production, is a distinct advantage over live-attenuated vaccines that must balance safety and immunogenicity. Previous studies have delivered VLVs encoded in modified Vaccinia Ankara vectors and we have developed the concept into a single-reading adenovirus-based technology capable of eliciting robust CD8+ and CD4+ T cells responses and trimer binding antibody responses. Such vaccines offer the potential to display the naturally produced immunogen directly and induce an integrated humoral and cellular immune response.
Collapse
|
21
|
Menon V, Kapulu MC, Taylor I, Jewell K, Li Y, Hill F, Long CA, Miura K, Biswas S. Assessment of Antibodies Induced by Multivalent Transmission-Blocking Malaria Vaccines. Front Immunol 2018; 8:1998. [PMID: 29403479 PMCID: PMC5780346 DOI: 10.3389/fimmu.2017.01998] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/22/2017] [Indexed: 01/20/2023] Open
Abstract
A malaria transmission-blocking vaccine would be a critical tool in achieving malaria elimination and eradication. By using chimpanzee adenovirus serotype 63 and modified vaccinia virus Ankara viral vectored vaccines, we investigated whether incorporating two antigens into one vaccine would result in higher transmission-reducing activity than one antigen. We demonstrated that when Pfs25 was administered with other antigens Pfs28 or Pfs230C, either concurrently as a mixed vaccine or co-expressed as a dual-antigen vaccine, the antibody response in mice to each antigen was comparable to a monoantigen vaccine, without immunological interference. However, we found that the transmission-reducing activity (functional activity) of dual-antigen vaccines was not additive. Dual-antigen vaccines generally only elicited similar transmission-reducing activity to monoantigen vaccines and in one instance had lower transmission-reducing activity. We found that despite the lack of immunological interference of dual-antigen vaccines, they are still not as effective at blocking malaria transmission as Pfs25-IMX313, the current leading candidate for viral vectored vaccines. Pfs25-IMX313 elicited similar quality antibodies to dual-antigen vaccines, but higher antibody titers.
Collapse
Affiliation(s)
- Vinay Menon
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Iona Taylor
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Kerry Jewell
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Yuanyuan Li
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Carole A. Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, United States
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, United States
| | - Sumi Biswas
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
22
|
An in vitro assay to measure antibody-mediated inhibition of P. berghei sporozoite invasion against P. falciparum antigens. Sci Rep 2017; 7:17011. [PMID: 29209029 PMCID: PMC5717233 DOI: 10.1038/s41598-017-17274-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/21/2017] [Indexed: 02/02/2023] Open
Abstract
A large research effort is currently underway to find an effective and affordable malaria vaccine. Tools that enable the rapid evaluation of protective immune responses are essential to vaccine development as they can provide selection criteria to rank order vaccine candidates. In this study we have revisited the Inhibition of Sporozoite Invasion (ISI) assay to assess the ability of antibodies to inhibit sporozoite infection of hepatocytes. By using GFP expressing sporozoites of the rodent parasite P. berghei we are able to robustly quantify parasite infection of hepatocyte cell lines by flow cytometry. In conjunction with recently produced transgenic P. berghei parasites that express P. falciparum sporozoite antigens, we have been able to use this assay to measure antibody mediated inhibition of sporozoite invasion against one of the lead malaria antigens P. falciparum CSP. By combining chimeric rodent parasites expressing P. falciparum antigens and a flow cytometric readout of infection, we are able to robustly assess vaccine-induced antibodies, from mice, rhesus macaques and human clinical trials, for their functional ability to block sporozoite invasion of hepatocytes.
Collapse
|
23
|
Alharbi NK, Padron-Regalado E, Thompson CP, Kupke A, Wells D, Sloan MA, Grehan K, Temperton N, Lambe T, Warimwe G, Becker S, Hill AVS, Gilbert SC. ChAdOx1 and MVA based vaccine candidates against MERS-CoV elicit neutralising antibodies and cellular immune responses in mice. Vaccine 2017; 35:3780-3788. [PMID: 28579232 PMCID: PMC5516308 DOI: 10.1016/j.vaccine.2017.05.032] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/30/2017] [Accepted: 05/10/2017] [Indexed: 01/28/2023]
Abstract
Inserting tPA leader sequence enhanced humoral responses of ChAdOx1 MERS. ChAdOx1 MERS elicited cellular immunity and neutralising antibodies. ChAdOx1 MERS responses were boosted by MVA MERS. Immunogenicity of a single dose of ChAdOx1 MERS was equivalent to 2 doses of MVA MERS. In MVA, F11 promoter enhanced cellular, but not humoral, immunogenicity, comparing to mH5 promoter.
The Middle East respiratory syndrome coronavirus (MERS-CoV) has infected more than 1900 humans, since 2012. The syndrome ranges from asymptomatic and mild cases to severe pneumonia and death. The virus is believed to be circulating in dromedary camels without notable symptoms since the 1980s. Therefore, dromedary camels are considered the only animal source of infection. Neither antiviral drugs nor vaccines are approved for veterinary or medical use despite active research on this area. Here, we developed four vaccine candidates against MERS-CoV based on ChAdOx1 and MVA viral vectors, two candidates per vector. All vaccines contained the full-length spike gene of MERS-CoV; ChAdOx1 MERS vaccines were produced with or without the leader sequence of the human tissue plasminogen activator gene (tPA) where MVA MERS vaccines were produced with tPA, but either the mH5 or F11 promoter driving expression of the spike gene. All vaccine candidates were evaluated in a mouse model in prime only or prime-boost regimens. ChAdOx1 MERS with tPA induced higher neutralising antibodies than ChAdOx1 MERS without tPA. A single dose of ChAdOx1 MERS with tPA elicited cellular immune responses as well as neutralising antibodies that were boosted to a significantly higher level by MVA MERS. The humoral immunogenicity of a single dose of ChAdOx1 MERS with tPA was equivalent to two doses of MVA MERS (also with tPA). MVA MERS with mH5 or F11 promoter induced similar antibody levels; however, F11 promoter enhanced the cellular immunogenicity of MVA MERS to significantly higher magnitudes. In conclusion, our study showed that MERS-CoV vaccine candidates could be optimized by utilising different viral vectors, various genetic designs of the vectors, or different regimens to increase immunogenicity. ChAdOx1 and MVA vectored vaccines have been safely evaluated in camels and humans and these MERS vaccine candidates should now be tested in camels and in clinical trials.
Collapse
Affiliation(s)
- Naif Khalaf Alharbi
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.
| | | | - Craig P Thompson
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; Department of Zoology, University of Oxford, Oxford, UK
| | - Alexandra Kupke
- Institute of Virology, Philipps University of Marburg, Marburg, Germany; German Center for Infection Research, TTU Emerging Infections, Germany
| | - Daniel Wells
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Megan A Sloan
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Keith Grehan
- Viral Pseudotype Unit, School of Pharmacy, University of Kent, Chatham Maritime, Kent ME4 4TB, UK
| | - Nigel Temperton
- Viral Pseudotype Unit, School of Pharmacy, University of Kent, Chatham Maritime, Kent ME4 4TB, UK
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - George Warimwe
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Stephan Becker
- Institute of Virology, Philipps University of Marburg, Marburg, Germany; German Center for Infection Research, TTU Emerging Infections, Germany
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Sarah C Gilbert
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
24
|
Maunder HE, Wright J, Kolli BR, Vieira CR, Mkandawire TT, Tatoris S, Kennedy V, Iqball S, Devarajan G, Ellis S, Lad Y, Clarkson NG, Mitrophanous KA, Farley DC. Enhancing titres of therapeutic viral vectors using the transgene repression in vector production (TRiP) system. Nat Commun 2017; 8:14834. [PMID: 28345582 PMCID: PMC5378976 DOI: 10.1038/ncomms14834] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/03/2017] [Indexed: 12/28/2022] Open
Abstract
A key challenge in the field of therapeutic viral vector/vaccine manufacturing is maximizing production. For most vector platforms, the ‘benchmark' vector titres are achieved with inert reporter genes. However, expression of therapeutic transgenes can often adversely affect vector titres due to biological effects on cell metabolism and/or on the vector virion itself. Here, we exemplify the novel ‘Transgene Repression In vector Production' (TRiP) system for the production of both RNA- and DNA-based viral vectors. The TRiP system utilizes a translational block of one or more transgenes by employing the bacterial tryptophan RNA-binding attenuation protein (TRAP), which binds its target RNA sequence close to the transgene initiation codon. We report enhancement of titres of lentiviral vectors expressing Cyclo-oxygenase-2 by 600-fold, and adenoviral vectors expressing the pro-apoptotic gene Bax by >150,000-fold. The TRiP system is transgene-independent and will be a particularly useful platform in the clinical development of viral vectors expressing problematic transgenes. The maximum titre of therapeutic viral vectors can be adversely affected by the encoded transgene. Here the authors repress transgene expression in producing cells by employing the tryptophan RNA-binding attenuation protein and show that it improves titre of RNA- and DNA-based viral vectors expressing toxic transgenes.
Collapse
Affiliation(s)
- H E Maunder
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - J Wright
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - B R Kolli
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - C R Vieira
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - T T Mkandawire
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - S Tatoris
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - V Kennedy
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - S Iqball
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - G Devarajan
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - S Ellis
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - Y Lad
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - N G Clarkson
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - K A Mitrophanous
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| | - D C Farley
- Research Department, Oxford BioMedica Ltd., Windrush Court, Transport Way, Oxford OX4 6LT, UK
| |
Collapse
|
25
|
Novel adenovirus encoded virus-like particles displaying the placental malaria associated VAR2CSA antigen. Vaccine 2017; 35:1140-1147. [PMID: 28131394 DOI: 10.1016/j.vaccine.2017.01.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/04/2017] [Accepted: 01/09/2017] [Indexed: 12/13/2022]
Abstract
The malaria parasite Plasmodium falciparum presents antigens on the infected erythrocyte surface that bind human receptors expressed on the vascular endothelium. The VAR2CSA mediated binding to a distinct chondroitin sulphate A (CSA) is a crucial step in the pathophysiology of placental malaria and the CSA binding region of VAR2CSA has been identified as a promising vaccine target against placental malaria. Here we designed adenovirus encoded virus-like particles (VLP) by co-encoding Simian Immunodeficiency Virus (SIV) gag and VAR2CSA. The VAR2CSA antigen was fused to the transmembrane (TM) and cytoplasmic tail (CT) domains of either the envelope protein of mouse mammary tumour virus (MMTV) or the hemagglutinin (HA) of influenza A. For a non-VLP incorporation control, a third design was made where VAR2CSA was expressed without TM-CT domains. In the primary immunogenicity study in Balb/c mice, VAR2CSA fused to HA TM-CT was significantly superior in inducing ID1-ID2a specific antibodies after the first immunization. A sequential study was performed to include a comparison to the soluble VAR2CSA protein vaccine, which has entered a phase I clinical trial (NCT02647489). The results revealed the induction of higher antibody responses and increased inhibition of parasite binding to CSA using either VAR2CSA HA TM-CT or VAR2CSA MMTV TM-CT as priming vaccines for protein double-boost immunizations, compared to protein prime-double boost regimen. Analysis of pooled serum samples on peptide arrays revealed a unique targeting of several epitopes in mice that had been primed with VAR2CSA HA TM-CT. Consequently, modification of VLP anchors is an important point of optimization in virus-encoded retroviral VLP-based vaccines, and adenovirus VLPs boosted by recombinant proteins offer hope of increasing the levels of protective VAR2CSA specific antibodies.
Collapse
|
26
|
Andersson AMC, Ragonnaud E, Seaton KE, Sawant S, Folgori A, Colloca S, Labranche C, Montefiori DC, Tomaras GD, Holst PJ. Effect of HIV-1 envelope cytoplasmic tail on adenovirus primed virus encoded virus-like particle immunizations. Vaccine 2016; 34:5344-5351. [PMID: 27633665 DOI: 10.1016/j.vaccine.2016.08.089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 08/25/2016] [Accepted: 08/28/2016] [Indexed: 11/27/2022]
Abstract
The low number of envelope (Env) spikes presented on native HIV-1 particles is a major impediment for HIV-1 prophylactic vaccine development. We designed virus-like particle encoding adenoviral vectors utilizing SIVmac239 Gag as an anchor for full length and truncated HIV-1 M consensus Env. Truncated Env overexpressed VRC01 and 17b binding antigen on the surface of transduced cells while the full length Env vaccine presented more and similar amounts of antigen binding to the trimer conformation sensitive antibodies PGT151 and PGT145, respectively. The adenoviral vectors were used to prime Balb/c mice followed by sequential boosting with chimpanzee type 63, and chimpanzee type 3 adenoviral vectors encoding SIVmac239 Gag and full length consensus Env. Both vaccine regimens induced increasing titers of binding antibody responses after each immunization, and significant differences in immune responses between the two groups were observed after the final immunization. Full length Env priming skewed antibody responses towards gp41, while truncated Env priming induced responses primarily targeting gp120 containing and derived antigens. Importantly, no differences in neutralizing antibody responses were found between the different priming regimens as both induced high titered tier 1 neutralizing antibodies, but no tier 2 antibodies, possibly reflecting the similar presentation of trimer specific antibody epitopes. The described vaccine regimens provide insight into the effects of the HIV-1 Env cytoplasmic tail on epitope presentation and subsequent immune responses, which is relevant for the interpretation of current clinical trials that are using truncated Env as an immunogen. The regimens described here provide similar neutralization titers, and thus are useful for investigating the importance of specificity in non-neutralizing antibody mediated protection against viral challenge.
Collapse
Affiliation(s)
- Anne-Marie C Andersson
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| | - Emeline Ragonnaud
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | | | | | | - Peter J Holst
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
27
|
Ewer KJ, Sierra-Davidson K, Salman AM, Illingworth JJ, Draper SJ, Biswas S, Hill AVS. Progress with viral vectored malaria vaccines: A multi-stage approach involving "unnatural immunity". Vaccine 2015; 33:7444-51. [PMID: 26476366 PMCID: PMC4687526 DOI: 10.1016/j.vaccine.2015.09.094] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 08/17/2015] [Accepted: 09/22/2015] [Indexed: 12/20/2022]
Abstract
Viral vectors used in heterologous prime-boost regimens are one of very few vaccination approaches that have yielded significant protection against controlled human malaria infections. Recently, protection induced by chimpanzee adenovirus priming and modified vaccinia Ankara boosting using the ME-TRAP insert has been correlated with the induction of potent CD8(+) T cell responses. This regimen has progressed to field studies where efficacy against infection has now been reported. The same vectors have been used pre-clinically to identify preferred protective antigens for use in vaccines against the pre-erythrocytic, blood-stage and mosquito stages of malaria and this work is reviewed here for the first time. Such antigen screening has led to the prioritization of the PfRH5 blood-stage antigen, which showed efficacy against heterologous strain challenge in non-human primates, and vectors encoding this antigen are in clinical trials. This, along with the high transmission-blocking activity of some sexual-stage antigens, illustrates well the capacity of such vectors to induce high titre protective antibodies in addition to potent T cell responses. All of the protective responses induced by these vectors exceed the levels of the same immune responses induced by natural exposure supporting the view that, for subunit vaccines to achieve even partial efficacy in humans, "unnatural immunity" comprising immune responses of very high magnitude will need to be induced.
Collapse
Affiliation(s)
- Katie J Ewer
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK.
| | - Kailan Sierra-Davidson
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; Vaccine Research Center (VRC), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Ahmed M Salman
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | | | - Simon J Draper
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Sumi Biswas
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
28
|
Comparative assessment of vaccine vectors encoding ten malaria antigens identifies two protective liver-stage candidates. Sci Rep 2015; 5:11820. [PMID: 26139288 PMCID: PMC4490344 DOI: 10.1038/srep11820] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/01/2015] [Indexed: 12/15/2022] Open
Abstract
The development of an efficacious Plasmodium falciparum malaria vaccine remains a top priority for global health. Vaccination with irradiated sporozoites is able to provide complete sterile protection through the action of CD8+ T cells at the liver-stage of infection. However, this method is currently unsuitable for large-scale deployment and focus has instead turned to the development of sub-unit vaccines. Sub-unit vaccine efforts have traditionally focused on two well-known pre-erythrocytic antigens, CSP and TRAP, yet thousands of genes are expressed in the liver-stage. We sought to assess the ability of eight alternative P. falciparum pre-erythrocytic antigens to induce a high proportion of CD8+ T cells. We show that all antigens, when expressed individually in the non-replicating viral vectors ChAd63 and MVA, are capable of inducing an immune response in mice. Furthermore, we also developed chimeric P. berghei parasites expressing the cognate P. falciparum antigen to enable assessment of efficacy in mice. Our preliminary results indicate that vectors encoding either PfLSA1 or PfLSAP2 are capable of inducing sterile protection dependent on the presence of CD8+ T cells. This work has identified two promising P. falciparum liver-stage candidate antigens that will now undergo further testing in humans.
Collapse
|
29
|
Longley RJ, Bauza K, Ewer KJ, Hill AVS, Spencer AJ. Development of an in vitro assay and demonstration of Plasmodium berghei liver-stage inhibition by TRAP-specific CD8+ T cells. PLoS One 2015; 10:e0119880. [PMID: 25822951 PMCID: PMC4379172 DOI: 10.1371/journal.pone.0119880] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 02/03/2015] [Indexed: 01/20/2023] Open
Abstract
The development of an efficacious vaccine against the Plasmodium parasite remains a top priority. Previous research has demonstrated the ability of a prime-boost virally vectored sub-unit vaccination regimen, delivering the liver-stage expressed malaria antigen TRAP, to produce high levels of antigen-specific T cells. The liver-stage of malaria is the main target of T cell-mediated immunity, yet a major challenge in assessing new T cell inducing vaccines has been the lack of a suitable pre-clinical assay. We have developed a flow-cytometry based in vitro T cell killing assay using a mouse hepatoma cell line, Hepa1-6, and Plasmodium berghei GFP expressing sporozoites. Using this assay, P. berghei TRAP-specific CD8+ T cell enriched splenocytes were shown to inhibit liver-stage parasites in an effector-to-target ratio dependent manner. Further development of this assay using human hepatocytes and P. falciparum would provide a new method to pre-clinically screen vaccine candidates and to elucidate mechanisms of protection in vitro.
Collapse
Affiliation(s)
- Rhea J Longley
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Karolis Bauza
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Katie J Ewer
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
30
|
Hodgson SH, Ewer KJ, Bliss CM, Edwards NJ, Rampling T, Anagnostou NA, de Barra E, Havelock T, Bowyer G, Poulton ID, de Cassan S, Longley R, Illingworth JJ, Douglas AD, Mange PB, Collins KA, Roberts R, Gerry S, Berrie E, Moyle S, Colloca S, Cortese R, Sinden RE, Gilbert SC, Bejon P, Lawrie AM, Nicosia A, Faust SN, Hill AVS. Evaluation of the efficacy of ChAd63-MVA vectored vaccines expressing circumsporozoite protein and ME-TRAP against controlled human malaria infection in malaria-naive individuals. J Infect Dis 2014; 211:1076-86. [PMID: 25336730 PMCID: PMC4354983 DOI: 10.1093/infdis/jiu579] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background. Circumsporozoite protein (CS) is the antigenic target for RTS,S, the most advanced malaria vaccine to date. Heterologous prime-boost with the viral vectors simian adenovirus 63 (ChAd63)-modified vaccinia virus Ankara (MVA) is the most potent inducer of T-cells in humans, demonstrating significant efficacy when expressing the preerythrocytic antigen insert multiple epitope–thrombospondin-related adhesion protein (ME-TRAP). We hypothesized that ChAd63-MVA containing CS may result in a significant clinical protective efficacy. Methods. We conducted an open-label, 2-site, partially randomized Plasmodium falciparum sporozoite controlled human malaria infection (CHMI) study to compare the clinical efficacy of ChAd63-MVA CS with ChAd63-MVA ME-TRAP. Results. One of 15 vaccinees (7%) receiving ChAd63-MVA CS and 2 of 15 (13%) receiving ChAd63-MVA ME-TRAP achieved sterile protection after CHMI. Three of 15 vaccinees (20%) receiving ChAd63-MVA CS and 5 of 15 (33%) receiving ChAd63-MVA ME-TRAP demonstrated a delay in time to treatment, compared with unvaccinated controls. In quantitative polymerase chain reaction analyses, ChAd63-MVA CS was estimated to reduce the liver parasite burden by 69%–79%, compared with 79%–84% for ChAd63-MVA ME-TRAP. Conclusions. ChAd63-MVA CS does reduce the liver parasite burden, but ChAd63-MVA ME-TRAP remains the most promising antigenic insert for a vectored liver-stage vaccine. Detailed analyses of parasite kinetics may allow detection of smaller but biologically important differences in vaccine efficacy that can influence future vaccine development. Clinical Trials Registration. NCT01623557.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eoghan de Barra
- Jenner Institute Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Tom Havelock
- NIHR Wellcome Trust Clinical Research Facility, University of Southampton and University Hospital Southampton NHS Foundation Trust
| | | | | | | | | | | | | | | | | | | | | | - Eleanor Berrie
- Clinical Biomanufacturing Facility, University of Oxford
| | - Sarah Moyle
- Clinical Biomanufacturing Facility, University of Oxford
| | | | | | - Robert E Sinden
- Jenner Institute Division of Cell and Molecular Biology, Imperial College London, United Kingdom
| | | | - Philip Bejon
- Centre for Geographical Medical Research (Coast), Kenya Medical Research Institute-Wellcome Trust, Kilifi
| | | | - Alfredo Nicosia
- Okairos, Rome CEINGE Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Italy
| | - Saul N Faust
- NIHR Wellcome Trust Clinical Research Facility, University of Southampton and University Hospital Southampton NHS Foundation Trust
| | | |
Collapse
|
31
|
Spencer AJ, Furze J, Honeycutt JD, Calvert A, Saurya S, Colloca S, Wyllie DH, Gilbert SC, Bregu M, Cottingham MG, Hill AVS. 4-1BBL enhances CD8+ T cell responses induced by vectored vaccines in mice but fails to improve immunogenicity in rhesus macaques. PLoS One 2014; 9:e105520. [PMID: 25140889 PMCID: PMC4139357 DOI: 10.1371/journal.pone.0105520] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 07/21/2014] [Indexed: 01/28/2023] Open
Abstract
T cells play a central role in the immune response to many of the world's major infectious diseases. In this study we investigated the tumour necrosis factor receptor superfamily costimulatory molecule, 4-1BBL (CD137L, TNFSF9), for its ability to increase T cell immunogenicity induced by a variety of recombinant vectored vaccines. To efficiently test this hypothesis, we assessed a number of promoters and developed a stable bi-cistronic vector expressing both the antigen and adjuvant. Co-expression of 4-1BBL, together with our model antigen TIP, was shown to increase the frequency of murine antigen-specific IFN-γ secreting CD8(+) T cells in three vector platforms examined. Enhancement of the response was not limited by co-expression with the antigen, as an increase in CD8(+) immunogenicity was also observed by co-administration of two vectors each expressing only the antigen or adjuvant. However, when this regimen was tested in non-human primates using a clinical malaria vaccine candidate, no adjuvant effect of 4-1BBL was observed limiting its potential use as a single adjuvant for translation into a clinical vaccine.
Collapse
Affiliation(s)
| | - Julie Furze
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Alice Calvert
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Saroj Saurya
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - David H. Wyllie
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Sarah C. Gilbert
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Migena Bregu
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | |
Collapse
|
32
|
Combined adenovirus vector and hepatitis C virus envelope protein prime-boost regimen elicits T cell and neutralizing antibody immune responses. J Virol 2014; 88:5502-10. [PMID: 24599994 DOI: 10.1128/jvi.03574-13] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Despite the recent progress in the development of new antiviral agents, hepatitis C virus (HCV) infection remains a major global health problem, and there is a need for a preventive vaccine. We previously reported that adenoviral vectors expressing HCV nonstructural proteins elicit protective T cell responses in chimpanzees and were immunogenic in healthy volunteers. Furthermore, recombinant HCV E1E2 protein formulated with adjuvant MF59 induced protective antibody responses in chimpanzees and was immunogenic in humans. To develop an HCV vaccine capable of inducing both T cell and antibody responses, we constructed adenoviral vectors expressing full-length and truncated E1E2 envelope glycoproteins from HCV genotype 1b. Heterologous prime-boost immunization regimens with adenovirus and recombinant E1E2 glycoprotein (genotype 1a) plus MF59 were evaluated in mice and guinea pigs. Adenovirus prime and protein boost induced broad HCV-specific CD8+ and CD4+ T cell responses and functional Th1-type IgG responses. Immune sera neutralized luciferase reporter pseudoparticles expressing HCV envelope glycoproteins (HCVpp) and a diverse panel of recombinant cell culture-derived HCV (HCVcc) strains and limited cell-to-cell HCV transmission. This study demonstrated that combining adenovirus vector with protein antigen can induce strong antibody and T cell responses that surpass immune responses achieved by either vaccine alone. IMPORTANCE HCV infection is a major health problem. Despite the availability of new directly acting antiviral agents for treating chronic infection, an affordable preventive vaccine provides the best long-term goal for controlling the global epidemic. This report describes a new anti-HCV vaccine targeting the envelope viral proteins based on adenovirus vector and protein in adjuvant. Rodents primed with the adenovirus vaccine and boosted with the adjuvanted protein developed cross-neutralizing antibodies and potent T cell responses that surpassed immune responses achieved with either vaccine component alone. If combined with the adenovirus vaccine targeting the HCV NS antigens now under clinical testing, this new vaccine might lead to a stronger and broader immune response and to a more effective vaccine to prevent HCV infection. Importantly, the described approach represents a valuable strategy for other infectious diseases in which both T and B cell responses are essential for protection.
Collapse
|
33
|
Cottingham MG, Carroll MW. Recombinant MVA vaccines: dispelling the myths. Vaccine 2013; 31:4247-51. [PMID: 23523407 DOI: 10.1016/j.vaccine.2013.03.021] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 03/11/2013] [Indexed: 12/22/2022]
Abstract
Diseases such as HIV/AIDS, tuberculosis, malaria and cancer are prime targets for prophylactic or therapeutic vaccination, but have proven partially or wholly resistant to traditional approaches to vaccine design. New vaccines based on recombinant viral vectors expressing a foreign antigen are under intense development for these and other indications. One of the most advanced and most promising vectors is the attenuated, non-replicating poxvirus MVA (modified vaccinia virus Ankara), a safer derivative of the uniquely successful smallpox vaccine. Despite the ability of recombinant MVA to induce potent humoral and cellular immune responses against transgenic antigen in humans, especially when used as the latter element of a heterologous prime-boost regimen, doubts are occasionally expressed about the ultimate feasibility of this approach. In this review, five common misconceptions over recombinant MVA are discussed, and evidence is cited to show that recombinant MVA is at least sufficiently genetically stable, manufacturable, safe, and immunogenic (even in the face of prior anti-vector immunity) to warrant reasonable hope over the feasibility of large-scale deployment, should useful levels of protection against target pathogens, or therapeutic benefit for cancer, be demonstrated in efficacy trials.
Collapse
Affiliation(s)
- Matthew G Cottingham
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, OX3 7DQ, UK.
| | | |
Collapse
|
34
|
Goodman AL, Forbes EK, Williams AR, Douglas AD, de Cassan SC, Bauza K, Biswas S, Dicks MDJ, Llewellyn D, Moore AC, Janse CJ, Franke-Fayard BM, Gilbert SC, Hill AVS, Pleass RJ, Draper SJ. The utility of Plasmodium berghei as a rodent model for anti-merozoite malaria vaccine assessment. Sci Rep 2013; 3:1706. [PMID: 23609325 PMCID: PMC3632886 DOI: 10.1038/srep01706] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 04/08/2013] [Indexed: 12/17/2022] Open
Abstract
Rodent malaria species Plasmodium yoelii and P. chabaudi have been widely used to validate vaccine approaches targeting blood-stage merozoite antigens. However, increasing data suggest the P. berghei rodent malaria may be able to circumvent vaccine-induced anti-merozoite responses. Here we confirm a failure to protect against P. berghei, despite successful antibody induction against leading merozoite antigens using protein-in-adjuvant or viral vectored vaccine delivery. No subunit vaccine approach showed efficacy in mice following immunization and challenge with the wild-type P. berghei strains ANKA or NK65, or against a chimeric parasite line encoding a merozoite antigen from P. falciparum. Protection was not improved in knockout mice lacking the inhibitory Fc receptor CD32b, nor against a Δsmac P. berghei parasite line with a non-sequestering phenotype. An improved understanding of the mechanisms responsible for protection, or failure of protection, against P. berghei merozoites could guide the development of an efficacious vaccine against P. falciparum.
Collapse
Affiliation(s)
- Anna L Goodman
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Dicks MDJ, Spencer AJ, Edwards NJ, Wadell G, Bojang K, Gilbert SC, Hill AVS, Cottingham MG. A novel chimpanzee adenovirus vector with low human seroprevalence: improved systems for vector derivation and comparative immunogenicity. PLoS One 2012; 7:e40385. [PMID: 22808149 PMCID: PMC3396660 DOI: 10.1371/journal.pone.0040385] [Citation(s) in RCA: 265] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 06/05/2012] [Indexed: 12/23/2022] Open
Abstract
Recombinant adenoviruses are among the most promising tools for vaccine antigen delivery. Recently, the development of new vectors has focused on serotypes to which the human population is less exposed in order to circumvent pre-existing anti vector immunity. This study describes the derivation of a new vaccine vector based on a chimpanzee adenovirus, Y25, together with a comparative assessment of its potential to elicit transgene product specific immune responses in mice. The vector was constructed in a bacterial artificial chromosome to facilitate genetic manipulation of genomic clones. In order to conduct a fair head-to-head immunological comparison of multiple adenoviral vectors, we optimised a method for accurate determination of infectious titre, since this parameter exhibits profound natural variability and can confound immunogenicity studies when doses are based on viral particle estimation. Cellular immunogenicity of recombinant E1 E3-deleted vector ChAdY25 was comparable to that of other species E derived chimpanzee adenovirus vectors including ChAd63, the first simian adenovirus vector to enter clinical trials in humans. Furthermore, the prevalence of virus neutralizing antibodies (titre >1:200) against ChAdY25 in serum samples collected from two human populations in the UK and Gambia was particularly low compared to published data for other chimpanzee adenoviruses. These findings support the continued development of new chimpanzee adenovirus vectors, including ChAdY25, for clinical use.
Collapse
Affiliation(s)
| | | | - Nick J. Edwards
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Göran Wadell
- Department of Clinical Microbiology, University of Umeå, Umeå, Sweden
| | - Kalifa Bojang
- Medical Research Council Laboratories, Fajara, The Gambia
| | - Sarah C. Gilbert
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | | |
Collapse
|