1
|
Modjarrad K, Scott PT, McCauley M, Ober-Shepherd B, Sondergaard E, Amare MF, Parikh AP, Omar B, Minutello AM, Adhikarla H, Wu Y, P AR, Delore V, Mantel N, Morrison MN, Kourbanova KS, Martinez ME, Guzman I, Greenleaf ME, Darden JM, Koren MA, Hamer MJ, Lee CE, Hutter JN, Peel SA, Robb ML, Vangelisti M, Feroldi E. Safety and immunogenicity of a next-generation live-attenuated yellow fever vaccine produced in a Vero cell line in the USA: a phase 1 randomised, observer-blind, active-controlled, dose-ranging clinical trial. THE LANCET. INFECTIOUS DISEASES 2024:S1473-3099(24)00406-7. [PMID: 39153488 DOI: 10.1016/s1473-3099(24)00406-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Recent outbreaks between 2015-17 and production delays have led to a yellow fever vaccine shortage. Therefore, there is an urgent need for new yellow fever vaccines with improved production scalability. A next-generation live-attenuated yellow fever vaccine candidate (vYF), produced in a Vero cell line has shown similar immunogenicity to licensed yellow fever vaccines in preclinical studies. In this study, we aimed to report the safety and immunogenicity of vYF in human clinical trial participants. METHODS In this first in-human, phase 1 randomised, observer-blind, active-controlled, dose-ranging clinical trial conducted at a single centre in the USA (Walter Reed Army Institute of Research, Silver Spring, MD, USA), 72 healthy adults (aged 18-60 years), without a known history of flavivirus infection or vaccination were randomly assigned (1:1:1:1) using interactive response technology to receive one dose of either vYF at 4, 5 or 6 Log CCID50 or the licensed YF-VAX (18 individuals per group). The primary outcomes were safety, neutralising antibody (NAb) titres through D180 post-vaccination in the per-protocol analysis set (comprised of yellow fever-naive participants who received their intended vaccine and provided a valid post-vaccination blood sample), and occurrence, and level of yellow fever viraemia in each vaccine group through D14 post-vaccination. FINDINGS All vYF doses had a safety and tolerability profile similar to YF-VAX. The most frequently reported solicited injection site reactions (vYF groups vs YF-VAX group) were pain (22% [12 of 54 participants, 95% CI 12-36] vs 28% [five of 18 participants, 10-54]), and erythema (13% [seven of 54 participants, 5-25] vs 39% [seven of 18 participants, 17-64]), with headache (32% [17 of 54 participants, 20-46] vs 44% [eight of 18 participants, 22-69]) and malaise (26% [14 of 54 participants, 15-40] vs 33% [six of 18 participants, 13-59]) as the most frequently reported solicited systemic reactions. One grade 3 solicited reaction (erythema) reported in the YF-VAX group resolved spontaneously. No serious unsolicited adverse events or deaths were reported. Viraemia was transiently detected in 50 participants between D4 and D10 in all groups and was observed in more participants or for a longer time in the vYF 6 Log CCID50 and YF-VAX groups. All yellow fever-naive vaccine recipients across the study groups seroconverted yielding four-fold increase from baseline in yellow fever NAb titres measured by yellow fever microneutralisation assay by D28 and were seroprotected with yellow fever NAb titres of at least 10 [1/dil]). Overall, 100% (18 of 18 participants, 95% CI 82-100), 89% (16 participants, 65-99), 100% (18 participants, 82-100), and 94% (17 participants, 73-100) of participants in the vYF 4 Log, vYF 5 Log, vYF 6 Log CCID50 groups, and YF-VAX group, respectively, remained seroprotected through D180. INTERPRETATION vYF has a similar safety and immunogenicity profile to YF-VAX. In general, the vYF 5 Log CCID50 dose appeared to show optimal viraemia, safety, and immunogenicity, and was chosen for subsequent development. FUNDING Sanofi.
Collapse
Affiliation(s)
- Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Paul T Scott
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Melanie McCauley
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Brittany Ober-Shepherd
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Erica Sondergaard
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Mihret F Amare
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Ajay P Parikh
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Badryah Omar
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | | | | | | | | | | | | | - Meshell N Morrison
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Kamila S Kourbanova
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Melissa E Martinez
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Ivelese Guzman
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Melissa E Greenleaf
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Janice M Darden
- Diagnostics and Countermeasures Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Michael A Koren
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Melinda J Hamer
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA; Department of Military and Emergency Medicine, Uniformed Services University, Bethesda, MD, USA; Department of Emergency Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Christine E Lee
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Jack N Hutter
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Sheila A Peel
- Diagnostics and Countermeasures Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Merlin L Robb
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | | | | |
Collapse
|
2
|
Mantel N, Piras-Douce F, Chautard E, Marcos-Lopez E, Bodinham CL, Cosma A, Courtois V, Dhooge N, Gautheron S, Kaufmann SHE, Pizzoferro K, Lewis DJM, Martinon F, Pagnon A, Raynal F, Dereuddre-Bosquet N, Le Grand R. Cynomolgus macaques as a translational model of human immune responses to yellow fever 17D vaccination. J Virol 2024; 98:e0151623. [PMID: 38567951 PMCID: PMC11092345 DOI: 10.1128/jvi.01516-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/22/2023] [Indexed: 05/15/2024] Open
Abstract
The non-human primate (NHP) model (specifically rhesus and cynomolgus macaques) has facilitated our understanding of the pathogenic mechanisms of yellow fever (YF) disease and allowed the evaluation of the safety and efficacy of YF-17D vaccines. However, the accuracy of this model in mimicking vaccine-induced immunity in humans remains to be fully determined. We used a systems biology approach to compare hematological, biochemical, transcriptomic, and innate and antibody-mediated immune responses in cynomolgus macaques and human participants following YF-17D vaccination. Immune response progression in cynomolgus macaques followed a similar course as in adult humans but with a slightly earlier onset. Yellow fever virus neutralizing antibody responses occurred earlier in cynomolgus macaques [by Day 7[(D7)], but titers > 10 were reached in both species by D14 post-vaccination and were not significantly different by D28 [plaque reduction neutralization assay (PRNT)50 titers 3.6 Log vs 3.5 Log in cynomolgus macaques and human participants, respectively; P = 0.821]. Changes in neutrophils, NK cells, monocytes, and T- and B-cell frequencies were higher in cynomolgus macaques and persisted for 4 weeks versus less than 2 weeks in humans. Low levels of systemic inflammatory cytokines (IL-1RA, IL-8, MIP-1α, IP-10, MCP-1, or VEGF) were detected in either or both species but with no or only slight changes versus baseline. Similar changes in gene expression profiles were elicited in both species. These included enriched and up-regulated type I IFN-associated viral sensing, antiviral innate response, and dendritic cell activation pathways D3-D7 post-vaccination in both species. Hematological and blood biochemical parameters remained relatively unchanged versus baseline in both species. Low-level YF-17D viremia (RNAemia) was transiently detected in some cynomolgus macaques [28% (5/18)] but generally absent in humans [except one participant (5%; 1/20)].IMPORTANCECynomolgus macaques were confirmed as a valid surrogate model for replicating YF-17D vaccine-induced responses in humans and suggest a key role for type I IFN.
Collapse
Affiliation(s)
| | | | | | - Ernesto Marcos-Lopez
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| | - Caroline L. Bodinham
- Surrey Clinical Research Centre, University of Surrey, Guildford, Surrey, United Kingdom
| | - Antonio Cosma
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| | | | - Nina Dhooge
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| | | | - Stefan H. E. Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany; Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, Texas, USA
| | - Kathleen Pizzoferro
- Surrey Clinical Research Centre, University of Surrey, Guildford, Surrey, United Kingdom
| | - David J. M. Lewis
- Surrey Clinical Research Centre, University of Surrey, Guildford, Surrey, United Kingdom
| | - Frédéric Martinon
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| | - Anke Pagnon
- Research and Development, Sanofi, Marcy L'Etoile, France
| | - Franck Raynal
- Research and Development, Sanofi, Marcy L'Etoile, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| | - Roger Le Grand
- Université Paris-Saclay, INSERM, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay aux Roses, France
| |
Collapse
|
3
|
Sanchez-Felipe L, Alpizar YA, Ma J, Coelmont L, Dallmeier K. YF17D-based vaccines - standing on the shoulders of a giant. Eur J Immunol 2024; 54:e2250133. [PMID: 38571392 DOI: 10.1002/eji.202250133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 04/05/2024]
Abstract
Live-attenuated yellow fever vaccine (YF17D) was developed in the 1930s as the first ever empirically derived human vaccine. Ninety years later, it is still a benchmark for vaccines made today. YF17D triggers a particularly broad and polyfunctional response engaging multiple arms of innate, humoral and cellular immunity. This unique immunogenicity translates into an extraordinary vaccine efficacy and outstanding longevity of protection, possibly by single-dose immunization. More recently, progress in molecular virology and synthetic biology allowed engineering of YF17D as a powerful vector and promising platform for the development of novel recombinant live vaccines, including two licensed vaccines against Japanese encephalitis and dengue, even in paediatric use. Likewise, numerous chimeric and transgenic preclinical candidates have been described. These include prophylactic vaccines against emerging viral infections (e.g. Lassa, Zika and SARS-CoV-2) and parasitic diseases (e.g. malaria), as well as therapeutic applications targeting persistent infections (e.g. HIV and chronic hepatitis), and cancer. Efforts to overcome historical safety concerns and manufacturing challenges are ongoing and pave the way for wider use of YF17D-based vaccines. In this review, we summarize recent insights regarding YF17D as vaccine platform, and how YF17D-based vaccines may complement as well as differentiate from other emerging modalities in response to unmet medical needs and for pandemic preparedness.
Collapse
Affiliation(s)
- Lorena Sanchez-Felipe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Yeranddy A Alpizar
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Lotte Coelmont
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| |
Collapse
|
4
|
Zhang Y, Yang R, Yuan G, Li W, Cui Z, Xiao Z, Dong X, Yang H, Liu X, Zhang L, Hou Y, Liu M, Liu S, Hao Y, Zhang Y, Zheng X. Enhancing Inactivated Yellow Fever 17D Vaccine-Induced Immune Responses in Balb/C Mice Using Alum/CpG. Vaccines (Basel) 2023; 11:1744. [PMID: 38140149 PMCID: PMC10747526 DOI: 10.3390/vaccines11121744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
There are some concerns about the safety of live attenuated yellow fever vaccines (YF-live), particularly viscerotropic adverse events, which have a high mortality rate. The cellular production of the vaccine will not cause these adverse effects and has the potential to extend applicability to those who have allergic reactions, immunosuppression, and age. In this study, inactivated yellow fever (YF) was prepared and adsorbed with Alum/CpG. The cellular and humoral immunities were investigated in a mouse model. The results showed that Alum/CpG (20 μg/mL) could significantly increase the binding and neutralizing activities of the antibodies against YF. Moreover, the antibody level at day 28 after one dose was similar to that of the attenuated vaccine, but significantly higher after two doses. At the same time, Alum/CpG significantly increased the levels of IFN-γ and IL-4 cytokines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yuntao Zhang
- Beijing Institute of Biological Products Company Limited, Beijing 100170, China; (Y.Z.); (R.Y.); (G.Y.); (W.L.); (Z.C.); (Z.X.); (X.D.); (H.Y.); (X.L.); (L.Z.); (Y.H.); (M.L.); (S.L.); (Y.H.)
| | - Xiaotong Zheng
- Beijing Institute of Biological Products Company Limited, Beijing 100170, China; (Y.Z.); (R.Y.); (G.Y.); (W.L.); (Z.C.); (Z.X.); (X.D.); (H.Y.); (X.L.); (L.Z.); (Y.H.); (M.L.); (S.L.); (Y.H.)
| |
Collapse
|
5
|
Piras-Douce F, Broudic K, Chautard E, Raynal F, Courtois V, Gautheron S, Mantel N. Evaluation of safety and immuno-efficacy of a next generation live-attenuated yellow fever vaccine in cynomolgus macaques. Vaccine 2023; 41:1457-1470. [PMID: 36702693 DOI: 10.1016/j.vaccine.2022.11.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/17/2022] [Accepted: 11/20/2022] [Indexed: 01/26/2023]
Abstract
The increased demand for yellow fever (YF) vaccines over the last decade, along with insufficient availability of specific pathogen-free embryonated eggs required for timely vaccine production, has led to global YF vaccine shortages. A new live-attenuated YF vaccine candidate (generically referred to as vYF) cloned from a YF-VAX® vaccine (YF-17D vaccine) substrain adapted for growth in Vero cells cultured in serum-free media is currently in development. Here, we assessed the safety and immunogenicity of vYF, and its protective activity upon virulent challenge with wild-type yellow fever virus (YFV) Asibi, compared to licensed YF-17D vaccines in the translational cynomolgus macaque model. vYF was well tolerated with no major safety concerns. Vaccine-related safety observations were limited to minimal/minor microscopic findings at the injection sites and in the draining lymph nodes, consistent with expected stimulation of the immune system. vYF induced early differential expression of genes involved in antiviral innate immunity previously described in humans vaccinated with YF-17D vaccines, as well as YFV-specific IgM and IgG antibodies, high and sustained YFV neutralizing antibody titers from Day 14 up to at least Day 258 post-immunization, IgM+ and IgG+ memory B cells from Day 14 up to at least Day 221 post-vaccination, and Th1 interferon (IFN)-γ and interleukin (IL)-2 secreting effector and memory T cells. Additionally, vYF provided effective resistance to virulent challenge with wild-type YFV Asibi including complete protection against YFV-induced mortality, pathology, dysregulation of blood and liver soluble biomarkers, and a significant reduction in viremia and viral load to the limit of detection. These NHP data suggest that vYF would provide protection against YFV infection in practice, at least similar to that achieved with currently marketed YF-17D vaccines.
Collapse
Affiliation(s)
| | | | - Emilie Chautard
- Scientific and Digital Innovation, Sanofi, Marcy l'Etoile, France.
| | - Franck Raynal
- Research and External Innovation, Sanofi, Marcy l'Etoile, France.
| | | | | | - Nathalie Mantel
- Research and External Innovation, Sanofi, Marcy l'Etoile, France.
| |
Collapse
|
6
|
Cajaraville ACDRA, Gomes MPDB, Azamor T, Pereira RC, Neves PCDC, De Luca PM, de Lima SMB, Gaspar LP, Caride E, Freire MDS, Medeiros MA. Evaluation of Two Adjuvant Formulations for an Inactivated Yellow Fever 17DD Vaccine Candidate in Mice. Vaccines (Basel) 2022; 11:73. [PMID: 36679918 PMCID: PMC9865672 DOI: 10.3390/vaccines11010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/10/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
The attenuated yellow fever (YF) vaccine is one of the most successful vaccines ever developed. After a single dose administration YF vaccine can induce balanced Th1/Th2 immune responses and long-lasting neutralizing antibodies. These attributes endorsed it as a model of how to properly stimulate the innate response to target protective immune responses. Despite their longstanding success, attenuated YF vaccines can cause rare fatal adverse events and are contraindicated for persons with immunosuppression, egg allergy and age < 6 months and >60 years. These drawbacks have encouraged the development of a non-live vaccine. The aim of the present study is to characterize and compare the immunological profile of two adjuvant formulations of an inactivated YF 17DD vaccine candidate. Inactivated YF vaccine formulations based on alum (Al(OH)3) or squalene (AddaVax®) were investigated by immunization of C57BL/6 mice in 3-dose or 2-dose schedules, respectively, and compared with a single dose of attenuated YF virus 17DD. Sera were analyzed by ELISA and Plaque Reduction Neutralization Test (PRNT) for detection of total IgG and neutralizing antibodies against YF virus. In addition, splenocytes were collected to evaluate cellular responses by ELISpot. Both inactivated formulations were able to induce high titers of IgG against YF, although neutralizing antibodies levels were borderline on pre-challenge samples. Analysis of IgG subtypes revealed a predominance of IgG2a associated with improved neutralizing capacity in animals immunized with the attenuated YF vaccine, and a predominance of IgG1 in groups immunized with experimental non-live formulations (alum and AddaVax®). After intracerebral (IC) challenge, attenuated and inactivated vaccine formulations showed an increase in neutralizing antibodies. The AddaVax®-based inactivated vaccine and the attenuated vaccine achieved 100% protection, and alum-based equivalent formulation achieved 70% protection.
Collapse
Affiliation(s)
| | - Mariana Pierre de Barros Gomes
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Tamiris Azamor
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Renata Carvalho Pereira
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Patrícia Cristina da Costa Neves
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Paula Mello De Luca
- Instituto Oswaldo Cruz (IOC), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Sheila Maria Barbosa de Lima
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Luciane Pinto Gaspar
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Elena Caride
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Marcos da Silva Freire
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Marco Alberto Medeiros
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
7
|
Tavares da Silva Fernandes A, Moreira SB, Gaspar LP, Simões M, Cajaraville ACDRA, Pereira RC, Gomes MPDB, Linhares JHR, Santos VDO, Santos RT, Amorim JF, Barros TADC, Melgaço JG, da Silva AMV, Fernandes CB, Tubarão LN, da Silva J, Caride EC, Borges MB, Guimarães RC, Marchevsky RS, de Lima SMB, Ano Bom APD, Neves PCDC, Pissinatti A, Freire MDS. Safety and immunogenicity of 17DD attenuated yellow fever vaccine in howler monkeys (Alouatta spp.). J Med Primatol 2020; 50:36-45. [PMID: 33219623 DOI: 10.1111/jmp.12501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 09/17/2020] [Accepted: 10/19/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND Alouatta spp. are highly susceptible to yellow fever (YF) infection and develop an often fatal disease. The threat posed by an outbreak started in 2016 leads us to investigate vaccination as a potential tool in preventing YF in non-human primates (NHP). METHODS Susceptible howler monkeys were immunized with three different concentrations of the human Brazilian commercial YF17DD vaccine. Post-vaccination viremia/RNAemia, immunogenicity, and safety were characterized. RESULTS The vaccine did not produce YF clinical manifestations in any of the NHPs. After immunization, all animals seroconverted demonstrating the ability of the YF vaccine to induce humoral response in Alouatta species. CONCLUSIONS The present work has demonstrated the safe and immunogenic profile of the existing YF 17DD vaccine in howler monkeys. This knowledge may support further studies with other susceptible monkey species and provide a possible solution for controlling epizootics and preventing the devastation of endangered species.
Collapse
Affiliation(s)
| | - Silvia Bahadian Moreira
- Centro de Primatologia do Rio de Janeiro - CPRJ, Instituto Estadual do Ambiente, Guapimirim, Brazil
| | - Luciane Pinto Gaspar
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | - Marisol Simões
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | | | - Renata Carvalho Pereira
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | | | | | - Vanessa de Oliveira Santos
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | - Renata Tourinho Santos
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | - Juliana Fernandes Amorim
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | | | - Juliana Gil Melgaço
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | | | - Camilla Bayma Fernandes
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | - Luciana Neves Tubarão
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | - Jane da Silva
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | - Elena Cristina Caride
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | - Maria Beatriz Borges
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | - Rosane Cuber Guimarães
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | - Renato Sérgio Marchevsky
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | - Sheila Maria Barbosa de Lima
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | - Ana Paula Dinis Ano Bom
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| | | | - Alcides Pissinatti
- Centro de Primatologia do Rio de Janeiro - CPRJ, Instituto Estadual do Ambiente, Guapimirim, Brazil.,Centro Universitário Serra dos Orgãos, Unifeso, Teresópolis, Brazil
| | - Marcos da Silva Freire
- Fundação Oswaldo Cruz-Fiocruz, Instituto de Tecnologia em Imunobiológicos/Bio-Manguinhos, Rio de Janeiro, Brazil
| |
Collapse
|
8
|
Bartish M, Del Rincón SV, Rudd CE, Saragovi HU. Aiming for the Sweet Spot: Glyco-Immune Checkpoints and γδ T Cells in Targeted Immunotherapy. Front Immunol 2020; 11:564499. [PMID: 33133075 PMCID: PMC7550643 DOI: 10.3389/fimmu.2020.564499] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/31/2020] [Indexed: 11/23/2022] Open
Abstract
Though a healthy immune system is capable of recognizing and eliminating emergent cancerous cells, an established tumor is adept at escaping immune surveillance. Altered and tumor-specific expression of immunosuppressive cell surface carbohydrates, also termed the “tumor glycocode,” is a prominent mechanism by which tumors can escape anti-tumor immunity. Given their persistent and homogeneous expression, tumor-associated glycans are promising targets to be exploited as biomarkers and therapeutic targets. However, the exploitation of these glycans has been a challenge due to their low immunogenicity, immunosuppressive properties, and the inefficient presentation of glycolipids in a conventional major histocompatibility complex (MHC)-restricted manner. Despite this, a subset of T-cells expressing the gamma and delta chains of the T-cell receptor (γδ T cells) exist with a capacity for MHC-unrestricted antigen recognition and potent inherent anti-tumor properties. In this review, we discuss the role of tumor-associated glycans in anti-tumor immunity, with an emphasis on the potential of γδ T cells to target the tumor glycocode. Understanding the many facets of this interaction holds the potential to unlock new ways to use both tumor-associated glycans and γδ T cells in novel therapeutic interventions.
Collapse
Affiliation(s)
- Margarita Bartish
- Lady Davis Institute, Jewish General Hospital, Translational Center for Research in Cancer, McGill University, Montreal, QC, Canada
| | - Sonia V Del Rincón
- Lady Davis Institute, Jewish General Hospital, Translational Center for Research in Cancer, McGill University, Montreal, QC, Canada.,Oncology and Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Christopher E Rudd
- Division of Immuno-Oncology, Research Center Maisonneuve-Rosemont Hospital, Montreal, QC, Canada.,Département de Médecine, Université de Montréal, Montreal, QC, Canada
| | - H Uri Saragovi
- Lady Davis Institute, Jewish General Hospital, Translational Center for Research in Cancer, McGill University, Montreal, QC, Canada.,Oncology and Experimental Medicine, McGill University, Montreal, QC, Canada.,Pharmacology and Therapeutics, and Ophthalmology and Vision Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
9
|
Olusola BA, Kabelitz D, Olaleye DO, Odaibo GN. Early HIV infection is associated with reduced proportions of gamma delta T subsets as well as high creatinine and urea levels. Scand J Immunol 2020; 91:e12868. [PMID: 32052490 PMCID: PMC7335456 DOI: 10.1111/sji.12868] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 12/19/2019] [Accepted: 02/01/2020] [Indexed: 01/02/2023]
Abstract
Renal dysfunctions are major predictors of co-morbidities and mortality in HIV-infected individuals. Unconventional T cells have been shown to regulate kidney functions. However, there is dearth of information on the effect of HIV-associated nephropathies on γδ and DN T cells. It is also not clear whether γδ T cell perturbations observed during the early stages of HIV infection occur before immune activation. In this study, we investigated the relationship between creatinine and urea on the number of unconventional T cells in HIV-infected individuals at the early and chronic stages of infection. Persons in the chronic stage of infection were divided into treatment naïve and exposed groups. Treatment exposed individuals were further subdivided into groups with undetectable and detectable HIV-1RNA in their blood. Creatinine and urea levels were significantly higher among persons in the early HIV infection compared with the other groups. Proportions of γδ T, γδ + CD8, γδ + CD16 cells were also significantly reduced in the early stage of HIV infection (P < .01). Markers of immune activation, CD4 + HLA-DR and CD8 + HLA-DR, were also significantly reduced during early HIV infection (P < .01). Taken together, our findings suggest that high levels of renal markers as well as reduced proportions of gamma delta T cells are associated with the early stages of HIV infection. This event likely occurs before systemic immune activation reaches peak levels. This study provides evidence for the need for early HIV infection diagnosis and treatment.
Collapse
Affiliation(s)
- Babatunde A. Olusola
- Department of Virology, College of Medicine, University of
Ibadan, Ibadan, Oyo State, Nigeria
| | - Dieter Kabelitz
- Institute of Immunology, UKSH Campus Kiel,
Christian-Albrechts-University, Kiel, Germany
| | - David O. Olaleye
- Department of Virology, College of Medicine, University of
Ibadan, Ibadan, Oyo State, Nigeria
| | - Georgina N. Odaibo
- Department of Virology, College of Medicine, University of
Ibadan, Ibadan, Oyo State, Nigeria
| |
Collapse
|
10
|
Sant S, Jenkins MR, Dash P, Watson KA, Wang Z, Pizzolla A, Koutsakos M, Nguyen TH, Lappas M, Crowe J, Loudovaris T, Mannering SI, Westall GP, Kotsimbos TC, Cheng AC, Wakim L, Doherty PC, Thomas PG, Loh L, Kedzierska K. Human γδ T-cell receptor repertoire is shaped by influenza viruses, age and tissue compartmentalisation. Clin Transl Immunology 2019; 8:e1079. [PMID: 31559018 PMCID: PMC6756999 DOI: 10.1002/cti2.1079] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background Although γδ T cells comprise up to 10% of human peripheral blood T cells, questions remain regarding their role in disease states and T‐cell receptor (TCR) clonal expansions. We dissected anti‐viral functions of human γδ T cells towards influenza viruses and defined influenza‐reactive γδ TCRs in the context of γδ‐TCRs across the human lifespan. Methods We performed 51Cr‐killing assay and single‐cell time‐lapse live video microscopy to define mechanisms underlying γδ T‐cell‐mediated killing of influenza‐infected targets. We assessed cytotoxic profiles of γδ T cells in influenza‐infected patients and IFN‐γ production towards influenza‐infected lung epithelial cells. Using single‐cell RT‐PCR, we characterised paired TCRγδ clonotypes for influenza‐reactive γδ T cells in comparison with TCRs from healthy neonates, adults, elderly donors and tissues. Results We provide the first visual evidence of γδ T‐cell‐mediated killing of influenza‐infected targets and show distinct features to those reported for CD8+ T cells. γδ T cells displayed poly‐cytotoxic profiles in influenza‐infected patients and produced IFN‐γ towards influenza‐infected cells. These IFN‐γ‐producing γδ T cells were skewed towards the γ9δ2 TCRs, particularly expressing the public GV9‐TCRγ, capable of pairing with numerous TCR‐δ chains, suggesting their significant role in γδ T‐cell immunity. Neonatal γδ T cells displayed extensive non‐overlapping TCRγδ repertoires, while adults had enriched γ9δ2‐pairings with diverse CDR3γδ regions. Conversely, the elderly showed distinct γδ‐pairings characterised by large clonal expansions, a profile also prominent in adult tissues. Conclusion Human TCRγδ repertoire is shaped by age, tissue compartmentalisation and the individual's history of infection, suggesting that these somewhat enigmatic γδ T cells indeed respond to antigen challenge.
Collapse
Affiliation(s)
- Sneha Sant
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Misty R Jenkins
- Immunology Division Walter and Eliza Hall Institute Melbourne VIC Australia.,LaTrobe Institute for Molecular Science La Trobe University Melbourne VIC Australia.,Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Pradyot Dash
- Department of Immunology St Jude Children's Research Hospital Memphis TN USA
| | - Katherine A Watson
- Immunology Division Walter and Eliza Hall Institute Melbourne VIC Australia
| | - Zhongfang Wang
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Angela Pizzolla
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Marios Koutsakos
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Thi Ho Nguyen
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group Department of Obstetrics & Gynaecology Mercy Hospital for Women University of Melbourne Melbourne VIC Australia
| | | | - Tom Loudovaris
- Immunology and Diabetes Unit St Vincent's Institute of Medical Research Fitzroy VIC Australia
| | - Stuart I Mannering
- Immunology and Diabetes Unit St Vincent's Institute of Medical Research Fitzroy VIC Australia
| | - Glen P Westall
- Lung Transplant Unit Alfred Hospital Melbourne VIC Australia
| | - Tom C Kotsimbos
- Department of Allergy, Immunology and Respiratory Medicine The Alfred Hospital Melbourne VIC Australia.,Department of Medicine Central Clinical School The Alfred Hospital Melbourne Monash University Melbourne VIC Australia
| | - Allen C Cheng
- School of Public Health and Preventive Medicine Monash University Melbourne VIC Australia.,Infection Prevention and Healthcare Epidemiology Unit Alfred Health Melbourne VIC Australia
| | - Linda Wakim
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Peter C Doherty
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia.,Immunology Division Walter and Eliza Hall Institute Melbourne VIC Australia
| | - Paul G Thomas
- Department of Immunology St Jude Children's Research Hospital Memphis TN USA
| | - Liyen Loh
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology University of Melbourne at The Peter Doherty Institute for Infection and Immunity Melbourne VIC Australia
| |
Collapse
|
11
|
Gamma-interferon exerts a critical early restriction on replication and dissemination of yellow fever virus vaccine strain 17D-204. NPJ Vaccines 2018; 3:5. [PMID: 29387474 PMCID: PMC5780476 DOI: 10.1038/s41541-017-0039-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/27/2017] [Accepted: 11/30/2017] [Indexed: 11/11/2022] Open
Abstract
Live attenuated viruses are historically among the most effective viral vaccines. Development of a safe vaccine requires the virus to be less virulent, a phenotype that is historically arrived by empirical evaluation often leaving the mechanisms of attenuation unknown. The yellow fever virus 17D live attenuated vaccine strain has been developed as a delivery vector for heterologous antigens; however, the mechanisms of attenuation remain elusive. The successful and safe progress of 17D as a vaccine vector and the development of live attenuated vaccines (LAVs) to related flaviviruses requires an understanding of the molecular mechanisms leading to attenuation. Using subcutaneous infection of interferon-deficient mouse models of wild type yellow fever virus (WT YFV) pathogenesis and 17D-mediated immunity, we found that, in the absence of type I IFN (IFN-α/β), type II interferon (IFN-γ) restricted 17D replication, but not that of WT YFV, by 1–2 days post-infection. In this context, IFN-γ responses protected 17D-infected animals from mortality, largely restricted the virus to lymphoid organs, and eliminated viscerotropic disease signs such as steatosis in the liver and inflammatory cell infiltration into the spleen. However, WT YFV caused a disseminated infection, gross liver pathology, and rapid death of the animals. In vitro, IFN-γ treatment of myeloid cells suppressed the replication of 17D significantly more than that of WT YFV, suggesting a direct differential effect on 17D virus replication. Together these data indicate that an important mechanism of 17D attenuation in vivo is increased sensitivity to IFN-γ stimulated responses elicited early after infection. The interferon gamma protein may play a key role in preventing yellow fever vaccine 17D from causing virus-like disease in recipients. The highly effective 17D vaccine is a less virulent form of the virus, but can induce severe disease in rare cases. A research group from the University of Pittsburgh, led by William Klimstra, investigated the impact of the vaccine on mice, as the mechanism by which hosts defend against its disease-causing potential is not fully understood. They found that interferon gamma restricted the replication and spread of the attenuated virus (the vaccine) but not its natural form. This study helps to inform efforts to improve the safety of the 17D vaccine, as well as the other vaccines that use it as a template for prophylaxis against other pathogens.
Collapse
|
12
|
Tottey S, Shoji Y, Jones RM, Chichester JA, Green BJ, Musiychuk K, Si H, Manceva SD, Rhee A, Shamloul M, Norikane J, Guimarães RC, Caride E, Silva ANMR, Simões M, Neves PCC, Marchevsky R, Freire MS, Streatfield SJ, Yusibov V. Plant-Produced Subunit Vaccine Candidates against Yellow Fever Induce Virus Neutralizing Antibodies and Confer Protection against Viral Challenge in Animal Models. Am J Trop Med Hyg 2017; 98:420-431. [PMID: 29231157 DOI: 10.4269/ajtmh.16-0293] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Yellow fever (YF) is a viral disease transmitted by mosquitoes and endemic mostly in South America and Africa with 20-50% fatality. All current licensed YF vaccines, including YF-Vax® (Sanofi-Pasteur, Lyon, France) and 17DD-YFV (Bio-Manguinhos, Rio de Janeiro, Brazil), are based on live attenuated virus produced in hens' eggs and have been widely used. The YF vaccines are considered safe and highly effective. However, a recent increase in demand for YF vaccines and reports of rare cases of YF vaccine-associated fatal adverse events have provoked interest in developing a safer YF vaccine that can be easily scaled up to meet this increased global demand. To this point, we have engineered the YF virus envelope protein (YFE) and transiently expressed it in Nicotiana benthamiana as a stand-alone protein (YFE) or as fusion to the bacterial enzyme lichenase (YFE-LicKM). Immunogenicity and challenge studies in mice demonstrated that both YFE and YFE-LicKM elicited virus neutralizing (VN) antibodies and protected over 70% of mice from lethal challenge infection. Furthermore, these two YFE-based vaccine candidates induced VN antibody responses with high serum avidity in nonhuman primates and these VN antibody responses were further enhanced after challenge infection with the 17DD strain of YF virus. These results demonstrate partial protective efficacy in mice of YFE-based subunit vaccines expressed in N. benthamiana. However, their efficacy is inferior to that of the live attenuated 17DD vaccine, indicating that formulation development, such as incorporating a more suitable adjuvant, may be required for product development.
Collapse
Affiliation(s)
- Stephen Tottey
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Yoko Shoji
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - R Mark Jones
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | | | - Brian J Green
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | | | - Huaxin Si
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | | | - Amy Rhee
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Moneim Shamloul
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Joey Norikane
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Rosane C Guimarães
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Elena Caride
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Andrea N M R Silva
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Marisol Simões
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Patricia C C Neves
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Renato Marchevsky
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Marcos S Freire
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | | | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| |
Collapse
|
13
|
Ferreira CDC, Campi-Azevedo AC, Peruhype-Magalhāes V, Costa-Pereira C, Albuquerque CPD, Muniz LF, Yokoy de Souza T, Oliveira ACV, Martins-Filho OA, da Mota LMH. The 17D-204 and 17DD yellow fever vaccines: an overview of major similarities and subtle differences. Expert Rev Vaccines 2017; 17:79-90. [PMID: 29172832 DOI: 10.1080/14760584.2018.1406800] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION The yellow fever vaccine is a live attenuated virus vaccine that is considered one of the most efficient vaccines produced to date. The original 17D strain generated the substrains 17D-204 and 17DD, which are used for the current production of vaccines against yellow fever. The 17D-204 and 17DD substrains present subtle differences in their nucleotide compositions, which can potentially lead to variations in immunogenicity and reactogenicity. We will address the main changes in the immune responses induced by the 17D-204 and 17DD yellow fever vaccines and report similarities and differences between these vaccines in cellular and humoral immunity . This is a relevant issue in view of the re-emergence of yellow fever in Uganda in 2016 and in Brazil in the beginning of 2017. AREAS COVERED This article will be divided into 8 sections that will analyze the innate immune response, adaptive immune response, humoral response, production of cytokines, immunity in children, immunity in the elderly, gene expression and adverse reactions. EXPERT COMMENTARY The 17D-204 and 17DD yellow fever vaccines present similar immunogenicity, with strong activation of the cellular and humoral immune responses. Additionally, both vaccines have similar adverse effects, which are mostly mild and thus are considered safe.
Collapse
Affiliation(s)
| | | | | | | | | | - Luciana Feitosa Muniz
- a Department of Rheumatology , University Hospital of Brasilia, University of Brasilia , Brasilia , Brazil
| | - Talita Yokoy de Souza
- a Department of Rheumatology , University Hospital of Brasilia, University of Brasilia , Brasilia , Brazil
| | | | | | | |
Collapse
|
14
|
Watson AM, Klimstra WB. T Cell-Mediated Immunity towards Yellow Fever Virus and Useful Animal Models. Viruses 2017; 9:E77. [PMID: 28398253 PMCID: PMC5408683 DOI: 10.3390/v9040077] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/04/2017] [Accepted: 04/06/2017] [Indexed: 12/31/2022] Open
Abstract
The 17D line of yellow fever virus vaccines is among the most effective vaccines ever created. The humoral and cellular immunity elicited by 17D has been well characterized in humans. Neutralizing antibodies have long been known to provide protection against challenge with a wild-type virus. However, a well characterized T cell immune response that is robust, long-lived and polyfunctional is also elicited by 17D. It remains unclear whether this arm of immunity is protective following challenge with a wild-type virus. Here we introduce the 17D line of yellow fever virus vaccines, describe the current state of knowledge regarding the immunity directed towards the vaccines in humans and conclude with a discussion of animal models that are useful for evaluating T cell-mediated immune protection to yellow fever virus.
Collapse
Affiliation(s)
- Alan M Watson
- Center for Vaccine Research, Departments of Microbiology and Molecular Genetics, and Immunology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA.
| | - William B Klimstra
- Center for Vaccine Research, Departments of Microbiology and Molecular Genetics, and Immunology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA 15261, USA.
| |
Collapse
|
15
|
Heinen MP, Cambier L, Fievez L, Mignon B. Are Th17 Cells Playing a Role in Immunity to Dermatophytosis? Mycopathologia 2016; 182:251-261. [PMID: 27878642 DOI: 10.1007/s11046-016-0093-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 11/13/2016] [Indexed: 12/24/2022]
Abstract
Despite their superficial localization in the skin, pathogenic dermatophytes can induce a complex but still misunderstood immune response in their hosts. The cell-mediated immunity (CMI) is correlated with both clinical recovery and protection against reinfection, and CD4+ T lymphocytes have been recognized as a crucial component of the immune defense against dermatophytes. Before the discovery of the Th17 pathway, CMI was considered to be only dependent of Th1 cells, and thus most studies on the immunology of dermatophytosis have focused on the Th1 pathway. Nevertheless, the fine comparative analysis of available scientific data on immunology of dermatophytosis in one hand and on the Th17 pathway mechanisms involved in opportunistic mucosal fungal infections in the other hand reveals that some key elements of the Th17 pathway can be activated by dermatophytes. Stimulation of the Th17 pathway could occur through the activation of some C-type lectin-like receptors and inflammasome in antigen-presenting cells. The Th17 cells could go back to the affected skin and by the production of signature cytokines could induce the effector mechanisms like the recruitment of polymorphonuclear neutrophils and the synthesis of antimicrobial peptides. In conclusion, besides the Th1 pathway, which is important to the immune response against dermatophytes, there are also growing evidences for the involvement of the Th17 pathway.
Collapse
Affiliation(s)
- Marie-Pierre Heinen
- Veterinary Mycology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Quartier Vallée 2, Avenue de Cureghem 6, B-43a, 4000, Liège, Belgium
| | - Ludivine Cambier
- Veterinary Mycology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Quartier Vallée 2, Avenue de Cureghem 6, B-43a, 4000, Liège, Belgium
| | - Laurence Fievez
- Laboratory of Cellular and Molecular Immunology, GIGA Research, Quartier Hôpital, University of Liège, Avenue de l'Hôpital 11, B-34, 4000, Liège, Belgium
| | - Bernard Mignon
- Veterinary Mycology, Fundamental and Applied Research for Animals & Health (FARAH), Faculty of Veterinary Medicine, University of Liège, Quartier Vallée 2, Avenue de Cureghem 6, B-43a, 4000, Liège, Belgium.
| |
Collapse
|
16
|
Abstract
Mammalian immune system is a complex amalgam of diverse cellular and noncellular components such as cytokines, receptors and co-receptors. FAM26F (family with sequence similarity 26, member F) is a recently identified tetraspanin-like membrane glycoprotein which is predicted to make homophilic interactions and potential synapses between several immune cells including CD4+, CD8+, NK, dendritic cells and macrophages. Various whole transcriptome analyses have demonstrated the differential expression of FAM26F in several bacterial, viral and parasitic infections, in certain pathophysiological conditions such as liver and heart transplantation, and in various cancers. The complete understanding of transcriptional regulation of FAM26F is in its infancy however it is up regulated by various stimulants such as polyI:C, LPS, INF gamma and TNF alpha, and via various proposed pathways including TLR3, TLR4 IFN-β and Dectin-1. These pathways can merge in STAT1 activation. The synergistic expression of FAM26F on both NK-cells and myeloid dendritic cells is required to activate NK-cells against tumors via its cytoplasmic tail, thus emphasizing therapeutic potential of FAM26F for NK sensitive tumors. Current review provides a comprehensive basis to propose that FAM26F expression level is at least a hallmark for IFN-γ-lead immune responses and thus can proficiently be regarded as an early diagnostic marker. Future investigation dissecting the role of FAM26F in activation of various immune cell populations in local amplification by cell-cell contact is crucial to provide the missing link imperative for elucidating the relevance of this protein in immune responses.
Collapse
Affiliation(s)
- Uzma Malik
- a Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB) , National University of Sciences and Technology (NUST) , Islamabad , Pakistan
| | - Aneela Javed
- a Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB) , National University of Sciences and Technology (NUST) , Islamabad , Pakistan
| |
Collapse
|
17
|
Cong Y, McArthur MA, Cohen M, Jahrling PB, Janosko KB, Josleyn N, Kang K, Zhang T, Holbrook MR. Characterization of Yellow Fever Virus Infection of Human and Non-human Primate Antigen Presenting Cells and Their Interaction with CD4+ T Cells. PLoS Negl Trop Dis 2016; 10:e0004709. [PMID: 27191161 PMCID: PMC4871483 DOI: 10.1371/journal.pntd.0004709] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 04/23/2016] [Indexed: 12/15/2022] Open
Abstract
Humans infected with yellow fever virus (YFV), a mosquito-borne flavivirus, can develop illness ranging from a mild febrile disease to hemorrhagic fever and death. The 17D vaccine strain of YFV was developed in the 1930s, has been used continuously since development and has proven very effective. Genetic differences between vaccine and wild-type viruses are few, yet viral or host mechanisms associated with protection or disease are not fully understood. Over the past 20 years, a number of cases of vaccine-associated disease have been identified following vaccination with 17D; these cases have been correlated with reduced immune status at the time of vaccination. Recently, several studies have evaluated T cell responses to vaccination in both humans and non-human primates, but none have evaluated the response to wild-type virus infection. In the studies described here, monocyte-derived macrophages (MDM) and dendritic cells (MoDC) from both humans and rhesus macaques were evaluated for their ability to support infection with either wild-type Asibi virus or the 17D vaccine strain and the host cytokine and chemokine response characterized. Human MoDC and MDM were also evaluated for their ability to stimulate CD4+ T cells. It was found that MoDC and MDM supported viral replication and that there were differential cytokine responses to infection with either wild-type or vaccine viruses. Additionally, MoDCs infected with live 17D virus were able to stimulate IFN-γ and IL-2 production in CD4+ T cells, while cells infected with Asibi virus were not. These data demonstrate that wild-type and vaccine YFV stimulate different responses in target antigen presenting cells and that wild-type YFV can inhibit MoDC activation of CD4+ T cells, a critical component in development of protective immunity. These data provide initial, but critical insight into regulatory capabilities of wild-type YFV in development of disease.
Collapse
Affiliation(s)
- Yu Cong
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft. Detrick, Frederick, Maryland, United States of America
| | - Monica A. McArthur
- Department of Pediatrics and Center for Vaccine Development, University of Maryland, Baltimore, Maryland, United States of America
| | - Melanie Cohen
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft. Detrick, Frederick, Maryland, United States of America
| | - Peter B. Jahrling
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft. Detrick, Frederick, Maryland, United States of America
| | - Krisztina B. Janosko
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft. Detrick, Frederick, Maryland, United States of America
| | - Nicole Josleyn
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft. Detrick, Frederick, Maryland, United States of America
| | - Kai Kang
- Gears Inc., Lanham, Maryland, United States of America
| | - Tengfei Zhang
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft. Detrick, Frederick, Maryland, United States of America
| | - Michael R. Holbrook
- National Institute of Allergy and Infectious Diseases (NIAID) Integrated Research Facility, Ft. Detrick, Frederick, Maryland, United States of America
| |
Collapse
|
18
|
Julander JG. Animal models of yellow fever and their application in clinical research. Curr Opin Virol 2016; 18:64-9. [PMID: 27093699 DOI: 10.1016/j.coviro.2016.03.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 03/30/2016] [Indexed: 11/18/2022]
Abstract
Yellow fever virus (YFV) is an arbovirus that causes significant human morbidity and mortality. This virus has been studied intensively over the past century, although there are still no treatment options for those who become infected. Periodic and unpredictable yellow fever (YF) outbreaks in Africa and South America continue to occur and underscore the ongoing need to further understand this viral disease and to develop additional countermeasures to prevent or treat cases of illness. The use of animal models of YF is critical to accomplishing this goal. There are several animal models of YF that replicate various aspects of clinical disease and have provided insight into pathogenic mechanisms of the virus. These typically include mice, hamsters and non-human primates (NHP). The utilities and shortcomings of the available animal models of YF are discussed. Information on recent discoveries that have been made in the field of YFV research is also included as well as important future directions in further ameliorating the morbidity and mortality that occur as a result of YFV infection. It is anticipated that these model systems will help facilitate further improvements in the understanding of this virus and in furthering countermeasures to prevent or treat infections.
Collapse
Affiliation(s)
- Justin G Julander
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, United States.
| |
Collapse
|
19
|
Abstract
Yellow fever 17D vaccine is one of the oldest live-attenuated vaccines in current use that is recognized historically for its immunogenic and safe properties. These unique properties of 17D are presently exploited in rationally designed recombinant vaccines targeting not only flaviviral antigens but also other pathogens of public health concern. Several candidate vaccines based on 17D have advanced to human trials, and a chimeric recombinant Japanese encephalitis vaccine utilizing the 17D backbone has been licensed. The mechanism(s) of attenuation for 17D are poorly understood; however, recent insights from large in silico studies have indicated particular host genetic determinants contributing to the immune response to the vaccine, which presumably influences the considerable durability of protection, now in many cases considered to be lifelong. The very rare occurrence of severe adverse events for 17D is discussed, including a recent fatal case of vaccine-associated viscerotropic disease.
Collapse
Affiliation(s)
- Andrew S Beck
- a 1 Department of Pathology, University of Texas Medical Branch, Galveston TX 77555-0609, USA
| | | |
Collapse
|
20
|
Pereira RC, Silva ANMR, Souza MCO, Silva MV, Neves PPCC, Silva AAMV, Matos DDCS, Herrera MAO, Yamamura AMY, Freire MS, Gaspar LP, Caride E. An inactivated yellow fever 17DD vaccine cultivated in Vero cell cultures. Vaccine 2015; 33:4261-8. [PMID: 25862300 DOI: 10.1016/j.vaccine.2015.03.077] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 03/10/2015] [Accepted: 03/24/2015] [Indexed: 02/03/2023]
Abstract
Yellow fever is an acute infectious disease caused by prototype virus of the genus Flavivirus. It is endemic in Africa and South America where it represents a serious public health problem causing epidemics of hemorrhagic fever with mortality rates ranging from 20% to 50%. There is no available antiviral therapy and vaccination is the primary method of disease control. Although the attenuated vaccines for yellow fever show safety and efficacy it became necessary to develop a new yellow fever vaccine due to the occurrence of rare serious adverse events, which include visceral and neurotropic diseases. The new inactivated vaccine should be safer and effective as the existing attenuated one. In the present study, the immunogenicity of an inactivated 17DD vaccine in C57BL/6 mice was evaluated. The yellow fever virus was produced by cultivation of Vero cells in bioreactors, inactivated with β-propiolactone, and adsorbed to aluminum hydroxide (alum). Mice were inoculated with inactivated 17DD vaccine containing alum adjuvant and followed by intracerebral challenge with 17DD virus. The results showed that animals receiving 3 doses of the inactivated vaccine (2 μg/dose) with alum adjuvant had neutralizing antibody titers above the cut-off of PRNT50 (Plaque Reduction Neutralization Test). In addition, animals immunized with inactivated vaccine showed survival rate of 100% after the challenge as well as animals immunized with commercial attenuated 17DD vaccine.
Collapse
Affiliation(s)
- Renata C Pereira
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Andrea N M R Silva
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Marta Cristina O Souza
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Marlon V Silva
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Patrícia P C C Neves
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Andrea A M V Silva
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Denise D C S Matos
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Miguel A O Herrera
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Anna M Y Yamamura
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Marcos S Freire
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| | - Luciane P Gaspar
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil.
| | - Elena Caride
- Oswaldo Cruz Foundation (FIOCRUZ), Bio-Manguinhos, Avenida Brasil 4365, 21045-900, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
21
|
Liao S, Zhang W, Hu X, Wang W, Deng D, Wang H, Wang C, Zhou J, Wang S, Zhang H, Ma D. A novel "priming-boosting" strategy for immune interventions in cervical cancer. Mol Immunol 2015; 64:295-305. [PMID: 25575128 DOI: 10.1016/j.molimm.2014.12.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 12/13/2014] [Accepted: 12/16/2014] [Indexed: 12/22/2022]
Abstract
Despite the encouraging development of a preventive vaccine for human papillomavirus (HPV), it cannot improve ongoing infections. Therefore, a new vaccine is urgently needed that can prevent and treat cervical cancer, and cure pre-cancerous lesions. In this study, we constructed two peptide-based vaccines. The first was a short-term, long-peptide (ST-LP) vaccine that simultaneously targeted three key carcinogenic epitopes (E5-E6-E7) on HPV16. We tested this vaccine in murine TC-1 cells infected with a recombinant adeno-associated virus (rAAV) fused with HPV16E5 DNA (rTC-1 cells), which served as a cell model; we also tested it in immune-competent mice loaded with rTC-1 cells, which served as an ectopic tumor model. The ST-LP injections resulted in strong, cell-mediated immunity, capable of attacking and eliminating abnormal antigen-bearing cells. Furthermore, to prolong immunogenic capability, we designed a unique rAAV that encoded the three predicted epitopes for a second, long-term, long-peptide (LT-LP) vaccine. Moreover, we used a new immune strategy of continuous re-injections, where three ST-LP injections were performed at one-week intervals (days 0, 7, 14), then one LT-LP injection was performed on day 120. Our in vitro and in vivo studies revealed that this strategy could boost the immune response to produce longer and stronger protection against target cells, and mice were thoroughly protected from tumor growth. Our results showed that priming the immune system with the ST-LP vaccine, followed by boosting the immune system with the LT-LP vaccine could generate a rapid, robust, durable cytotoxic T-lymphocyte response to HPV16-positive tumors.
Collapse
Affiliation(s)
- Shujie Liao
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | | | - Xiaoji Hu
- Wuhan General Hospita of Guangzhou Military Region, PR China
| | - Wei Wang
- Southern Medical University South Hospital, PR China
| | - Dongrui Deng
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Hui Wang
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Changyu Wang
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Jianfeng Zhou
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Shixuan Wang
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China
| | - Hanwang Zhang
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China.
| | - Ding Ma
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, PR China.
| |
Collapse
|
22
|
Bonaldo MC, Sequeira PC, Galler R. The yellow fever 17D virus as a platform for new live attenuated vaccines. Hum Vaccin Immunother 2014; 10:1256-65. [PMID: 24553128 DOI: 10.4161/hv.28117] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The live-attenuated yellow fever 17D virus is one of the most outstanding human vaccines ever developed. It induces efficacious immune responses at a low production cost with a well-established manufacture process. These advantages make the YF17D virus attractive as a vector for the development of new vaccines. At the beginning of vector development studies, YF17D was genetically manipulated to express other flavivirus prM and E proteins, components of the viral envelope. While these 17D recombinants are based on the substitution of equivalent YF17D genes, other antigens from unrelated pathogens have also been successfully expressed and delivered by recombinant YF17D viruses employing alternative strategies for genetic manipulation of the YF17D genome. Herein, we discuss these strategies in terms of possibilities of single epitope or larger sequence expression and the main properties of these replication-competent viral platforms.
Collapse
Affiliation(s)
- Myrna C Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, IOC, Fiocruz; Rio de Janeiro, Brazil
| | - Patrícia C Sequeira
- Laboratório de Biologia Molecular de Flavivírus, IOC, Fiocruz; Rio de Janeiro, Brazil
| | - Ricardo Galler
- Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
de Santana MGV, Neves PCC, dos Santos JR, Lima NS, dos Santos AAC, Watkins DI, Galler R, Bonaldo MC. Improved genetic stability of recombinant yellow fever 17D virus expressing a lentiviral Gag gene fragment. Virology 2014; 452-453:202-11. [PMID: 24606697 DOI: 10.1016/j.virol.2014.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/11/2013] [Accepted: 01/20/2014] [Indexed: 10/25/2022]
Abstract
We have previously designed a method to construct viable recombinant Yellow Fever (YF) 17D viruses expressing heterologous polypeptides including part of the Simian Immunodeficiency Virus (SIV) Gag protein. However, the expressed region, encompassing amino acid residues from 45 to 269, was genetically unstable. In this study, we improved the genetic stability of this recombinant YF 17D virus by introducing mutations in the IRES element localized at the 5' end of the SIV gag gene. The new stable recombinant virus elicited adaptive immune responses similar to those induced by the original recombinant virus. It is, therefore, possible to increase recombinant stability by removing functional motifs from the insert that may have deleterious effects on recombinant YF viral fitness.
Collapse
Affiliation(s)
- Marlon G Veloso de Santana
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil; Department of Pathology, University of Miami, Miller School of Medicine, United States of America
| | - Patrícia C C Neves
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Juliana Ribeiro dos Santos
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Noemia S Lima
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - Alexandre A C dos Santos
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil
| | - David I Watkins
- Department of Pathology, University of Miami, Miller School of Medicine, United States of America
| | - Ricardo Galler
- Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Myrna C Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro, Brazil.
| |
Collapse
|
24
|
Neves PCC, Santos JR, Tubarão LN, Bonaldo MC, Galler R. Early IFN-gamma production after YF 17D vaccine virus immunization in mice and its association with adaptive immune responses. PLoS One 2013; 8:e81953. [PMID: 24324734 PMCID: PMC3855709 DOI: 10.1371/journal.pone.0081953] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 10/18/2013] [Indexed: 01/01/2023] Open
Abstract
Yellow Fever vaccine is one of the most efficacious human vaccines ever made. The vaccine (YF 17D) virus induces polyvalent immune responses, with a mixed TH1/TH2 CD4(+) cell profile, which results in robust T CD8(+) responses and high titers of neutralizing antibody. In recent years, it has been suggested that early events after yellow fever vaccination are crucial to the development of adequate acquired immunity. We have previously shown that primary immunization of humans and monkeys with YF 17D virus vaccine resulted in the early synthesis of IFN-γ. Herein we have demonstrated, for the first time that early IFN-γ production after yellow fever vaccination is a feature also of murine infection and is much more pronounced in the C57BL/6 strain compared to the BALB/c strain. Likewise, in C57BL/6 strain, we have observed the highest CD8(+) T cells responses as well as higher titers of neutralizing antibodies and total anti-YF IgG. Regardless of this intense IFN-γ response in mice, it was not possible to see higher titers of IgG2a in relation to IgG1 in both mice lineages. However, IgG2a titers were positively correlated to neutralizing antibodies levels, pointing to an important role of IFN-γ in eliciting high quality responses against YF 17D, therefore influencing the immunogenicity of this vaccine.
Collapse
Affiliation(s)
- Patrícia C. C. Neves
- Vice-diretoria de Desenvolvimento Tecnológico, Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Juliana R. Santos
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Luciana N. Tubarão
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Myrna C. Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Ricardo Galler
- Vice-diretoria de Desenvolvimento Tecnológico, Instituto de Tecnologia em Imunobiológicos, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
25
|
HPV16 E5 peptide vaccine in treatment of cervical cancer in vitro and in vivo. ACTA ACUST UNITED AC 2013; 33:735-742. [DOI: 10.1007/s11596-013-1189-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/13/2013] [Indexed: 11/24/2022]
|
26
|
Recombinant yellow fever viruses elicit CD8+ T cell responses and protective immunity against Trypanosoma cruzi. PLoS One 2013; 8:e59347. [PMID: 23527169 PMCID: PMC3601986 DOI: 10.1371/journal.pone.0059347] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 02/13/2013] [Indexed: 12/19/2022] Open
Abstract
Chagas’ disease is a major public health problem affecting nearly 10 million in Latin America. Despite several experimental vaccines have shown to be immunogenic and protective in mouse models, there is not a current vaccine being licensed for humans or in clinical trial against T. cruzi infection. Towards this goal, we used the backbone of Yellow Fever (YF) 17D virus, one of the most effective and well-established human vaccines, to express an immunogenic fragment derived from T. cruzi Amastigote Surface Protein 2 (ASP-2). The cDNA sequence of an ASP-2 fragment was inserted between E and NS1 genes of YF 17D virus through the construction of a recombinant heterologous cassette. The replication ability and genetic stability of recombinant YF virus (YF17D/ENS1/Tc) was confirmed for at least six passages in Vero cells. Immunogenicity studies showed that YF17D/ENS1/Tc virus elicited neutralizing antibodies and gamma interferon (IFN-γ) producing-cells against the YF virus. Also, it was able to prime a CD8+ T cell directed against the transgenic T. cruzi epitope (TEWETGQI) which expanded significantly as measured by T cell-specific production of IFN-γ before and after T. cruzi challenge. However, most important for the purposes of vaccine development was the fact that a more efficient protective response could be seen in mice challenged after vaccination with the YF viral formulation consisting of YF17D/ENS1/Tc and a YF17D recombinant virus expressing the TEWETGQI epitope at the NS2B-3 junction. The superior protective immunity observed might be due to an earlier priming of epitope-specific IFN-γ-producing T CD8+ cells induced by vaccination with this viral formulation. Our results suggest that the use of viral formulations consisting of a mixture of recombinant YF 17D viruses may be a promising strategy to elicit protective immune responses against pathogens, in general.
Collapse
|
27
|
Abstract
γδ T cells are a unique and conserved population of lymphocytes that have been the subject of a recent explosion of interest owing to their essential contributions to many types of immune response and immunopathology. But what does the integration of recent and long-established studies really tell us about these cells and their place in immunology? The time is ripe to consider the evidence for their unique and crucial functions. We conclude that whereas B cells and αβ T cells are commonly thought to contribute primarily to the antigen-specific effector and memory phases of immunity, γδ T cells are distinct in that they combine conventional adaptive features (inherent in their T cell receptors and pleiotropic effector functions) with rapid, innate-like responses that can place them in the initiation phase of immune reactions. This underpins a revised perspective on lymphocyte biology and the regulation of immunogenicity.
Collapse
|
28
|
Martins MA, Bonaldo MC, Rudersdorf RA, Piaskowski SM, Rakasz EG, Weisgrau KL, Furlott JR, Eernisse CM, Veloso de Santana MG, Hidalgo B, Friedrich TC, Chiuchiolo MJ, Parks CL, Wilson NA, Allison DB, Galler R, Watkins DI. Immunogenicity of seven new recombinant yellow fever viruses 17D expressing fragments of SIVmac239 Gag, Nef, and Vif in Indian rhesus macaques. PLoS One 2013; 8:e54434. [PMID: 23336000 PMCID: PMC3545953 DOI: 10.1371/journal.pone.0054434] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 12/11/2012] [Indexed: 12/31/2022] Open
Abstract
An effective vaccine remains the best solution to stop the spread of human immunodeficiency virus (HIV). Cellular immune responses have been repeatedly associated with control of viral replication and thus may be an important element of the immune response that must be evoked by an efficacious vaccine. Recombinant viral vectors can induce potent T-cell responses. Although several viral vectors have been developed to deliver HIV genes, only a few have been advanced for clinical trials. The live-attenuated yellow fever vaccine virus 17D (YF17D) has many properties that make it an attractive vector for AIDS vaccine regimens. YF17D is well tolerated in humans and vaccination induces robust T-cell responses that persist for years. Additionally, methods to manipulate the YF17D genome have been established, enabling the generation of recombinant (r)YF17D vectors carrying genes from unrelated pathogens. Here, we report the generation of seven new rYF17D viruses expressing fragments of simian immunodeficiency virus (SIV)mac239 Gag, Nef, and Vif. Studies in Indian rhesus macaques demonstrated that these live-attenuated vectors replicated in vivo, but only elicited low levels of SIV-specific cellular responses. Boosting with recombinant Adenovirus type-5 (rAd5) vectors resulted in robust expansion of SIV-specific CD8+ T-cell responses, particularly those targeting Vif. Priming with rYF17D also increased the frequency of CD4+ cellular responses in rYF17D/rAd5-immunized macaques compared to animals that received rAd5 only. The effect of the rYF17D prime on the breadth of SIV-specific T-cell responses was limited and we also found evidence that some rYF17D vectors were more effective than others at priming SIV-specific T-cell responses. Together, our data suggest that YF17D – a clinically relevant vaccine vector – can be used to prime AIDS virus-specific T-cell responses in heterologous prime boost regimens. However, it will be important to optimize rYF17D-based vaccine regimens to ensure maximum delivery of all immunogens in a multivalent vaccine.
Collapse
MESH Headings
- AIDS Vaccines/genetics
- AIDS Vaccines/immunology
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Epitopes, T-Lymphocyte/immunology
- Female
- Gene Order
- Gene Products, gag/genetics
- Gene Products, gag/immunology
- Gene Products, nef/genetics
- Gene Products, nef/immunology
- Gene Products, vif/genetics
- Gene Products, vif/immunology
- Genetic Vectors/genetics
- Humans
- Immunization
- Immunization, Secondary
- Kinetics
- Macaca mulatta
- Male
- Simian Immunodeficiency Virus/genetics
- Simian Immunodeficiency Virus/immunology
- T-Lymphocytes/immunology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Virus Replication
- Yellow fever virus/genetics
Collapse
Affiliation(s)
- Mauricio A. Martins
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Myrna C. Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz – FIOCRUZ, Rio de Janeiro, Brazil
| | - Richard A. Rudersdorf
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shari M. Piaskowski
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kim L. Weisgrau
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jessica R. Furlott
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Christopher M. Eernisse
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | | | - Bertha Hidalgo
- Section on Statistical Genetics, Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Thomas C. Friedrich
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Maria J. Chiuchiolo
- International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn Army Terminal, Brooklyn, New York, United States of America
| | - Christopher L. Parks
- International AIDS Vaccine Initiative, AIDS Vaccine Design and Development Laboratory, Brooklyn Army Terminal, Brooklyn, New York, United States of America
| | - Nancy A. Wilson
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - David B. Allison
- Section on Statistical Genetics, Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Ricardo Galler
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz – FIOCRUZ, Rio de Janeiro, Brazil
| | - David I. Watkins
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail:
| |
Collapse
|
29
|
|
30
|
Singleterry WL, Henderson H, Cruse JM. Depletion of pro-inflammatory CD161(+) double negative (CD3(+)CD4(-)CD8(-)) T cells in AIDS patients is ameliorated by expansion of the γδ T cell population. Exp Mol Pathol 2011; 92:155-9. [PMID: 22100877 DOI: 10.1016/j.yexmp.2011.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 11/02/2011] [Indexed: 11/28/2022]
Abstract
In this present investigation, flow cytometry was utilized to evaluate 13 healthy controls and 31 HIV-1 infected patients who had advanced to the AIDS stage of infection (CD4 count below 200 cells/mm(3)), for the expression of CD161 on CD3(+) double negative (DN) (CD3(+)CD4(-)CD8(-)) T cells, CD4(+) T cells, CD8(+) T cells and γδ T cells. The observed depletion of CD161(+) T cells from peripheral circulation was due primarily to the loss of CD4(+)CD161(+) T cells; as these cells represented 8.67±0.74% of the total healthy control peripheral T cell population, while the CD4(+)CD161(+) T cells of the AIDS group represented only 3.35±0.41% (p=<0.0001) of the total peripheral T cell population. We have also shown here that the DN T cell population was more than doubled in the AIDS group, with the DN T cell population expanding from 3.29±0.45% of the healthy control peripheral T cell population to 8.64±1.16% (p=0.0001) of the AIDS group peripheral T cell population. By evaluating the expression of CD161 on the surface of the DN T cells we showed that within the healthy control group, 47.4±4.99% of the DN T cells were positive for the expression of CD161, while only 26.4±3.54% (p=0.002) of the AIDS group's DN T cells expressed CD161. Despite CD161 expression being halved on the DN T cells of the AIDS group, when we compared the total peripheral T cell percentage of CD161(+) DN T cells between the healthy control group and the AIDS group, there was no statistical difference. Even though only 26.4% DN T cells within the AIDS group were positive for CD161(+), the overall DN T cell population had expanded to such an extent that there was no statistical difference between the groups with regard to CD161(+) DN T cells as a percentage of the total peripheral T cell population. Furthermore, we showed that within the DN T cell population, there was an approximate 2:1 ratio of γδ to αβ T cells, and this ratio was maintained in both the healthy control group and the AIDS group. While evaluating γδ T cells we also discovered that CD8(+) γδ T cells were expanded from 0.62±.09% of the healthy control peripheral T cell population to 5.01±.88% (p=<0.0001) of the peripheral T cell population of the AIDS group; and that this population of CD8(+) γδ T cells underwent the same reduction in percentage of cells expressing CD161(+), further demonstrated that the phenomenon of CD161(+) percentage reduction and compensatory increase in total cell population was affecting the entire circulating γδ T cell population.
Collapse
Affiliation(s)
- Will L Singleterry
- University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, United States.
| | | | | |
Collapse
|