78 results on '"Dowall S"'
Search Results
2. Longitudinal blood cell transcriptomic profiling and in vitro temporal proteomics provides novel insights into metabolic reprogramming and host-immune responses against Crimean-Congo Hemorrhagic Fever Virus
- Author
-
Friedemann Weber, Binnur Bagci, Soham Gupta, Vegvari A, Banerjea A, Emma Kennedy, Anoop T. Ambikan, Sofia Appelberg, Dowall S, Roger Hewson, Monteil, Akusjärvi Ss, Rodriguez J, Ali Mirazimi, Neogi U, and Nazif Elaldi
- Subjects
Transcriptome ,Blood cell ,Immune system ,medicine.anatomical_structure ,Host (biology) ,Metabolic reprogramming ,medicine ,Biology ,Proteomics ,Virology ,Crimean Congo hemorrhagic fever virus ,In vitro - Abstract
The pathogenesis and host-viral interactions of the Crimean–Congo hemorrhagic fever orthonairovirus (CCHFV) are convoluted and is not well evaluated. To understand the host immune responses against CCHFV, we have performed a global transcriptomic analysis of peripheral blood mononuclear cells from a longitudinal cohort of CCHF patients who survived the infection and temporal untargeted proteomics analysis of CCHFV infected Huh7 cells. Our results indicate that during the acute phase of CCHFV infection, the host's metabolic reprogramming towards central carbon metabolism including glycolysis and glutaminolysis occurs that favours the virus replication as blocking these pathways in vitro inhibits CCHFV cellular replication. Furthermore, CCHFV replication was inhibited by blocking Akt with MK-2206 suggesting a regulatory role of PI3K/Akt/mTOR pathways. We also show activation of key interferon stimulating genes during infection, suggesting a role for type I and II interferon-mediated antiviral mechanisms. Targeting immune-metabolic pathways could be attractive therapeutic intervention for CCHFV.
- Published
- 2021
- Full Text
- View/download PDF
3. Prophylactic intranasal administration of a TLR2/6 agonist reduces upper respiratory tract viral shedding in a SARS-CoV-2 challenge ferret model
- Author
-
Proud, PC, Tsitoura, D, Watson, RJ, Chua, BY, Aram, MJ, Bewley, KR, Cavell, BE, Cobb, R, Dowall, S, Fotheringham, SA, Ho, CMK, Lucas, V, Ngabo, D, Rayner, E, Ryan, KA, Slack, GS, Thomas, S, Wand, N, Yeates, P, Demaison, C, Zeng, W, Holmes, I, Jackson, DC, Bartlett, NW, Mercuri, F, Carroll, MW, Proud, PC, Tsitoura, D, Watson, RJ, Chua, BY, Aram, MJ, Bewley, KR, Cavell, BE, Cobb, R, Dowall, S, Fotheringham, SA, Ho, CMK, Lucas, V, Ngabo, D, Rayner, E, Ryan, KA, Slack, GS, Thomas, S, Wand, N, Yeates, P, Demaison, C, Zeng, W, Holmes, I, Jackson, DC, Bartlett, NW, Mercuri, F, and Carroll, MW
- Abstract
BACKGROUND: The novel human coronavirus SARS-CoV-2 is a major ongoing global threat with huge economic burden. Like all respiratory viruses, SARS-CoV-2 initiates infection in the upper respiratory tract (URT). Infected individuals are often asymptomatic, yet highly infectious and readily transmit virus. A therapy that restricts initial replication in the URT has the potential to prevent progression of severe lower respiratory tract disease as well as limiting person-to-person transmission. METHODS: SARS-CoV-2 Victoria/01/2020 was passaged in Vero/hSLAM cells and virus titre determined by plaque assay. Challenge virus was delivered by intranasal instillation to female ferrets at 5.0 × 106 pfu/ml. Treatment groups received intranasal INNA-051, developed by Ena Respiratory. SARS-CoV-2 RNA was detected using the 2019-nCoV CDC RUO Kit and QuantStudio™ 7 Flex Real-Time PCR System. Histopathological analysis was performed using cut tissues stained with haematoxylin and eosin (H&E). FINDINGS: We show that prophylactic intra-nasal administration of the TLR2/6 agonist INNA-051 in a SARS-CoV-2 ferret infection model effectively reduces levels of viral RNA in the nose and throat. After 5 days post-exposure to SARS-CoV-2, INNA-051 significantly reduced virus in throat swabs (p=<0.0001) by up to a 24 fold (96% reduction) and in nasal wash (p=0.0107) up to a 15 fold (93% reduction) in comparison to untreated animals. INTERPRETATION: The results of our study support clinical development of a therapy based on prophylactic TLR2/6 innate immune activation in the URT, to reduce SARS-CoV-2 transmission and provide protection against COVID-19. FUNDING: This work was funded by Ena Respiratory, Melbourne, Australia.
- Published
- 2021
4. Emerging viruses and current strategies for vaccine intervention
- Author
-
Afrough, B, primary, Dowall, S, additional, and Hewson, R, additional
- Published
- 2019
- Full Text
- View/download PDF
5. Severe undifferentiated febrile illness outbreaks in the Federal Republic of Sudan – A retrospective epidemiological and diagnostic study
- Author
-
Bower, H., primary, Fletcher, T.E., additional, Mohamed, R., additional, Alzain, M., additional, Elhalawi, A., additional, Osman, A., additional, Semper, A., additional, Brooks, T., additional, Osborne, J., additional, Furneaux, J., additional, Dowall, S., additional, Graham, V., additional, Slack, G., additional, Hewson, R., additional, Beeching, N., additional, Whitworth, J., additional, Bausch, D., additional, and Mustafa, M., additional
- Published
- 2019
- Full Text
- View/download PDF
6. West Nile virus in Tunisia, 2014: First isolation from mosquitoes
- Author
-
Wasfi, F., Dachraoui, K., Cherni, S., Bosworth, A., Barhoumi, W., Dowall, S., Chelbi, I., Derbali, M., Zoghlami, Z., Beier, J.C., and Zhioua, E.
- Published
- 2016
- Full Text
- View/download PDF
7. Development of a primate model for tuberculosis vaccine research: IFN-gamma responses induced by BCG vaccination and M-tuberculosis infection in macaques
- Author
-
Dowall, S, Sharpe, S, Mcshane, H, Pathan, A, Williams, A, Dennis, M, and Marsh, P
- Published
- 2016
8. Minimal in vivo efficacy of iminosugars in a lethal ebola virus guinea pig model
- Author
-
Miller, J L, Spiro, S G, Dowall, S D, Taylor, I, Rule, A, Alonzi, D S, Sayce, A C, Wright, E, Bentley, E M, Thom, R, Hall, G, Dwek, R A, Hewson, R, and Zitzmann, N
- Subjects
Science ,Medicine - Abstract
The antiviral properties of iminosugars have been reported previously in vitro and in small animal models against Ebola virus (EBOV); however, their effects have not been tested in larger animal models such as guinea pigs. We tested the iminosugars N-butyl-deoxynojirimycin (NB-DNJ) and N-(9- methoxynonyl)-1deoxynojirimycin (MON-DNJ) for safety in uninfected animals, and for antiviral efficacy in animals infected with a lethal dose of guinea pig adapted EBOV. 1850 mg/kg/day NB-DNJ and 120 mg/kg/day MON-DNJ administered intravenously, three times daily, caused no adverse effects and were well tolerated. A pilot study treating infected animals three times within an 8 hour period was promising with 1 of 4 infected NB-DNJ treated animals surviving and the remaining three showing improved clinical signs. MON-DNJ showed no protective effects when EBOV-infected guinea pigs were treated. On histopathological examination, animals treated with NB-DNJ had reduced lesion severity in liver and spleen. However, a second study, in which NB-DNJ was administered at equally- spaced 8 hour intervals, could not confirm drug-associated benefits. Neither was any antiviral effect of iminosugars detected in an EBOV glycoprotein pseudotyped virus assay. Overall, this study provides evidence that NB-DNJ and MON-DNJ do not protect guinea pigs from a lethal EBOV-infection at the dose levels and regimens tested. However, the one surviving animal and signs of improvements in three animals of the NB-DNJ treated cohort could indicate that NB-DNJ at these levels may have a marginal beneficial effect. Future work could be focused on the development of more potent iminosugars
- Published
- 2016
9. Specificity, cross-reactivity, and function of antibodies elicited by Zika virus infection
- Author
-
Stettler, K, Beltramello, M, Espinosa, DA, Graham, V, Cassotta, A, Bianchi, S, Vanzetta, F, Minola, A, Jaconi, S, Mele, F, Foglierini, M, Pedotti, M, Simonelli, L, Dowall, S, Atkinson, B, Percivalle, E, Simmons, CP, Varani, L, Blum, J, Baldanti, F, Cameroni, E, Hewson, R, Harris, E, Lanzavecchia, A, Sallusto, F, Corti, D, Stettler, K, Beltramello, M, Espinosa, DA, Graham, V, Cassotta, A, Bianchi, S, Vanzetta, F, Minola, A, Jaconi, S, Mele, F, Foglierini, M, Pedotti, M, Simonelli, L, Dowall, S, Atkinson, B, Percivalle, E, Simmons, CP, Varani, L, Blum, J, Baldanti, F, Cameroni, E, Hewson, R, Harris, E, Lanzavecchia, A, Sallusto, F, and Corti, D
- Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus with homology to Dengue virus (DENV), has become a public health emergency. By characterizing memory lymphocytes from ZIKV-infected patients, we dissected ZIKV-specific and DENV-cross-reactive immune responses. Antibodies to nonstructural protein 1 (NS1) were largely ZIKV-specific and were used to develop a serological diagnostic tool. In contrast, antibodies against E protein domain I/II (EDI/II) were cross-reactive and, although poorly neutralizing, potently enhanced ZIKV and DENV infection in vitro and lethally enhanced DENV disease in mice. Memory T cells against NS1 or E proteins were poorly cross-reactive, even in donors preexposed to DENV. The most potent neutralizing antibodies were ZIKV-specific and targeted EDIII or quaternary epitopes on infectious virus. An EDIII-specific antibody protected mice from lethal ZIKV infection, illustrating the potential for antibody-based therapy.
- Published
- 2016
10. Effective Binding of a Phosphatidylserine-Targeting Antibody to Ebola Virus Infected Cells and Purified Virions
- Author
-
Dowall, S. D., primary, Graham, V. A., additional, Corbin-Lickfett, K., additional, Empig, C., additional, Schlunegger, K., additional, Bruce, C. B., additional, Easterbrook, L., additional, and Hewson, R., additional
- Published
- 2015
- Full Text
- View/download PDF
11. Non-fatal case of Crimean-Congo haemorrhagic fever imported into the United Kingdom (ex Bulgaria), June 2014
- Author
-
Lumley, S, primary, Atkinson, B, additional, Dowall, S D, additional, Pitman, J K, additional, Staplehurst, S, additional, Busuttil, J, additional, Simpson, A J, additional, Aarons, E J, additional, Petridou, C, additional, Nijjar, M, additional, Glover, S, additional, Brooks, T J, additional, and Hewson, R, additional
- Published
- 2014
- Full Text
- View/download PDF
12. Bunyaviruses in human, animal and mosquito samples from southeast Austria
- Author
-
Huemer, HP, primary, Seidel, B, additional, Hufnagl, P, additional, Deutz, A, additional, Posautz, A, additional, Dowall, S, additional, Hewson, R, additional, Hubalek, Z, additional, and Allerberger, F, additional
- Published
- 2014
- Full Text
- View/download PDF
13. Establishment of an Aerosol Challenge Model of Tuberculosis in Rhesus Macaques and an Evaluation of Endpoints for Vaccine Testing
- Author
-
Sharpe, S. A., primary, McShane, H., additional, Dennis, M. J., additional, Basaraba, R. J., additional, Gleeson, F., additional, Hall, G., additional, McIntyre, A., additional, Gooch, K., additional, Clark, S., additional, Beveridge, N. E. R., additional, Nuth, E., additional, White, A., additional, Marriott, A., additional, Dowall, S., additional, Hill, A. V. S., additional, Williams, A., additional, and Marsh, P. D., additional
- Published
- 2010
- Full Text
- View/download PDF
14. Absence of Crimean-Congo haemorrhagic fever virus in the tick Hyalomma aegyptium parasitizing the spur-thighed tortoise (Testudo graeca) in Tunisia
- Author
-
Fares Wasfi, Dachraoui Khalil, Najjar Chawki, Younsi Hend, Findlay-Wilson Stephen, Petretto Marie, Dowall Stuart, Hewson Roger, and Zhioua Elyes
- Subjects
Crimean-Congo haemorrhagic fever virus ,Hyalomma aegyptium ,Testudo graeca ,North Africa ,Infectious and parasitic diseases ,RC109-216 - Abstract
Free-ranging spur-thighed tortoises Testudo graeca, captured in different habitat types of Northern Tunisia from March to April 2017, were examined for tick infestation: 134/147 (91%) were infested. The overall infestation intensity and abundance was 8.5 and 7.8, respectively. From these tortoises, 1174 ticks were collected, of which 10% (n = 120) taken from 18 randomly-selected tortoises were identified at the species level; the remaining ticks were examined for the presence of Crimean-Congo haemorrhagic fever virus (CCHFv) by real time RT-PCR. Only adult Hyalomma aegyptium were found, suggesting a high degree of host specificity to tortoises. No CCHFv was detected in ticks. Considering the absence of CCHFv in Hyalomma aegyptium infesting its main host, the spur-thighed tortoise, this tick species is unlikely to play a major role in the epidemiology of CCHF. Therefore, more studies are needed to investigate the circulation of this arbovirus between livestock and other tick species from North Africa.
- Published
- 2019
- Full Text
- View/download PDF
15. "Correct to a T"
- Author
-
Dowall, S. S. M., primary
- Published
- 1909
- Full Text
- View/download PDF
16. Sero-epidemiological survey of Crimean-Congo hemorrhagic fever virus in Tunisia
- Author
-
Wasfi Fares, Dowall Stuart, Ghabbari Tayssir, Bosworth Andrew, Chakroun Mohamed, Varghese Anitha, Tiouiri Hanene, Ben Jemaa Mounir, Znazen Abir, Hewson Roger, Zhioua Elyes, and Letaief Amel
- Subjects
CCHF ,CCHF virus ,Seroprevalence ,Ticks ,Tunisia ,Infectious and parasitic diseases ,RC109-216 - Abstract
Crimean-Congo hemorrhagic fever (CCHF) is a tick-borne disease associated with a high case fatality rate and transmitted mainly by Hyalomma marginatum. The geographical distribution of H. marginatum covers most of the Western Mediterranean basin. We aimed to investigate whether CCHF virus (CCHFv) is circulating in Tunisia. Samples from unexplained acute febrile patients (n = 181) and a high risk group of humans, mainly slaughter workers (n = 38), were collected in the summer of 2014 and analyzed for exposure to CCHFv using serological tests and real-time RT-PCR. Ticks were collected from Northern and Southern Tunisia during May–June 2014 and examined for the presence of CCHFv by real-time RT-PCR. Of the 181 febrile patients, 5 showed only high titers of IgM suggesting a recent exposure to CCHFv. Among 38 slaughter workers, 2 had IgG anti-CCHFv responses yielding a seroprevalence of 5.2%. No CCHFv was detected in ticks and sera. Our results provide evidence of human exposure to CCHFv in Tunisia.
- Published
- 2016
- Full Text
- View/download PDF
17. Heterologous prime-boost-boost immunisation of Chinese cynomolgus macaques using DNA and recombinant poxvirus vectors expressing HIV-1 virus-like particles
- Author
-
Anson Donald S, Getty Brian, Dowall Stuart D, Dennis Mike J, Sharpe Sally A, Bridge Simon H, Skinner Michael A, Stewart James P, and Blanchard Tom J
- Subjects
Prime-boost HIV vaccine ,broadly reactive neutralising antibodies ,recombinant poxvirus ,modified vaccinia virus Ankara ,fowlpox virus ,cholera toxin B ,human complement protein C3d ,virus-like particle ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Background There is renewed interest in the development of poxvirus vector-based HIV vaccines due to the protective effect observed with repeated recombinant canarypox priming with gp120 boosting in the recent Thai placebo-controlled trial. This study sought to investigate whether a heterologous prime-boost-boost vaccine regimen in Chinese cynomolgus macaques with a DNA vaccine and recombinant poxviral vectors expressing HIV virus-like particles bearing envelopes derived from the most prevalent clades circulating in sub-Saharan Africa, focused the antibody response to shared neutralising epitopes. Methods Three Chinese cynomolgus macaques were immunised via intramuscular injections using a regimen composed of a prime with two DNA vaccines expressing clade A Env/clade B Gag followed by boosting with recombinant fowlpox virus expressing HIV-1 clade D Gag, Env and cholera toxin B subunit followed by the final boost with recombinant modified vaccinia virus Ankara expressing HIV-1 clade C Env, Gag and human complement protein C3d. We measured the macaque serum antibody responses by ELISA, enumerated T cell responses by IFN-γ ELISpot and assessed seroneutralisation of HIV-1 using the TZM-bl β-galactosidase assay with primary isolates of HIV-1. Results This study shows that large and complex synthetic DNA sequences can be successfully cloned in a single step into two poxvirus vectors: MVA and FPV and the recombinant poxviruses could be grown to high titres. The vaccine candidates showed appropriate expression of recombinant proteins with the formation of authentic HIV virus-like particles seen on transmission electron microscopy. In addition the b12 epitope was shown to be held in common by the vaccine candidates using confocal immunofluorescent microscopy. The vaccine candidates were safely administered to Chinese cynomolgus macaques which elicited modest T cell responses at the end of the study but only one out of the three macaques elicited an HIV-specific antibody response. However, the antibodies did not neutralise primary isolates of HIV-1 or the V3-sensitive isolate SF162 using the TZM-bl β-galactosidase assay. Conclusions MVA and FP9 are ideal replication-deficient viral vectors for HIV-1 vaccines due to their excellent safety profile for use in humans. This study shows this novel prime-boost-boost regimen was poorly immunogenic in Chinese cynomolgus macaques.
- Published
- 2011
- Full Text
- View/download PDF
18. A Review of Nonhuman Primate Models of Rift Valley Fever Virus Infection: Progress, Challenge Strains, and Future Directions.
- Author
-
Ebisine K, Quist D, Findlay-Wilson S, Kennedy E, and Dowall S
- Abstract
Rift Valley fever (RVF) is a mosquito-borne viral disease that primarily affects animals, especially ruminants, but has the capacity to infect humans and result in outbreaks. Infection with the causative agent, RVF virus (RVFV), causes severe disease in domestic animals, especially sheep, resulting in fever, anorexia, immobility, abortion, and high morbidity and mortality rates in neonate animals. Humans become infected through exposure to infected animals and, less frequently, directly via a mosquito bite. A greater awareness of RVFV and its epidemic potential has resulted in increased investment in the development of interventions, especially vaccines. There is currently no substitute for the use of animal models in order to evaluate these vaccines. As outbreaks of RVF disease are difficult to predict or model, conducting Phase III clinical trials will likely not be feasible. Therefore, representative animal model systems are essential for establishing efficacy data to support licensure. Nonhuman primate (NHP) species are often chosen due to their closeness to humans, reflecting similar susceptibility and disease kinetics. This review covers the use of NHP models in RVFV research, with much of the work having been conducted in rhesus macaques and common marmosets. The future direction of RVF work conducted in NHP is discussed in anticipation of the importance of it being a key element in the development and approval of a human vaccine.
- Published
- 2024
- Full Text
- View/download PDF
19. Recombinant Vaccine Production: Production of a Recombinant CCHF MVA Vaccine.
- Author
-
Kennedy E, Hewson R, and Dowall S
- Subjects
- Humans, Vaccinia virus genetics, Vaccinia virus immunology, Hemorrhagic Fever Virus, Crimean-Congo immunology, Hemorrhagic Fever Virus, Crimean-Congo genetics, Vaccines, Synthetic immunology, Vaccines, Synthetic genetics, Vaccines, DNA immunology, Vaccines, DNA genetics, Animals, Genetic Vectors genetics, Hemorrhagic Fever, Crimean prevention & control, Hemorrhagic Fever, Crimean immunology, Viral Vaccines immunology, Viral Vaccines genetics
- Abstract
One of the key interventions against infection is immunization, including an increasing focus on development of vaccines against pathogenic bunyaviruses. Whilst different vaccine development approaches exist, recombinant viral vaccines have a strong safety record, are rapid to produce, are cost-effective, and have been demonstrated to be rolled out in response to outbreaks, including in low- and middle-income countries. One viral vector, modified Vaccinia Ankara (MVA), has been used to develop vaccine candidates against Crimean-Congo Haemorrhagic Fever (CCHF) virus through incorporation of the nucleoprotein (NP) and glycoprotein (GP) regions, with the former candidate having now progressed to being the first vaccine against CCHF virus to enter Phase 1 clinical trials. Herein, we report the method used to generate this MVA-based vaccine construct., (© 2025. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2025
- Full Text
- View/download PDF
20. Comparison of Chikungunya Virus-Induced Disease Progression and Pathogenesis in Type-I Interferon Receptor-Deficient Mice (A129) and Two Wild-Type (129Sv/Ev and C57BL/6) Mouse Strains.
- Author
-
Graham VA, Easterbrook L, Rayner E, Findlay-Wilson S, Flett L, Kennedy E, Fotheringham S, Kempster S, Almond N, and Dowall S
- Subjects
- Animals, Mice, Female, Mice, Knockout, Chikungunya Fever virology, Chikungunya Fever immunology, Chikungunya Fever pathology, Chikungunya virus genetics, Chikungunya virus pathogenicity, Chikungunya virus immunology, Mice, Inbred C57BL, Viral Load, Disease Models, Animal, Receptor, Interferon alpha-beta deficiency, Receptor, Interferon alpha-beta genetics, Disease Progression
- Abstract
Chikungunya virus (CHIKV) is a mosquito-borne alphavirus causing a debilitating febrile illness with rheumatic disease symptoms of arthralgia and arthritis. Since its spread outside of Africa in 2005, it continues to cause outbreaks and disseminates into new territories. Intervention strategies are urgently required, including vaccination and antiviral approaches. To test efficacy, the use of small animal models is required. Two mouse strains, A129, with a deficiency in their type-I interferon (IFN) receptor, and C57BL/6 are widely used. A direct comparison of these strains alongside the wild-type parental strain of the A129 mice, 129Sv/Ev, was undertaken to assess clinical disease progression, viral loads in key tissues, histological changes and levels of sera biomarkers. Our results confirm the severe disease course in A129 mice which was not observed in the parental 129Sv/Ev strain. Of the two wild-type strains, viral loads were higher in 129Sv/Ev mice compared to C57BL/6 counterparts. Our results have established these models and parameters for the future testing of vaccines and antiviral approaches., Competing Interests: The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.
- Published
- 2024
- Full Text
- View/download PDF
21. Convalescent human plasma candidate reference materials protect against Crimean-Congo haemorrhagic fever virus (CCHFV) challenge in an A129 mouse model.
- Author
-
Kempster S, Hassall M, Graham V, Kennedy E, Findlay-Wilson S, Salguero FJ, Bagci B, Elaldi N, Oz M, Tasseten T, Charlton FW, Barr JN, Fontana J, Duru C, Ezeajughi E, Matejtschuk P, Arnold U, Adedeji Y, Mirazimi A, Hewson R, Dowall S, and Almond N
- Subjects
- Animals, Mice, Humans, Female, Neutralization Tests, Plasma immunology, Male, Hemorrhagic Fever, Crimean immunology, Hemorrhagic Fever, Crimean prevention & control, Hemorrhagic Fever Virus, Crimean-Congo immunology, Antibodies, Viral blood, Antibodies, Viral immunology, Disease Models, Animal, Antibodies, Neutralizing blood, Antibodies, Neutralizing immunology
- Abstract
Crimean-Congo Haemorrhagic Fever Virus (CCHFV) is spread by infected ticks or direct contact with blood, tissues and fluids from infected patients or livestock. Infection with CCHFV causes severe haemorrhagic fever in humans which is fatal in up to 83 % of cases. CCHFV is listed as a priority pathogen by the World Health Organization (WHO) and there are currently no widely-approved vaccines. Defining a serological correlate of protection against CCHFV infection would support the development of vaccines by providing a 'target threshold' for pre-clinical and clinical immunogenicity studies to achieve in subjects and potentially obviate the need for in vivo protection studies. We therefore sought to establish titratable protection against CCHFV using pooled human convalescent plasma, in a mouse model. Convalescent plasma collected from seven individuals with a known previous CCHFV virus infection were characterised using binding antibody and neutralisation assays. All plasma recognised nucleoprotein and the Gc glycoprotein, but some had a lower Gn glycoprotein response by ELISA. Pooled plasma and two individual donations from convalescent donors were administered intraperitoneally to A129 mice 24 h prior to intradermal challenge with CCHFV (strain IbAr10200). A partial protective effect was observed with all three convalescent plasmas characterised by longer survival post-challenge and reduced clinical score. These protective responses were titratable. Further characterisation of the serological reactivities within these samples will establish their value as reference materials to support assay harmonisation and accelerate vaccine development for CCHFV., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Crown Copyright © 2024. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
22. Potent immunogenicity and protective efficacy of a multi-pathogen vaccination targeting Ebola, Sudan, Marburg and Lassa viruse.
- Author
-
Flaxman A, Sebastian S, Appelberg S, Cha KM, Ulaszewska M, Purushotham J, Gilbride C, Sharpe H, Spencer AJ, Bibi S, Wright D, Schmidt I, Dowall S, Easterbrook L, Findlay-Wilson S, Gilbert S, Mirazimi A, and Lambe T
- Subjects
- Animals, Mice, Viral Vaccines immunology, Humans, Vaccination, Female, Antibodies, Viral immunology, Immunogenicity, Vaccine, Ebola Vaccines immunology, Ebolavirus immunology, Lassa virus immunology, Marburgvirus immunology, Hemorrhagic Fever, Ebola prevention & control, Hemorrhagic Fever, Ebola immunology, Lassa Fever immunology, Lassa Fever prevention & control, Marburg Virus Disease immunology, Marburg Virus Disease prevention & control
- Abstract
Viral haemorrhagic fevers (VHF) pose a significant threat to human health. In recent years, VHF outbreaks caused by Ebola, Marburg and Lassa viruses have caused substantial morbidity and mortality in West and Central Africa. In 2022, an Ebola disease outbreak in Uganda caused by Sudan virus resulted in 164 cases with 55 deaths. In 2023, a Marburg disease outbreak was confirmed in Equatorial Guinea and Tanzania resulting in over 49 confirmed or suspected cases; 41 of which were fatal. There are no clearly defined correlates of protection against these VHF, impeding targeted vaccine development. Any vaccine developed should therefore induce strong and preferably long-lasting humoral and cellular immunity against these viruses. Ideally this immunity should also cross-protect against viral variants, which are known to circulate in animal reservoirs and cause human disease. We have utilized two viral vectored vaccine platforms, an adenovirus (ChAdOx1) and Modified Vaccinia Ankara (MVA), to develop a multi-pathogen vaccine regime against three filoviruses (Ebola virus, Sudan virus, Marburg virus) and an arenavirus (Lassa virus). These platform technologies have consistently demonstrated the capability to induce robust cellular and humoral antigen-specific immunity in humans, most recently in the rollout of the licensed ChAdOx1-nCoV19/AZD1222. Here, we show that our multi-pathogen vaccines elicit strong cellular and humoral immunity, induce a diverse range of chemokines and cytokines, and most importantly, confers protection after lethal Ebola virus, Sudan virus and Marburg virus challenges in a small animal model., Competing Interests: TL reports consulting fees from Vaccitech on an unrelated project, an honorarium from Seqirus, grant support from the DHSC for this work, and is named as an inventor on a patent application for a vaccine against SARS-CoV-2. SG is named as an inventor on patents covering use of ChAdOx1-vectored vaccines and a vaccine against SARS-CoV-2., (Copyright: © 2024 Flaxman et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
23. Third International Conference on Crimean-Congo Hemorrhagic Fever in Thessaloniki, Greece, September 19-21, 2023.
- Author
-
Welch SR, Garrison AR, Bente DA, Burt F, D'Addiego J, Devignot S, Dowall S, Fischer K, Hawman DW, Hewson R, Mirazimi A, Oestereich L, Vatansever Z, Spengler JR, and Papa A
- Subjects
- Animals, Humans, Greece, Disease Outbreaks, Hemorrhagic Fever, Crimean epidemiology, Hemorrhagic Fever Virus, Crimean-Congo, Ticks
- Abstract
The Third International Conference on Crimean-Congo Hemorrhagic Fever (CCHF) was held in Thessaloniki, Greece, September 19-21, 2023, bringing together a diverse group of international partners, including public health professionals, clinicians, ecologists, epidemiologists, immunologists, and virologists. The conference was attended by 118 participants representing 24 countries and the World Health Organization (WHO). Meeting sessions covered the epidemiology of CCHF in humans; Crimean-Congo hemorrhagic fever virus (CCHFV) in ticks; wild and domestic animal hosts; molecular virology; pathogenesis and animal models; immune response related to therapeutics; and CCHF prevention in humans. The concluding session focused on recent WHO recommendations regarding disease prevention, control strategies, and innovations against CCHFV outbreaks. This meeting report summarizes lectures by the invited speakers and highlights advances in the field., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
24. Seroprevalence and Risk Factors Associated with Phleboviruses and Crimean-Congo Hemorrhagic Fever Virus among Blood Donors in Central Tunisia.
- Author
-
Ayari R, Chaouch H, Findlay-Wilson S, Hachfi W, Ben Lasfar N, Bellazreg F, Dowall S, Hannachi N, and Letaief A
- Abstract
The aim of this study was to determine the prevalence of six viruses, from two families of the order Bunyavirales , in the general population of central Tunisia. Sera collected from 377 asymptomatic blood donors were serologically assayed for Rift Valley fever virus (RVFV), Crimean-Congo hemorrhagic fever virus (CCHFV), and four sandfly-borne phleboviruses: Toscana virus (TOSV), sandfly fever Naples virus (SFNV), sandfly fever Sicilian virus (SFSV), and sandfly fever Cyprus virus (SFCV). Of the 377 subjects enrolled in this study, 17.3% were IgG positive for at least one of the viruses tested. The most frequently detected antibodies were against TOSV (13.3%), followed by SFCV (2.9%), RVFV (1.9%), SFSV (1.3%), and SFNV (1.1%). Only one sample was IgG positive for CCHFV. Dual reactivity was observed in nine cases: SFSV + SFCV in three cases (0.8%) and TOSV + SFNV, TOSV + SFCV, and TOSV + RVFV in two cases (0.5%) each. 15.9% of donors were IgG positive against sandfly-borne phleboviruses. Among the 65 donors IgG positive for phleboviruses, 50.8% were from rural areas compared to 12.3% from urban areas ( p < 0.001); 92.3% had animals in their living quarters ( p = 0.009); and 70.8% lived in the vicinity of stagnant water ( p = 0.062). Seroprevalence was significantly higher among donors living with chronic diseases ( p = 0.039). Furthermore, the seroprevalence of phleboviruses was higher in Kairouan, the central governorate, than in the two coastal governorates: Monastir and Sousse, with 33.4%, 24.2%, and 14.9%, respectively. The presence of antibodies in the general population needs further investigation to better assess the extent of these viruses. Only TOSV was known to have an extensive circulation in Tunisia and in North Africa. Continued surveillance and interventions are necessary to detect the emergence of all arboviruses and to prevent further transmission., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2024
- Full Text
- View/download PDF
25. Pathogenesis of Rift Valley Fever Virus in a BALB/c Mouse Model Is Affected by Virus Culture Conditions and Sex of the Animals.
- Author
-
Graham VA, Easterbrook L, Kennedy E, Rayner E, Findlay-Wilson S, Flett L, Wise EL, Treagus S, Fotheringham S, Kempster S, Almond N, and Dowall S
- Subjects
- Male, Female, Humans, Animals, Mice, Mice, Inbred BALB C, Reproducibility of Results, Disease Progression, Mammals, Rift Valley fever virus, Rift Valley Fever
- Abstract
Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic pathogen causing disease in livestock and humans. Whilst initially restricted to the African continent, recent spread to the Arabian Peninsula has highlighted the likelihood of entry into new regions. Due to the absence of a regulatory-approved human vaccine, work is ongoing to develop and assess countermeasures. As such, small animal models play a pivotal role in providing information on disease pathogenesis and elucidating which intervention strategies confer protection. To develop and establish the BALB/c mouse model, we challenged mice with RVFV grown from two separate cell lines: one derived from mosquitoes (C6/36) and the other mammalian derived (Vero E6). Following infection, we assessed the clinical course of disease progression at days 1 and 3 post-challenge and evaluated viral tropism and immune analytes. The results demonstrated that RVFV infection was affected by the cell line used to propagate the challenge virus, with those grown in insect cells resulting in a more rapid disease progression. The lowest dose that caused uniform severe disease remained the same across both virus preparations. In addition, to demonstrate reproducibility, the lowest dose was used for a subsequent infection study using male and female animals. The results further demonstrated that male mice succumbed to infection more rapidly than their female counterparts. Our results establish an RVFV mouse model and key parameters that affect the course of disease progression in BALB/c mice.
- Published
- 2023
- Full Text
- View/download PDF
26. Publisher Correction: Refinement of an ovine-based immunoglobulin therapy against SARS-CoV-2, with comparison of whole IgG versus F(ab') 2 fragments.
- Author
-
Findlay-Wilson S, Easterbrook L, Smith S, Pope N, Aldridge M, Humphries G, Schuhmann H, Ngabo D, Rayner E, Otter A, Coleman T, Hicks B, Halkerston R, Apostolakis K, Taylor S, Fotheringham S, Horton A, CanoCejas I, Wand M, Tree JA, Sutton M, Graham V, Hewson R, and Dowall S
- Published
- 2023
- Full Text
- View/download PDF
27. Refinement of an ovine-based immunoglobulin therapy against SARS-CoV-2, with comparison of whole IgG versus F(ab') 2 fragments.
- Author
-
Findlay-Wilson S, Easterbrook L, Smith S, Pope N, Aldridge M, Humphries G, Schuhmann H, Ngabo D, Rayner E, Otter A, Coleman T, Hicks B, Halkerston R, Apostolakis K, Taylor S, Fotheringham S, Horton A, CanoCejas I, Wand M, Tree JA, Sutton M, Graham V, Hewson R, and Dowall S
- Subjects
- Cricetinae, Animals, Sheep, Immunization, Passive, Kinetics, Immunoglobulin G, SARS-CoV-2, COVID-19
- Abstract
The development of new therapies against SARS-CoV-2 is required to extend the toolkit of intervention strategies to combat the global pandemic. In this study, hyperimmune plasma from sheep immunised with whole spike SARS-CoV-2 recombinant protein has been used to generate candidate products. In addition to purified IgG, we have refined candidate therapies by removing non-specific IgG via affinity binding along with fragmentation to eliminate the Fc region to create F(ab')
2 fragments. These preparations were evaluated for in vitro activity and demonstrated to be strongly neutralising against a range of SARS-CoV-2 strains, including Omicron B2.2. In addition, their protection against disease manifestations and viral loads were assessed using a hamster SARS-CoV-2 infection model. Results demonstrated protective effects of both IgG and F(ab')2 , with the latter requiring sequential dosing to maintain in vivo activity due to rapid clearance from the circulation., (© 2023. Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
28. Establishment of a Nipah Virus Disease Model in Hamsters, including a Comparison of Intranasal and Intraperitoneal Routes of Challenge.
- Author
-
Findlay-Wilson S, Flett L, Salguero FJ, Ruedas-Torres I, Fotheringham S, Easterbrook L, Graham V, and Dowall S
- Abstract
Nipah virus (NiV) is an emerging pathogen that can cause severe respiratory illness and encephalitis in humans. The main reservoir is fruit bats, distributed across a large geographical area that includes Australia, Southeast Asia, and Africa. Incursion into humans is widely reported through exposure of infected pigs, ingestion of contaminated food, or through contact with an infected person. With no approved treatments or vaccines, NiV poses a threat to human public health and has epidemic potential. To aid with the assessment of emerging interventions being developed, an expansion of preclinical testing capability is required. Given variations in the model parameters observed in different sites during establishment, optimisation of challenge routes and doses is required. Upon evaluating the hamster model, an intranasal route of challenge was compared with intraperitoneal delivery, demonstrating a more rapid dissemination to wider tissues in the latter. A dose effect was observed between those causing respiratory illness and those resulting in neurological disease. The data demonstrate the successful establishment of the hamster model of NiV disease for subsequent use in the evaluation of vaccines and antivirals.
- Published
- 2023
- Full Text
- View/download PDF
29. Adenoviral vectored vaccination protects against Crimean-Congo Haemorrhagic Fever disease in a lethal challenge model.
- Author
-
Saunders JE, Gilbride C, Dowall S, Morris S, Ulaszewska M, Spencer AJ, Rayner E, Graham VA, Kennedy E, Thomas K, Hewson R, Gilbert SC, Belij-Rammerstorfer S, and Lambe T
- Subjects
- Humans, Animals, Mice, Vaccination, Genetic Vectors genetics, Vaccinia virus, Hemorrhagic Fever, Crimean prevention & control, Hemorrhagic Fever Virus, Crimean-Congo genetics, Viral Vaccines
- Abstract
Background: The tick-borne bunyavirus, Crimean-Congo Haemorrhagic Fever virus (CCHFV), can cause severe febrile illness in humans and has a wide geographic range that continues to expand due to tick migration. Currently, there are no licensed vaccines against CCHFV for widespread usage., Methods: In this study, we describe the preclinical assessment of a chimpanzee adenoviral vectored vaccine (ChAdOx2 CCHF) which encodes the glycoprotein precursor (GPC) from CCHFV., Findings: We demonstrate here that vaccination with ChAdOx2 CCHF induces both a humoral and cellular immune response in mice and 100% protection in a lethal CCHF challenge model. Delivery of the adenoviral vaccine in a heterologous vaccine regimen with a Modified Vaccinia Ankara vaccine (MVA CCHF) induces the highest levels of CCHFV-specific cell-mediated and antibody responses in mice. Histopathological examination and viral load analysis of the tissues of ChAdOx2 CCHF immunised mice reveals an absence of both microscopic changes and viral antigen associated with CCHF infection, further demonstrating protection against disease., Interpretation: There is the continued need for an effective vaccine against CCHFV to protect humans from lethal haemorrhagic disease. Our findings support further development of the ChAd platform expressing the CCHFV GPC to seek an effective vaccine against CCHFV., Funding: This research was supported by funding from the Biotechnology and Biological Sciences Research Council (UKRI-BBSRC) [BB/R019991/1 and BB/T008784/1]., Competing Interests: Declaration of interests SCG is co-founder and board member of Vaccitech and named as an inventor on a patent covering use of ChAdOx2-vectored vaccines. TL was consultant to Vaccitech. The remaining authors declare no competing interests., (Copyright © 2023 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
30. Exploring the Potential of Iminosugars as Antivirals for Crimean-Congo Haemorrhagic Fever Virus, Using the Surrogate Hazara Virus: Liquid-Chromatography-Based Mapping of Viral N-Glycosylation and In Vitro Antiviral Assays.
- Author
-
Tyrrell BE, Kumar A, Gangadharan B, Alonzi D, Brun J, Hill M, Bharucha T, Bosworth A, Graham V, Dowall S, Miller JL, and Zitzmann N
- Abstract
Crimean-Congo haemorrhagic fever virus (CCHFV) is a pathogen of increasing public health concern, being a widely distributed arbovirus and the causative agent of the potentially fatal Crimean-Congo haemorrhagic fever. Hazara virus (HAZV) is a genetically and serologically related virus that has been proposed as a surrogate for antiviral and vaccine testing for CCHFV. Glycosylation analysis of HAZV has been limited; first, we confirmed for the first time the occupation of two N-glycosylation sites in the HAZV glycoprotein. Despite this, there was no apparent antiviral efficacy of a panel of iminosugars against HAZV, as determined by quantification of the total secretion and infectious virus titres produced following infection of SW13 and Vero cells. This lack of efficacy was not due to an inability of deoxynojirimycin (DNJ)-derivative iminosugars to access and inhibit endoplasmic reticulum α-glucosidases, as demonstrated by free oligosaccharide analysis in uninfected and infected SW13 and uninfected Vero cells. Even so, iminosugars may yet have potential as antivirals for CCHFV since the positions and importance of N-linked glycans may differ between the viruses, a hypothesis requiring further evaluation.
- Published
- 2023
- Full Text
- View/download PDF
31. Screening of wild deer populations for exposure to SARS-CoV-2 in the United Kingdom, 2020-2021.
- Author
-
Holding M, Otter AD, Dowall S, Takumi K, Hicks B, Coleman T, Hemingway G, Royds M, Findlay-Wilson S, Curran-French M, Vipond R, Sprong H, and Hewson R
- Subjects
- Animals, Animals, Wild, Antibodies, Viral, COVID-19 Testing veterinary, Humans, SARS-CoV-2, Spike Glycoprotein, Coronavirus, COVID-19 epidemiology, COVID-19 veterinary, Deer
- Abstract
Following findings in Northern America of SARS-CoV-2 infections in white-tailed deer, there is concern of similar infections in European deer and their potential as reservoirs of SARS-CoV-2 including opportunities for the emergence of new variants. UK deer sera were collected in 2020-2021 from 6 species and a hybrid with 1748 tested using anti-spike and anti-nucleocapsid serology assays. No samples were positive on both assays nor by surrogate neutralization testing. There is no evidence that spill-over infections of SARS-CoV-2 occurred from the human population to UK deer or that SARS-CoV-2 has been circulating in UK deer (over the study period). Although it cannot be ruled out, study results indicate that spill-over infections followed by circulation of SARS-CoV-2 to the most common European deer species is small., (© 2022 Crown copyright. Transboundary and Emerging Diseases published by Wiley-VCH GmbH. This article is published with the permission of the Controller of HMSO and the Queen's Printer for Scotland.)
- Published
- 2022
- Full Text
- View/download PDF
32. Activity of a Carbohydrate-Binding Module Therapy, Neumifil, against SARS-CoV-2 Disease in a Hamster Model of Infection.
- Author
-
Fell R, Potter JA, Yuille S, Salguero FJ, Watson R, Ngabo D, Gooch K, Hewson R, Howat D, and Dowall S
- Subjects
- Carbohydrates, Cricetinae, Humans, SARS-CoV-2, Spike Glycoprotein, Coronavirus, Peptidyl-Dipeptidase A metabolism, COVID-19 Drug Treatment
- Abstract
The rapid global spread of severe acute respiratory coronavirus 2 (SARS-CoV-2) has resulted in an urgent effort to find efficacious therapeutics. Broad-spectrum therapies which could be used for other respiratory pathogens confer advantages, as do those based on targeting host cells that are not prone to the development of resistance by the pathogen. We tested an intranasally delivered carbohydrate-binding module (CBM) therapy, termed Neumifil, which is based on a CBM that has previously been shown to offer protection against the influenza virus through the binding of sialic acid receptors. Using the recognised hamster model of SARS-CoV-2 infection, we demonstrate that Neumifil significantly reduces clinical disease severity and pathological changes in the nasal cavity. Furthermore, we demonstrate Neumifil binding to the human angiotensin-converting enzyme 2 (ACE2) receptor and spike protein of SARS-CoV-2. This is the first report describing the testing of this type of broad-spectrum antiviral therapy in vivo and provides evidence for the advancement of Neumifil in further preclinical and clinical studies.
- Published
- 2022
- Full Text
- View/download PDF
33. Multi-omics insights into host-viral response and pathogenesis in Crimean-Congo hemorrhagic fever viruses for novel therapeutic target.
- Author
-
Neogi U, Elaldi N, Appelberg S, Ambikan A, Kennedy E, Dowall S, Bagci BK, Gupta S, Rodriguez JE, Svensson-Akusjärvi S, Monteil V, Vegvari A, Benfeitas R, Banerjea A, Weber F, Hewson R, and Mirazimi A
- Subjects
- Antiviral Agents therapeutic use, Cross-Sectional Studies, Humans, Interferons, Leukocytes, Mononuclear, Hemorrhagic Fever Virus, Crimean-Congo genetics, Hemorrhagic Fever, Crimean
- Abstract
The pathogenesis and host-viral interactions of the Crimean-Congo hemorrhagic fever orthonairovirus (CCHFV) are convoluted and not well evaluated. Application of the multi-omics system biology approaches, including biological network analysis in elucidating the complex host-viral response, interrogates the viral pathogenesis. The present study aimed to fingerprint the system-level alterations during acute CCHFV-infection and the cellular immune responses during productive CCHFV-replication in vitro. We used system-wide network-based system biology analysis of peripheral blood mononuclear cells (PBMCs) from a longitudinal cohort of CCHF patients during the acute phase of infection and after one year of recovery (convalescent phase) followed by untargeted quantitative proteomics analysis of the most permissive CCHFV-infected Huh7 and SW13 cells. In the RNAseq analysis of the PBMCs, comparing the acute and convalescent-phase, we observed system-level host's metabolic reprogramming towards central carbon and energy metabolism (CCEM) with distinct upregulation of oxidative phosphorylation (OXPHOS) during CCHFV-infection. Upon application of network-based system biology methods, negative coordination of the biological signaling systems like FOXO/Notch axis and Akt/mTOR/HIF-1 signaling with metabolic pathways during CCHFV-infection were observed. The temporal quantitative proteomics in Huh7 showed a dynamic change in the CCEM over time and concordant with the cross-sectional proteomics in SW13 cells. By blocking the two key CCEM pathways, glycolysis and glutaminolysis, viral replication was inhibited in vitro. Activation of key interferon stimulating genes during infection suggested the role of type I and II interferon-mediated antiviral mechanisms both at the system level and during progressive replication., Competing Interests: UN, NE, SA, AA, EK, SD, BB, SG, JR, SS, VM, AV, RB, AB, FW, RH, AM No competing interests declared, (© 2022, Neogi et al.)
- Published
- 2022
- Full Text
- View/download PDF
34. Development of a Hamster Natural Transmission Model of SARS-CoV-2 Infection.
- Author
-
Dowall S, Salguero FJ, Wiblin N, Fotheringham S, Hatch G, Parks S, Gowan K, Harris D, Carnell O, Fell R, Watson R, Graham V, Gooch K, Hall Y, Mizen S, and Hewson R
- Subjects
- Animals, COVID-19 pathology, COVID-19 virology, Cricetinae, Female, Lung pathology, Male, Nasal Cavity pathology, Viral Load, Virus Shedding, COVID-19 transmission, Disease Models, Animal, Mesocricetus, SARS-CoV-2 physiology
- Abstract
The global pandemic of coronavirus disease (COVID-19) caused by infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has led to an international thrust to study pathogenesis and evaluate interventions. Experimental infection of hamsters and the resulting respiratory disease is one of the preferred animal models since clinical signs of disease and virus shedding are similar to more severe cases of human COVID-19. The main route of challenge has been direct inoculation of the virus via the intranasal route. To resemble the natural infection, we designed a bespoke natural transmission cage system to assess whether recipient animals housed in physically separate adjacent cages could become infected from a challenged donor animal in a central cage, with equal airflow across the two side cages. To optimise viral shedding in the donor animals, a low and moderate challenge dose were compared after direct intranasal challenge, but similar viral shedding responses were observed and no discernible difference in kinetics. The results from our natural transmission set-up demonstrate that most recipient hamsters are infected within the system developed, with variation in the kinetics and levels of disease between individual animals. Common clinical outputs used for the assessment in directly-challenged hamsters, such as weight loss, are less obvious in hamsters who become infected from naturally acquiring the infection. The results demonstrate the utility of a natural transmission model for further work on assessing the differences between virus strains and evaluating interventions using a challenge system which more closely resembles human infection.
- Published
- 2021
- Full Text
- View/download PDF
35. Plant-made dengue virus-like particles produced by co-expression of structural and non-structural proteins induce a humoral immune response in mice.
- Author
-
Ponndorf D, Meshcheriakova Y, Thuenemann EC, Dobon Alonso A, Overman R, Holton N, Dowall S, Kennedy E, Stocks M, Lomonossoff GP, and Peyret H
- Subjects
- Animals, Antibodies, Neutralizing, Antibodies, Viral, Dengue Virus genetics, Mice, Mice, Inbred BALB C, Nicotiana, Dengue Vaccines, Immunity, Humoral, Vaccines, Virus-Like Particle genetics, Viral Proteins immunology
- Abstract
Dengue virus (DENV) is an emerging threat causing an estimated 390 million infections per year. Dengvaxia, the only licensed vaccine, may not be adequately safe in young and seronegative patients; hence, development of a safer, more effective vaccine is of great public health interest. Virus-like particles (VLPs) are a safe and very efficient vaccine strategy, and DENV VLPs have been produced in various expression systems. Here, we describe the production of DENV VLPs in Nicotiana benthamiana using transient expression. The co-expression of DENV structural proteins (SP) and a truncated version of the non-structural proteins (NSPs), lacking NS5 that contains the RNA-dependent RNA polymerase, led to the assembly of DENV VLPs in plants. These VLPs were comparable in appearance and size to VLPs produced in mammalian cells. Contrary to data from other expression systems, expression of the protein complex prM-E was not successful, and strategies used in other expression systems to improve the VLP yield did not result in increased yields in plants but, rather, increased purification difficulties. Immunogenicity assays in BALB/c mice revealed that plant-made DENV1-SP + NSP VLPs led to a higher antibody response in mice compared with DENV-E domain III displayed inside bluetongue virus core-like particles and a DENV-E domain III subunit. These results are consistent with the idea that VLPs could be the optimal approach to creating candidate vaccines against enveloped viruses., (© 2020 The Authors. Plant Biotechnology Journal published by Society for Experimental Biology and The Association of Applied Biologists and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
36. Prophylactic intranasal administration of a TLR2/6 agonist reduces upper respiratory tract viral shedding in a SARS-CoV-2 challenge ferret model.
- Author
-
Proud PC, Tsitoura D, Watson RJ, Chua BY, Aram MJ, Bewley KR, Cavell BE, Cobb R, Dowall S, Fotheringham SA, Ho CMK, Lucas V, Ngabo D, Rayner E, Ryan KA, Slack GS, Thomas S, Wand NI, Yeates P, Demaison C, Zeng W, Holmes I, Jackson DC, Bartlett NW, Mercuri F, and Carroll MW
- Subjects
- Administration, Intranasal, Animals, COVID-19 pathology, Disease Models, Animal, Female, Ferrets, Immunity, Innate, Lipopeptides chemistry, Lipopeptides pharmacology, Nasal Cavity pathology, Nasal Cavity virology, Pharynx pathology, Pharynx virology, RNA, Viral metabolism, Real-Time Polymerase Chain Reaction, Respiratory System pathology, SARS-CoV-2 genetics, SARS-CoV-2 isolation & purification, Viral Load drug effects, COVID-19 Drug Treatment, Lipopeptides administration & dosage, Respiratory System virology, SARS-CoV-2 pathogenicity, Toll-Like Receptor 2 agonists, Toll-Like Receptor 6 agonists, Virus Shedding
- Abstract
Background: The novel human coronavirus SARS-CoV-2 is a major ongoing global threat with huge economic burden. Like all respiratory viruses, SARS-CoV-2 initiates infection in the upper respiratory tract (URT). Infected individuals are often asymptomatic, yet highly infectious and readily transmit virus. A therapy that restricts initial replication in the URT has the potential to prevent progression of severe lower respiratory tract disease as well as limiting person-to-person transmission., Methods: SARS-CoV-2 Victoria/01/2020 was passaged in Vero/hSLAM cells and virus titre determined by plaque assay. Challenge virus was delivered by intranasal instillation to female ferrets at 5.0 × 10
6 pfu/ml. Treatment groups received intranasal INNA-051, developed by Ena Respiratory. SARS-CoV-2 RNA was detected using the 2019-nCoV CDC RUO Kit and QuantStudio™ 7 Flex Real-Time PCR System. Histopathological analysis was performed using cut tissues stained with haematoxylin and eosin (H&E)., Findings: We show that prophylactic intra-nasal administration of the TLR2/6 agonist INNA-051 in a SARS-CoV-2 ferret infection model effectively reduces levels of viral RNA in the nose and throat. After 5 days post-exposure to SARS-CoV-2, INNA-051 significantly reduced virus in throat swabs (p=<0.0001) by up to a 24 fold (96% reduction) and in nasal wash (p=0.0107) up to a 15 fold (93% reduction) in comparison to untreated animals., Interpretation: The results of our study support clinical development of a therapy based on prophylactic TLR2/6 innate immune activation in the URT, to reduce SARS-CoV-2 transmission and provide protection against COVID-19., Funding: This work was funded by Ena Respiratory, Melbourne, Australia., Competing Interests: Declaration of Competing Interests Authors report grants from Ena Respiratory, during the conduct of the study. W. Zeng and D.C. Jackson reports grants from Ena Therapeutics, during the conduct of the study. D. Tsitoura, C. Demaison, F. Mercuri, I. Holmes and N.W. Bartlett reports personal fees and other from Ena Therapeutics, outside the submitted work. D.C. Jackson, W. Zeng and B.Y. Chua reports other from Ena Therapeutics, outside the submitted work. Dr. Holmes reports personal fees from Ena Therapeutics, outside the submitted work. In addition, D. Tsitoura, C. Demaison and F. Mercuri have a patent AU 2020901709 pending to Ena Therapeutics. D.C Jackson, W. Zeng and C. Demaison have a patent PCT/AU2011/001225 issued to Ena Therapeutics. D.C Jackson, W. Zeng, I. Holmes and C. Demaison have a patent PCT/AU2020/050660 pending to Ena Therapeutics., (Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
37. X-ray inactivation of RNA viruses without loss of biological characteristics.
- Author
-
Afrough B, Eakins J, Durley-White S, Dowall S, Findlay-Wilson S, Graham V, Lewandowski K, Carter DP, and Hewson R
- Subjects
- Animals, Chlorocebus aethiops, Civil Defense, Containment of Biohazards, Feeder Cells, Humans, Monte Carlo Method, Nairovirus physiology, Nairovirus radiation effects, RNA Viruses radiation effects, RNA, Viral radiation effects, Sequence Analysis, RNA, Togaviridae physiology, Togaviridae radiation effects, Vero Cells, Viral Zoonoses prevention & control, Zika Virus physiology, Zika Virus radiation effects, RNA Viruses physiology, RNA, Viral genetics, Virus Inactivation, X-Rays adverse effects
- Abstract
In the event of an unpredictable viral outbreak requiring high/maximum biosafety containment facilities (i.e. BSL3 and BSL4), X-ray irradiation has the potential to relieve pressures on conventional diagnostic bottlenecks and expediate work at lower containment. Guided by Monte Carlo modelling and in vitro 1-log
10 decimal-reduction value (D-value) predictions, the X-ray photon energies required for the effective inactivation of zoonotic viruses belonging to the medically important families of Flaviviridae, Nairoviridae, Phenuiviridae and Togaviridae are demonstrated. Specifically, it is shown that an optimized irradiation approach is attractive for use in a multitude of downstream detection and functional assays, as it preserves key biochemical and immunological properties. This study provides evidence that X-ray irradiation can support emergency preparedness, outbreak response and front-line diagnostics in a safe, reproducible and scalable manner pertinent to operations that are otherwise restricted to higher containment BSL3 or BSL4 laboratories.- Published
- 2020
- Full Text
- View/download PDF
38. Immunogenicity and Efficacy of Zika Virus Envelope Domain III in DNA, Protein, and ChAdOx1 Adenoviral-Vectored Vaccines.
- Author
-
López-Camacho C, De Lorenzo G, Slon-Campos JL, Dowall S, Abbink P, Larocca RA, Kim YC, Poggianella M, Graham V, Findlay-Wilson S, Rayner E, Carmichael J, Dejnirattisai W, Boyd M, Hewson R, Mongkolsapaya J, Screaton GR, Barouch DH, Burrone OR, Patel AH, and Reyes-Sandoval A
- Abstract
The flavivirus envelope protein domain III (EDIII) was an effective immunogen against dengue virus (DENV) and other related flaviviruses. Whether this can be applied to the Zika virus (ZIKV) vaccinology remains an open question. Here, we tested the efficacy of ZIKV-EDIII against ZIKV infection, using several vaccine platforms that present the antigen in various ways. We provide data demonstrating that mice vaccinated with a ZIKV-EDIII as DNA or protein-based vaccines failed to raise fully neutralizing antibodies and did not control viremia, following a ZIKV challenge, despite eliciting robust antibody responses. Furthermore, we showed that ZIKV-EDIII encoded in replication-deficient Chimpanzee adenovirus (ChAdOx1-EDIII) elicited anti-ZIKV envelope antibodies in vaccinated mice but also provided limited protection against ZIKV in two physiologically different mouse challenge models. Taken together, our data indicate that contrary to what was shown for other flaviviruses like the dengue virus, which has close similarities with ZIKV-EDIII, this antigen might not be a suitable vaccine candidate for the correct induction of protective immune responses against ZIKV.
- Published
- 2020
- Full Text
- View/download PDF
39. A Multi-Filovirus Vaccine Candidate: Co-Expression of Ebola, Sudan, and Marburg Antigens in a Single Vector.
- Author
-
Sebastian S, Flaxman A, Cha KM, Ulaszewska M, Gilbride C, Sharpe H, Wright E, Spencer AJ, Dowall S, Hewson R, Gilbert S, and Lambe T
- Abstract
In the infectious diseases field, protective immunity against individual virus species or strains does not always confer cross-reactive immunity to closely related viruses, leaving individuals susceptible to disease after exposure to related virus species. This is a significant hurdle in the field of vaccine development, in which broadly protective vaccines represent an unmet need. This is particularly evident for filoviruses, as there are multiple family members that can cause lethal haemorrhagic fever, including Zaire ebolavirus, Sudan ebolavirus, and Marburg virus. In an attempt to address this need, both pre-clinical and clinical studies previously used mixed or co-administered monovalent vaccines to prevent filovirus mediated disease. However, these multi-vaccine and multi-dose vaccination regimens do not represent a practical immunisation scheme when considering the target endemic areas. We describe here the development of a single multi-pathogen filovirus vaccine candidate based on a replication-deficient simian adenoviral vector. Our vaccine candidate encodes three different filovirus glycoproteins in one vector and induces strong cellular and humoral immunity to all three viral glycoproteins after a single vaccination. Crucially, it was found to be protective in a stringent Zaire ebolavirus challenge in guinea pigs in a one-shot vaccination regimen. This trivalent filovirus vaccine offers a tenable vaccine product that could be rapidly translated to the clinic to prevent filovirus-mediated viral haemorrhagic fever.
- Published
- 2020
- Full Text
- View/download PDF
40. Detection of tick-borne encephalitis virus in the UK.
- Author
-
Holding M, Dowall S, and Hewson R
- Subjects
- Antibodies, Viral, United Kingdom, Encephalitis Viruses, Tick-Borne
- Published
- 2020
- Full Text
- View/download PDF
41. A probable case of tick-borne encephalitis (TBE) acquired in England, July 2019.
- Author
-
Kreusch TM, Holding M, Hewson R, Harder T, Medlock JM, Hansford KM, Dowall S, Semper A, Brooks T, Walsh A, Russell K, and Wichmann O
- Subjects
- Animals, England, Flavivirus Infections diagnosis, Flavivirus Infections virology, Germany, Humans, Immunoglobulin G blood, Immunoglobulin M blood, Infant, Magnetic Resonance Imaging, Meningoencephalitis diagnostic imaging, Travel, Encephalitis Viruses, Tick-Borne isolation & purification, Encephalitis, Tick-Borne diagnosis, Ixodes virology, Tick Bites
- Abstract
The United Kingdom (UK) has thus far been considered to be free from tick-borne encephalitis (TBE), yet in July 2019, a German infant developed serologically diagnosed TBE following a tick bite in southern England. This first report of a probable human case together with recent findings of TBE virus in ticks in foci in England suggest that TBE may be acquired in parts of England and should be considered in patients with aetiologically-unexplained neurological manifestations.
- Published
- 2019
- Full Text
- View/download PDF
42. High susceptibility, viral dynamics and persistence of South American Zika virus in New World monkey species.
- Author
-
Berry N, Ferguson D, Ham C, Hall J, Jenkins A, Giles E, Devshi D, Kempster S, Rose N, Dowall S, Fritzsche M, Bleazard T, Hewson R, and Almond N
- Subjects
- Animals, Callithrix virology, Disease Models, Animal, Humans, Macaca mulatta virology, Monkey Diseases pathology, Monkey Diseases virology, Platyrrhini virology, Puerto Rico epidemiology, South America epidemiology, Viremia pathology, Viremia virology, Zika Virus Infection pathology, Zika Virus Infection virology, Monkey Diseases epidemiology, Viremia epidemiology, Zika Virus pathogenicity, Zika Virus Infection epidemiology
- Abstract
South American Zika virus (ZIKV) recently emerged as a novel human pathogen, linked with neurological disorders. However, comparative ZIKV infectivity studies in New World primates are lacking. Two members of the Callitrichidae family, common marmosets (Callithrix jacchus) and red-bellied tamarins (Saguinus labiatus), were highly susceptible to sub-cutaneous challenge with the Puerto Rico-origin ZIKV
PRVABC59 strain. Both exhibited rapid, high, acute viraemia with early neuroinvasion (3 days) in peripheral and central nervous tissue. ZIKV RNA levels in blood and tissues were significantly higher in New World hosts compared to Old World species (Macaca mulatta, Macaca fascicularis). Tamarins and rhesus macaques exhibited loss of zonal occludens-1 (ZO-1) staining, indicative of a compromised blood-brain barrier 3 days post-ZIKV exposure. Early, widespread dissemination across multiple anatomical sites distant to the inoculation site preceded extensive ZIKV persistence after 100 days in New and Old World lineages, especially lymphoid, neurological and reproductive sites. Prolonged persistence in brain tissue has implications for otherwise resolved human ZIKV infection. High susceptibility of distinct New World species underscores possible establishment of ZIKV sylvatic cycles in primates indigenous to ZIKV endemic regions. Tamarins and marmosets represent viable New World models for ZIKV pathogenesis and therapeutic intervention studies, including vaccines, with contemporary strains.- Published
- 2019
- Full Text
- View/download PDF
43. A vaccine based on recombinant modified Vaccinia Ankara containing the nucleoprotein from Lassa virus protects against disease progression in a guinea pig model.
- Author
-
Kennedy EM, Dowall SD, Salguero FJ, Yeates P, Aram M, and Hewson R
- Subjects
- Animals, Cell Line, Cricetinae, Enzyme-Linked Immunosorbent Assay, Female, Guinea Pigs, Vaccination, Vaccines, Synthetic immunology, Vaccines, Synthetic therapeutic use, Vaccinia virus immunology, Vaccinia virus pathogenicity, Lassa virus immunology, Nucleoproteins immunology, Vaccinia immunology, Vaccinia prevention & control
- Abstract
Lassa fever remains the most imported viral haemorrhagic fever in Europe and is responsible for 5000 deaths per year throughout Western Africa. There is no vaccine and treatment is often ineffective. We have developed a vaccine based on modified Vaccinia Ankara expressing the nucleoprotein from Lassa virus (MVALassaNP). This study investigated the immunogenicity (in mice) and efficacy (in guinea pigs) of the MVALassaNP vaccine as a prime/boost or single vaccination regime. ELISA and ELISpot assays confirmed humoral and T-cell immunity following both a prime and prime/boost vaccination, with the prime/boost regime producing a statistically increased response compared to a prime only vaccine (P < 0.0001). The vaccine offered protection in guinea pigs against disease manifestations after challenge with virulent Lassa virus. Clinical signs, weight loss and temperature increases were observed in all animals receiving a control MVA vaccine, after challenge with Lassa virus. In contrast, no clinical signs, fever or weight loss were observed in any of the MVALassaNP vaccinated animals demonstrating that both a single immunisation, and prime/boost regime confer protection against disease progression. In conclusion, the MVALassaNP vaccine candidate elicits an immune response, demonstrates efficacy against Lassa virus disease and is suitable for further preclinical and clinical development., (Crown Copyright © 2019. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
44. Detection of Crimean-Congo Haemorrhagic Fever cases in a severe undifferentiated febrile illness outbreak in the Federal Republic of Sudan: A retrospective epidemiological and diagnostic cohort study.
- Author
-
Bower H, El Karsany M, Alzain M, Gannon B, Mohamed R, Mahmoud I, Eldegail M, Taha R, Osman A, Mohamednour S, Semper A, Atkinson B, Carter D, Dowall S, Furneaux J, Graham V, Mellors J, Osborne J, Pullan ST, Slack GS, Brooks T, Hewson R, Beeching NJ, Whitworth J, Bausch DG, and Fletcher TE
- Subjects
- Adolescent, Adult, Child, Child, Preschool, Cohort Studies, Disease Outbreaks, Female, Fever epidemiology, Fever virology, Hemorrhagic Fever, Crimean epidemiology, Hemorrhagic Fever, Crimean virology, Humans, Male, Real-Time Polymerase Chain Reaction, Retrospective Studies, Sudan epidemiology, Young Adult, Fever diagnosis, Hemorrhagic Fever, Crimean diagnosis
- Abstract
Background: Undifferentiated febrile illness (UFI) is one of the most common reasons for people seeking healthcare in low-income countries. While illness and death due to specific infections such as malaria are often well-quantified, others are frequently uncounted and their impact underappreciated. A number of high consequence infectious diseases, including Ebola virus, are endemic or epidemic in the Federal Republic of Sudan which has experienced at least 12 UFI outbreaks, frequently associated with haemorrhage and high case fatality rates (CFR), since 2012. One of these occurred in Darfur in 2015/2016 with 594 cases and 108 deaths (CFR 18.2%). The aetiology of these outbreaks remains unknown., Methodology/principal Findings: We report a retrospective cohort study of the 2015/2016 Darfur outbreak, using a subset of 65 of 263 outbreak samples received by the National Public Health Laboratory which met selection criteria of sufficient sample volume and epidemiological data. Clinical features included fever (95.8%), bleeding (95.7%), headache (51.6%) and arthralgia (42.2%). No epidemiological patterns indicative of person-to-person transmission or health-worker cases were reported. Samples were tested at the Public Health England Rare and Imported Pathogens Laboratory using a bespoke panel of likely pathogens including haemorrhagic fever viruses, arboviruses and Rickettsia, Leptospira and Borrelia spp. Seven (11%) were positive for Crimean-Congo haemorrhagic fever virus (CCHFV) by real-time reverse transcription PCR. The remaining samples tested negative on all assays., Conclusions/significance: CCHFV is an important cause of fever and haemorrhage in Darfur, but not the sole major source of UFI outbreaks in Sudan. Prospective studies are needed to explore other aetiologies, including novel pathogens. The presence of CCHFV has critical infection, prevention and control as well as clinical implications for future response. Our study reinforces the need to boost surveillance, lab and investigative capacity to underpin effective response, and for local and international health security., Competing Interests: I have read the journal's policy and the authors of this manuscript have the following competing interests: HB, BG, JW & DB are members of the UK Public Health Rapid Support Team. AS, NJB, RH, SD and TB are affiliated to the National Institute for Health Research Health Protection Research Unit (NIHR HPRU) in Emerging and Zoonotic Infections at University of Liverpool in partnership with PHE, in collaboration with the Liverpool School of Tropical Medicine. The views expressed are those of the authors and not necessarily those of the National Health Service, the National Institute for Health Research, the Department of Health and Social Care, or Public Health England.
- Published
- 2019
- Full Text
- View/download PDF
45. Therapeutic Monoclonal Antibodies for Ebola Virus Infection Derived from Vaccinated Humans.
- Author
-
Rijal P, Elias SC, Machado SR, Xiao J, Schimanski L, O'Dowd V, Baker T, Barry E, Mendelsohn SC, Cherry CJ, Jin J, Labbé GM, Donnellan FR, Rampling T, Dowall S, Rayner E, Findlay-Wilson S, Carroll M, Guo J, Xu XN, Huang KA, Takada A, Burgess G, McMillan D, Popplewell A, Lightwood DJ, Draper SJ, and Townsend AR
- Subjects
- Adolescent, Adult, Animals, Antibodies, Monoclonal blood, Antibodies, Monoclonal therapeutic use, Antibodies, Neutralizing blood, Antibodies, Neutralizing isolation & purification, Antibodies, Neutralizing therapeutic use, Antibodies, Viral blood, Antibodies, Viral isolation & purification, Antibodies, Viral therapeutic use, Cells, Cultured, Dogs, Female, Guinea Pigs, HEK293 Cells, Hemorrhagic Fever, Ebola blood, Hemorrhagic Fever, Ebola immunology, Humans, Madin Darby Canine Kidney Cells, Male, Middle Aged, Young Adult, Antibodies, Monoclonal isolation & purification, Ebola Vaccines isolation & purification, Ebola Vaccines therapeutic use, Ebolavirus immunology, Hemorrhagic Fever, Ebola therapy, Vaccination methods
- Abstract
We describe therapeutic monoclonal antibodies isolated from human volunteers vaccinated with recombinant adenovirus expressing Ebola virus glycoprotein (EBOV GP) and boosted with modified vaccinia virus Ankara. Among 82 antibodies isolated from peripheral blood B cells, almost half neutralized GP pseudotyped influenza virus. The antibody response was diverse in gene usage and epitope recognition. Although close to germline in sequence, neutralizing antibodies with binding affinities in the nano- to pico-molar range, similar to "affinity matured" antibodies from convalescent donors, were found. They recognized the mucin-like domain, glycan cap, receptor binding region, and the base of the glycoprotein. A cross-reactive cocktail of four antibodies, targeting the latter three non-overlapping epitopes, given on day 3 of EBOV infection, completely protected guinea pigs. This study highlights the value of experimental vaccine trials as a rich source of therapeutic human monoclonal antibodies., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
46. Differences in monocyte: lymphocyte ratio and Tuberculosis disease progression in genetically distinct populations of macaques.
- Author
-
Sibley L, Gooch K, Wareham A, Gray S, Chancellor A, Dowall S, Bate S, Marriott A, Dennis M, White AD, Marsh PD, Fletcher H, and Sharpe S
- Subjects
- Animals, Disease Models, Animal, Enzyme-Linked Immunosorbent Assay, Interferon-gamma biosynthesis, Transcriptome, Tuberculosis genetics, Tuberculosis pathology, Lymphocytes pathology, Macaca fascicularis genetics, Macaca mulatta genetics, Monocytes pathology, Tuberculosis immunology
- Abstract
Monocyte:lymphocyte ratio (M:L) has been identified as a risk factor in development of TB disease in children and those undergoing treatment for HIV in co-infected individuals. Retrospective analysis was performed using M:L data collected from TB modelling studies performed in Rhesus macaques of Indian genotype (RM), cynomolgus macaque of Chinese genotype (CCM) and cynomolgus macaque of Mauritian genotype (MCM), which found that the more susceptible populations (RM and MCM) had higher M:L ratios than the least susceptible population (CCM). Following Mycobacterium tuberculosis exposure, significant increases in M:L ratio were observed in susceptible RM and MCM within 12 weeks of TB infection, whereas M:L in CCM remained stable, suggesting that changes in M:L ratio may also act as a biomarker of TB disease progression. The frequency of PPD-specific interferon gamma (IFNγ) secreting cells (SFU) were compared, with the more susceptible macaque populations showing an association between M:L and IFNγ SFU frequency. Investigation of the genes associated with monocyte-derived antigen presenting cells revealed differences between RM and CCM, highlighting differences in their monocyte populations, as well as overall M:L ratio. Differences in M:L ratio between macaque populations could be used to explore immunological mechanisms in susceptible populations that would complement human population studies.
- Published
- 2019
- Full Text
- View/download PDF
47. A Human Bi-specific Antibody against Zika Virus with High Therapeutic Potential.
- Author
-
Wang J, Bardelli M, Espinosa DA, Pedotti M, Ng TS, Bianchi S, Simonelli L, Lim EXY, Foglierini M, Zatta F, Jaconi S, Beltramello M, Cameroni E, Fibriansah G, Shi J, Barca T, Pagani I, Rubio A, Broccoli V, Vicenzi E, Graham V, Pullan S, Dowall S, Hewson R, Jurt S, Zerbe O, Stettler K, Lanzavecchia A, Sallusto F, Cavalli A, Harris E, Lok SM, Varani L, and Corti D
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal chemistry, Antibodies, Neutralizing administration & dosage, Antibodies, Neutralizing chemistry, Antibodies, Viral administration & dosage, Antibodies, Viral chemistry, Cryoelectron Microscopy, Epitopes, Humans, Magnetic Resonance Spectroscopy, Mice, Models, Molecular, Sequence Alignment, Viral Envelope Proteins chemistry, Zika Virus immunology, Antibodies, Monoclonal therapeutic use, Antibodies, Neutralizing therapeutic use, Antibodies, Viral therapeutic use, Zika Virus chemistry, Zika Virus Infection therapy
- Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus, causes devastating congenital birth defects. We isolated a human monoclonal antibody (mAb), ZKA190, that potently cross-neutralizes multi-lineage ZIKV strains. ZKA190 is highly effective in vivo in preventing morbidity and mortality of ZIKV-infected mice. NMR and cryo-electron microscopy show its binding to an exposed epitope on DIII of the E protein. ZKA190 Fab binds all 180 E protein copies, altering the virus quaternary arrangement and surface curvature. However, ZIKV escape mutants emerged in vitro and in vivo in the presence of ZKA190, as well as of other neutralizing mAbs. To counter this problem, we developed a bispecific antibody (FIT-1) comprising ZKA190 and a second mAb specific for DII of E protein. In addition to retaining high in vitro and in vivo potencies, FIT-1 robustly prevented viral escape, warranting its development as a ZIKV immunotherapy., (Crown Copyright © 2017. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
48. Specificity, cross-reactivity, and function of antibodies elicited by Zika virus infection.
- Author
-
Stettler K, Beltramello M, Espinosa DA, Graham V, Cassotta A, Bianchi S, Vanzetta F, Minola A, Jaconi S, Mele F, Foglierini M, Pedotti M, Simonelli L, Dowall S, Atkinson B, Percivalle E, Simmons CP, Varani L, Blum J, Baldanti F, Cameroni E, Hewson R, Harris E, Lanzavecchia A, Sallusto F, and Corti D
- Subjects
- Animals, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal therapeutic use, Antibodies, Neutralizing chemistry, Antibodies, Neutralizing therapeutic use, Antibodies, Viral chemistry, Antibodies, Viral therapeutic use, Antibody Specificity, Cross Reactions, Dengue Virus immunology, Disease Models, Animal, Humans, Immunodominant Epitopes immunology, Immunologic Memory, Protein Structure, Tertiary, T-Lymphocytes immunology, Viral Envelope Proteins immunology, Viral Nonstructural Proteins immunology, Zika Virus Infection prevention & control, Zika Virus Infection therapy, Antibodies, Monoclonal immunology, Antibodies, Neutralizing immunology, Antibodies, Viral immunology, Zika Virus immunology, Zika Virus Infection immunology
- Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus with homology to Dengue virus (DENV), has become a public health emergency. By characterizing memory lymphocytes from ZIKV-infected patients, we dissected ZIKV-specific and DENV-cross-reactive immune responses. Antibodies to nonstructural protein 1 (NS1) were largely ZIKV-specific and were used to develop a serological diagnostic tool. In contrast, antibodies against E protein domain I/II (EDI/II) were cross-reactive and, although poorly neutralizing, potently enhanced ZIKV and DENV infection in vitro and lethally enhanced DENV disease in mice. Memory T cells against NS1 or E proteins were poorly cross-reactive, even in donors preexposed to DENV. The most potent neutralizing antibodies were ZIKV-specific and targeted EDIII or quaternary epitopes on infectious virus. An EDIII-specific antibody protected mice from lethal ZIKV infection, illustrating the potential for antibody-based therapy., (Copyright © 2016, American Association for the Advancement of Science.)
- Published
- 2016
- Full Text
- View/download PDF
49. A Crimean-Congo hemorrhagic fever (CCHF) viral vaccine expressing nucleoprotein is immunogenic but fails to confer protection against lethal disease.
- Author
-
Dowall SD, Buttigieg KR, Findlay-Wilson SJ, Rayner E, Pearson G, Miloszewska A, Graham VA, Carroll MW, and Hewson R
- Subjects
- Animals, Antibodies, Viral immunology, Cell Line, Chick Embryo, Chlorocebus aethiops, Cricetinae, Hemorrhagic Fever, Crimean immunology, Humans, Immunization, Mice, Mice, Knockout, Receptor, Interferon alpha-beta genetics, Vero Cells, Viral Load immunology, Antibodies, Viral blood, Hemorrhagic Fever Virus, Crimean-Congo immunology, Hemorrhagic Fever, Crimean prevention & control, Immunogenicity, Vaccine immunology, Nucleoproteins immunology, Vaccines, Synthetic immunology, Viral Proteins immunology, Viral Vaccines immunology
- Abstract
Crimean-Congo Hemorrhagic Fever (CCHF) is a severe tick-borne disease, endemic in many countries in Africa, the Middle East, Eastern Europe and Asia. Between 15-70% of reported cases are fatal with no approved vaccine available. In the present study, the attenuated poxvirus vector, Modified Vaccinia virus Ankara, was used to develop a recombinant candidate vaccine expressing the CCHF virus nucleoprotein. Cellular and humoral immunogenicity was confirmed in 2 mouse strains, including type I interferon receptor knockout mice, which are susceptible to CCHF disease. Despite the immune responses generated post-immunisation, the vaccine failed to protect animals from lethal disease in a challenge model.
- Published
- 2016
- Full Text
- View/download PDF
50. Serologic evidence of exposure to Rift Valley fever virus detected in Tunisia.
- Author
-
Bosworth A, Ghabbari T, Dowall S, Varghese A, Fares W, Hewson R, Zhioua E, Chakroun M, Tiouiri H, Ben Jemaa M, Znazen A, and Letaief A
- Abstract
Rift Valley fever virus (RVFv) is capable of causing dramatic outbreaks amongst economically important animal species and is capable of causing severe symptoms and mortality in humans. RVFv is known to circulate widely throughout East Africa; serologic evidence of exposure has also been found in some northern African countries, including Mauritania. This study aimed to ascertain whether RVFv is circulating in regions beyond its known geographic range. Samples from febrile patients (n = 181) and nonfebrile healthy agricultural and slaughterhouse workers (n = 38) were collected during the summer of 2014 and surveyed for exposure to RVFv by both serologic tests and PCR. Of the 219 samples tested, 7.8% of nonfebrile participants showed immunoglobulin G reactivity to RVFv nucleoprotein and 8.3% of febrile patients showed immunoglobulin M reactivity, with the latter samples indicating recent exposure to the virus. Our results suggest an active circulation of RVFv and evidence of human exposure in the population of Tunisia.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.