1
|
Himmler GE, Mladinich MC, Conde JN, Gorbunova EE, Lindner MR, Kim HK, Mackow ER. Passage-attenuated Powassan virus LI9P protects mice from lethal LI9 challenge and links envelope residue D308 to neurovirulence. mBio 2025; 16:e0006525. [PMID: 39998203 PMCID: PMC11980571 DOI: 10.1128/mbio.00065-25] [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: 01/14/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Powassan virus (POWV) is an emergent tick-borne flavivirus that causes lethal encephalitic disease and chronic neurologic deficits in surviving patients. POWV-LI9 is a tick-derived isolate that causes neurovirulent disease and age-dependent lethality in mice. Serial passage of VeroE6 cells infected with LI9 resulted in eight amino acid changes in a POWV strain LI9P. LI9P fails to cause neurological sequelae, or lethality in C57BL/6 mice yet elicits neutralizing POWV antibody responses and protects mice from lethal LI9 challenge. Analysis revealed that LI9, but not LI9P, is present at high levels in the CNS, suggesting that LI9P is restricted from neuroinvasion or CNS replication. LI9 and LI9P are distinguished by a D308N envelope change within a domain associated with cell attachment. We evaluated the roles of Env-Domain III residue changes in LI9 virulence and LI9P attenuation using recombinant POWVs (recPOWVs) generated by reverse genetics. Remarkably, mutating D308N in LI9 completely abolished viral lethality and neuroinvasion in 50-week-old mice, reflecting the avirulent phenotype of LI9P. Analysis of the reciprocal N308D change in LI9P only partially restored neuroinvasion and lethality to the LI9P-N308D mutant, indicating that further LI9P residue changes contribute to LI9P attenuation. Consistent with differences in neuroinvasion, we found that rapid LI9P RNA synthesis and corresponding early IFN induction may contribute to LI9P clearance. Collectively, these findings define D308 as a determinant of POWV neuroinvasion and lethality, suggest potential mechanisms for restricted LI9P CNS entry, and reveal passage-attenuated LI9P as a candidate POWV vaccine platform. IMPORTANCE Powassan virus (POWV) infection causes a 10% lethal encephalitis, resulting in chronic neurological symptoms in half of survivors. POWV is transmitted in as short as 15 min following tick attachment, demonstrating the need for the development of POWV vaccines and therapeutics. Mechanisms of POWV neurovirulence remain to be defined to inform vaccine and therapeutic design. Cell culture passage has successfully been used to generate live-attenuated flavivirus vaccines. Accordingly, we serially passaged POWV LI9-infected VeroE6 cells and isolated an attenuated POWV strain, LI9P, that fails to cause neurologic sequelae or murine lethality. LI9P elicits neutralizing antibody responses, protects mice from a lethal WT POWV challenge, and is a potential POWV vaccine. Analysis of attenuating mutations in LI9P revealed that changing envelope residue D308N alone in LI9 prevents POWV neurovirulence and lethality in immunocompetent mice. Altogether, this study defines viral determinants of POWV pathogenesis and attenuating mutations that inform the development of live-attenuated POWV vaccines.
Collapse
MESH Headings
- Animals
- Encephalitis Viruses, Tick-Borne/genetics
- Encephalitis Viruses, Tick-Borne/pathogenicity
- Encephalitis Viruses, Tick-Borne/immunology
- Encephalitis Viruses, Tick-Borne/growth & development
- Virulence
- Mice
- Mice, Inbred C57BL
- Encephalitis, Tick-Borne/prevention & control
- Encephalitis, Tick-Borne/virology
- Encephalitis, Tick-Borne/immunology
- Encephalitis, Tick-Borne/pathology
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/administration & dosage
- Disease Models, Animal
- Chlorocebus aethiops
- Serial Passage
- Vero Cells
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Female
- Viral Envelope
Collapse
Affiliation(s)
- Grace E. Himmler
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Megan C. Mladinich
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
- Department of Biological Sciences, SUNY Old Westbury, Old Westbury, New York, USA
| | - Jonas N. Conde
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Elena E. Gorbunova
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Marissa R. Lindner
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Hwan Keun Kim
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Erich R. Mackow
- Department of Microbiology and Immunology, Center for Infectious Disease, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
2
|
Barrett J, Muntengesa E, Warrell C, Rampling T, Owen J, Patel D, Bhagani S, Moores R, Scobie A. Sofosbuvir as post-exposure prophylaxis for yellow fever-associated viscerotropic disease (YEL-AVD). J Antimicrob Chemother 2025; 80:825-827. [PMID: 39821333 DOI: 10.1093/jac/dkae484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025] Open
Abstract
OBJECTIVES Yellow fever-associated viscerotropic disease (YEL-AVD) is a rare but serious complication arising from administration of live-attenuated yellow fever vaccine to individuals with risk factors such as thymectomy. At present there is no evidence-based treatment, and case fatality rates are high. Sofosbuvir, an NS5B nucleotide inhibitor, has activity against yellow fever virus in vitro and in vivo. PATIENT AND METHODS Here we describe clinical and virological response to use of off-licence sofosbuvir as post-exposure prophylaxis for a patient inadvertently given yellow fever vaccine despite previous thymectomy. RESULTS A 14-day course of oral sofosbuvir was administered in an outpatient setting with regular clinical and biochemical monitoring. The patient remained well without developing clinical features of YEL-AVD and did not experience adverse effects from the treatment. CONCLUSIONS This supports the use of sofosbuvir as post-exposure prophylaxis in patients at high risk of developing YEL-AVD. Ongoing trials of efficacy of sofosbuvir in yellow fever infection may result in stronger support for this approach in the future.
Collapse
Affiliation(s)
- Jessica Barrett
- Department of Infection, North Bristol NHS Trust, Bristol, UK
| | - Ernest Muntengesa
- Department of Infectious Diseases, Royal Free London NHS Foundation Trust, London, UK
| | - Clare Warrell
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Salisbury, UK
| | - Tommy Rampling
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Salisbury, UK
| | - Jodie Owen
- Rare and Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, Salisbury, UK
| | - Dipti Patel
- National Travel Health Network and Centre (NaTHNaC), UCLH NHS Foundation Trust, London, UK
| | - Sanjay Bhagani
- Department of Infectious Diseases, Royal Free London NHS Foundation Trust, London, UK
| | - Rachel Moores
- Department of Infectious Diseases, Royal Free London NHS Foundation Trust, London, UK
| | - Antonia Scobie
- Department of Infectious Diseases, Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
3
|
Farnsworth MG, Khanipov K, Botnar K, Weaver SC, Barrett ADT, Golovko G. Real-world evidence of yellow Fever vaccination data-driven study. Vaccine 2025; 48:126758. [PMID: 39848130 DOI: 10.1016/j.vaccine.2025.126758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/16/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
Yellow Fever (YF), a mosquito-borne viral disease caused by yellow fever virus (YFV), remains endemic in tropical Sub-Saharan Africa and South America. The 17D live-attenuated vaccine has significantly reduced YF incidence with minimal risk of vaccine-associated adverse events, including Yellow Fever Vaccine-fever-associated Neurotropic Disease (YEL-AND) and Yellow Fever Vaccine-Associated Viscerotropic Disease (YEL-AVD). This study investigates the potential of Real-World Evidence (RWE) to enhance vaccine surveillance by analyzing electronic health records (EHRs) from the TriNetX platform, which identified a total of 15,835 individuals who were vaccinated with the Stamaril® YF vaccine between 2017 and 2021 in the United States. We compared adverse event rates obtained from RWE with those reported by the manufacturer in a recent study of Stamaril® used in the United States during this period. Our findings were consistent with those published previously and suggest no significant increase in adverse medical outcomes post-vaccination across all age groups, particularly in long-term analysis. This proof-of-concept study underscores the value of RWE in monitoring vaccine safety and supports its potential to complement traditional surveillance methods, offering a robust tool for continuous post-marketing vaccine evaluation.
Collapse
Affiliation(s)
- Madison G Farnsworth
- Department of Human Pathophysiology and Translational Medicine, Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, United States.
| | - Kamil Khanipov
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Kostiantyn Botnar
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| | - Scott C Weaver
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Alan D T Barrett
- Sealy Institute for Vaccine Sciences and Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - George Golovko
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
4
|
Gervais A, Marchal A, Fortova A, Berankova M, Krbkova L, Pychova M, Salat J, Zhao S, Kerrouche N, Le Voyer T, Stiasny K, Raffl S, Schieber Pachart A, Fafi-Kremer S, Gravier S, Robbiani DF, Abel L, MacDonald MR, Rice CM, Weissmann G, Kamal Eldin T, Robatscher E, Erne EM, Pagani E, Borghesi A, Puel A, Bastard P, Velay A, Martinot M, Hansmann Y, Aberle JH, Ruzek D, Cobat A, Zhang SY, Casanova JL. Autoantibodies neutralizing type I IFNs underlie severe tick-borne encephalitis in ∼10% of patients. J Exp Med 2024; 221:e20240637. [PMID: 39316018 PMCID: PMC11448868 DOI: 10.1084/jem.20240637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/10/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
Tick-borne encephalitis (TBE) virus (TBEV) is transmitted to humans via tick bites. Infection is benign in >90% of the cases but can cause mild (<5%), moderate (<4%), or severe (<1%) encephalitis. We show here that ∼10% of patients hospitalized for severe TBE in cohorts from Austria, Czech Republic, and France carry auto-Abs neutralizing IFN-α2, -β, and/or -ω at the onset of disease, contrasting with only ∼1% of patients with moderate and mild TBE. These auto-Abs were found in two of eight patients who died and none of 13 with silent infection. The odds ratios (OR) for severe TBE in individuals with these auto-Abs relative to those without them in the general population were 4.9 (95% CI: 1.5-15.9, P < 0.0001) for the neutralization of only 100 pg/ml IFN-α2 and/or -ω, and 20.8 (95% CI: 4.5-97.4, P < 0.0001) for the neutralization of 10 ng/ml IFN-α2 and -ω. Auto-Abs neutralizing type I IFNs accounted for ∼10% of severe TBE cases in these three European cohorts.
Collapse
Affiliation(s)
- Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| | - Astrid Marchal
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
| | - Andrea Fortova
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Science, České Budějovice, Czech Republic
| | - Michaela Berankova
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Science, České Budějovice, Czech Republic
| | - Lenka Krbkova
- Department of Children’s Infectious Diseases, University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martina Pychova
- Department of Infectious Diseases, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jiri Salat
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Science, České Budějovice, Czech Republic
| | - Shuxiang Zhao
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Nacim Kerrouche
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Clinical Immunology Department, Assistance Publique Hôpitaux de Paris (AP-HP), Saint-Louis Hospital, Paris, France
| | - Karin Stiasny
- Medical University of Vienna, Center for Virology, Vienna, Austria
| | - Simon Raffl
- Medical University of Vienna, Center for Virology, Vienna, Austria
| | | | - Samira Fafi-Kremer
- Institut de Virologie, Strasbourg University Hospital, Strasbourg University, INSERM Unité Mixte de Recherche (UMR) S1109, Strasbourg, France
| | - Simon Gravier
- Infectious Diseases Department, Hôpitaux Civils, Colmar, France
| | - Davide F. Robbiani
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Margaret R. MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Gaia Weissmann
- Department of Pediatrics and Neonatology, F. Tappeiner Hospital, Merano, Italy
| | - Tarek Kamal Eldin
- Infectious Disease Unit, Provincial Hospital of Bolzano (SABES-ASDAA), Lehrkrankenhaus der Paracelsus Medizinischen Privatuniversität, Bolzano, Italy
| | - Eva Robatscher
- Laboratory of Microbiology and Virology, SABES-ASDAA, Lehrkrankenhaus der Paracelsus Medizinischen Privatuniversität, Bolzano, Italy
| | - Elke Maria Erne
- Infectious Disease Unit, Provincial Hospital of Bolzano (SABES-ASDAA), Lehrkrankenhaus der Paracelsus Medizinischen Privatuniversität, Bolzano, Italy
| | - Elisabetta Pagani
- Laboratory of Microbiology and Virology, SABES-ASDAA, Lehrkrankenhaus der Paracelsus Medizinischen Privatuniversität, Bolzano, Italy
| | - Alessandro Borghesi
- Neonatal Intensive Care Unit, San Matteo Research Hospital, Pavia, Italy
- School of Life Sciences, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Aurélie Velay
- Institut de Virologie, Strasbourg University Hospital, Strasbourg University, INSERM Unité Mixte de Recherche (UMR) S1109, Strasbourg, France
| | - Martin Martinot
- Infectious Diseases Department, Hôpitaux Civils, Colmar, France
| | - Yves Hansmann
- CHU de Strasbourg, Service des Maladies Infectieuses et Tropicales, Strasbourg, France
| | - Judith H. Aberle
- Medical University of Vienna, Center for Virology, Vienna, Austria
| | - Daniel Ruzek
- Laboratory of Emerging Viral Diseases, Veterinary Research Institute, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Institute of Parasitology, Biology Centre of the Czech Academy of Science, České Budějovice, Czech Republic
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, Paris, France
| |
Collapse
|
5
|
Göbel S, Kazemi O, Ma J, Jordan I, Sandig V, Paulissen J, Kerstens W, Thibaut HJ, Reichl U, Dallmeier K, Genzel Y. Parallel Multifactorial Process Optimization and Intensification for High-Yield Production of Live YF17D-Vectored Zika Vaccine. Vaccines (Basel) 2024; 12:755. [PMID: 39066393 PMCID: PMC11281342 DOI: 10.3390/vaccines12070755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
The live-attenuated yellow fever 17D strain is a potent vaccine and viral vector. Its manufacture is based on embryonated chicken eggs or adherent Vero cells. Both processes are unsuitable for rapid and scalable supply. Here, we introduce a high-throughput workflow to identify suspension cells that are fit for the high-yield production of live YF17D-based vaccines in an intensified upstream process. The use of an automated parallel ambr15 microbioreactor system for screening and process optimization has led to the identification of two promising cell lines (AGE1.CR.pIX and HEKDyn) and the establishment of optimized production conditions, which have resulted in a >100-fold increase in virus titers compared to the current state of the art using adherent Vero cells. The process can readily be scaled up from the microbioreactor scale (15 mL) to 1 L stirred tank bioreactors. The viruses produced are genetically stable and maintain their favorable safety and immunogenicity profile, as demonstrated by the absence of neurovirulence in suckling BALB/c mice and consistent seroprotection in AG129 mice. In conclusion, the presented workflow allows for the rapid establishment of a robust, scalable, and high-yield process for the production of live-attenuated orthoflavivirus vaccines, which outperforms current standards. The approach described here can serve as a model for the development of scalable processes and the optimization of yields for other virus-based vaccines that face challenges in meeting growing demands.
Collapse
Affiliation(s)
- Sven Göbel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106 Magdeburg, Germany; (S.G.)
| | - Ozeir Kazemi
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery (MVVD), 3000 Leuven, Belgium (K.D.)
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery (MVVD), 3000 Leuven, Belgium (K.D.)
| | | | | | - Jasmine Paulissen
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), 3000 Leuven, Belgium
| | - Winnie Kerstens
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), 3000 Leuven, Belgium
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), 3000 Leuven, Belgium
| | - Udo Reichl
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106 Magdeburg, Germany; (S.G.)
- Bioprocess Engineering, Otto-von-Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology & Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery (MVVD), 3000 Leuven, Belgium (K.D.)
| | - Yvonne Genzel
- Bioprocess Engineering, Max Planck Institute for Dynamics of Complex Technical Systems, Sandtorstr. 1, 39106 Magdeburg, Germany; (S.G.)
| |
Collapse
|
6
|
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
|
7
|
Le Hir A, Durand GA, Boucraut J, Garnier A, Mura M, Diamantis S, Carles M, Durand C, Schweitzer C, Audouard C, Decroix V, Boyez R, Van Dendriessche A, Leclancher A, Kaphan E, Barbat du Closel L, Verdon R, du Cheyron D, Vabret A, Vergnon D, Grard G, Charrel R, de Lamballerie X, Eldin C. Yellow fever vaccine-associated neurologic and viscerotropic disease: a 10-year case series of the French National Reference Center for Arboviruses with clinical and immunological insights. J Travel Med 2024; 31:taad160. [PMID: 38123499 DOI: 10.1093/jtm/taad160] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Immunization against the Yellow fever virus (YFV) with the 17D live-attenuated vaccine is the most effective way to prevent the disease. However, unexpected severe adverse events can occur. They consist in a neurological impairment - neurological disease (YEL-AND), a YF-like illness - viscerotropic disease (YEL-AVD) or anaphylaxis. In this article, we describe the epidemiology, clinical and biological features of YEL-AND and YEL-AVD cases reported to the French National Reference Center for Arboviruses (NRCA) in the past 10 years. METHODS We conducted a national, retrospective study using the database of the NRCA from June 2012 to June 2022. All patients whose biological samples were sent to the NRCA for detection of YFV by serology and/or RT-qPCR for a suspected vaccine-associated adverse event were included. We collected data by reading medical records and conducted complementary neuro-immunological analysis, followed by a search for autoimmunity against type-1-interferon when samples were available at the NRCA. RESULTS There were 10 cases of YEL-AND and 2 cases of YEL-AVD reported to the NRCA in the past 10 years, which represented an overall incidence of 0.6 for 100 000 doses. A total of 6/12 cases were previously healthy patients (50%, mean age 31 years), and 4/12 cases had cardiovascular co-morbidities (42%, mean age 56 years). The majority of YEL-AND had a favourable outcome at 6 months of follow up. One YEL-AVD patient passed. In secondary analyses, we evidenced a significant blood cerebrospinal fluid (CSF) barrier dysfunction, without intrathecal synthesis of immunoglobulin and without argument for a neuron damage. We further detected a significant rate of anti-type-1alpha interferon antibodies in 3/10 tested patients (2 YEL-AND and 1 YEL-AVD). CONCLUSION YEL-AND and YEL-AVD are rare events that can underlie defect in the innate immunity of apparently healthy or mild co-morbid subjects. Outcome was generally favourable in the YEL-AND cases of our series, but still life-threatening or even fatal in the YEL-AVD cases.
Collapse
Affiliation(s)
- Anne Le Hir
- Assistance Publique des Hôpitaux de Marseille, Marseille 13005, France
| | - Guillaume A Durand
- Unité des Virus Émergents (UVE: Aix-Marseille Univ-IRD 190-Inserm 1207), Marseille 13005, France
- National Reference Center for Arboviruses, National Institute of Health and Medical Research (Inserm) and French Armed Forces Biomedical Research Institute (IRBA), Marseille 13005, France
| | - José Boucraut
- Laboratoire d'Immunologie, Assistance-Publique des Hôpitaux de Marseille, Marseille 13005, France
- Institut de Neurosciences des Systèmes (INS, UMR1106), Marseille 13005, France
| | - Annabelle Garnier
- French Armed Forces Biomedical Research Institute, Brétigny-sur-Orge 91220, France
| | - Marie Mura
- French Armed Forces Biomedical Research Institute, Brétigny-sur-Orge 91220, France
- Institut Pasteur, Laboratoire d'innovation: vaccins, Paris 75015, France
| | - Sylvain Diamantis
- Infectious Diseases Unit, Groupe Hospitalier Sud Ile de France, Melun 77000, France
- DYNAMIC Research Unit, Université Paris-Est-Creteil, Thiais 94320, France
| | - Michel Carles
- Service de Maladies Infectieuses et Tropicales, CHU de Nice 06200, France
| | - Claire Durand
- Service de Maladies Infectieuses et Tropicales, CHU de Nice 06200, France
| | - Cyril Schweitzer
- Service de Médecine Infantile, Hôpital d'enfants, CHRU de Nancy, Vandœuvre-lès-Nancy 54500, France
- DeVAH EA 3450, Université de Lorraine, Faculté de Médecine de Nancy, Vandoeuvre lès Nancy 54500, France
| | - Claire Audouard
- Service de Médecine Infantile, Hôpital d'enfants, CHRU de Nancy, Vandœuvre-lès-Nancy 54500, France
| | - Véronique Decroix
- Laboratoire de biologie médicale, CH de Saint-Quentin, Saint-Quentin 02100, France
| | - Romain Boyez
- Service de neurologie, CH de Lunéville, Lunéville 54300, France
| | - Anne Van Dendriessche
- Service de médecine interne et maladies infectieuses, Groupe Hospitalier du Havre, Montivilliers 76290, France
| | | | - Elsa Kaphan
- Pôle de Médecine Oncologie, Service de médecine interne, CHU Conception, Assistance Publique Hôpitaux de Marseille, Marseille 13005, France
| | - Luce Barbat du Closel
- Service de Neurologie, CHU Timone, Assistance Publique des Hôpitaux de Marseille, Marseille 13005, France
| | - Renaud Verdon
- Service de maladies infectieuses et tropicales, CHU Côte-de-Nacre, Caen 14000, France
| | - Damien du Cheyron
- Service de médecine intensive et de réanimation, CHU de Caen, Caen 14000, France
| | - Astrid Vabret
- INSERM, DYNAMICURE UMR1311, CHU Caen, Department of Virology, Univ de Caen Normandie, Univ Rouen Normandie, Caen 14000, France
| | | | - Gilda Grard
- Unité des Virus Émergents (UVE: Aix-Marseille Univ-IRD 190-Inserm 1207), Marseille 13005, France
- National Reference Center for Arboviruses, National Institute of Health and Medical Research (Inserm) and French Armed Forces Biomedical Research Institute (IRBA), Marseille 13005, France
| | - Rémi Charrel
- Unité des Virus Émergents (UVE: Aix-Marseille Univ-IRD 190-Inserm 1207), Marseille 13005, France
- Comité de Lutte contre les Infections Nosocomiales (CLIN), Hôpital Nord, Assistance Publique des Hôpitaux de Marseille, Marseille 13005, France
- Laboratoire des Infections Virales Aigues et Tropicales, Pôle des Maladies Infectieuses, Assistance Publique des Hôpitaux de Marseille, Marseille 13005, France
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE: Aix-Marseille Univ-IRD 190-Inserm 1207), Marseille 13005, France
- National Reference Center for Arboviruses, National Institute of Health and Medical Research (Inserm) and French Armed Forces Biomedical Research Institute (IRBA), Marseille 13005, France
| | - Carole Eldin
- Comité de Lutte contre les Infections Nosocomiales (CLIN), Hôpital Nord, Assistance Publique des Hôpitaux de Marseille, Marseille 13005, France
| |
Collapse
|
8
|
Fam BSDO, Feira MF, Cadore NA, Sbruzzi R, Hünemeier T, Abel L, Zhang Q, Casanova JL, Vianna FSL. Human genetic determinants of COVID-19 in Brazil: challenges and future plans. Genet Mol Biol 2024; 46:e20230128. [PMID: 38226654 PMCID: PMC10792479 DOI: 10.1590/1678-4685-gmb-2023-0128] [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: 04/28/2023] [Accepted: 11/14/2023] [Indexed: 01/17/2024] Open
Abstract
COVID-19 pandemic represented a worldwide major challenge in different areas, and efforts undertaken by the scientific community led to the understanding of some of the genetic determinants that influence the different COVID-19 outcomes. In this paper, we review the studies about the role of human genetics in COVID-19 severity and how Brazilian studies also contributed to those findings. Rare variants in genes related to Inborn Errors of Immunity (IEI) in the type I interferons pathway, and its phenocopies, have been described as being causative of severe outcomes. IEI and its phenocopies are present in Brazil, not only in COVID-19 patients, but also in autoimmune conditions and severe reactions to yellow fever vaccine. In addition, studies focusing on common variants and GWAS studies encompassing worldwide patients have found several loci associated with COVID-19 severity. A GWAS study including only Brazilian COVID-19 patients identified a new locus 1q32.1 associated with COVID-19 severity. Thus, more comprehensive studies considering the Brazilian genomic diversity should be performed, since they can help to reveal not only what are the genetic determinants that contribute to the different outcomes for COVID-19 in the Brazilian population, but in the understanding of human genetics in different health conditions.
Collapse
Affiliation(s)
- Bibiana S. de Oliveira Fam
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Instituto de Biociências, Departamento de Genética, Laboratório de Imunogenética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
- Instituto Nacional de Genética Médica Populacional (INaGeMP), Porto Alegre, RS, Brazil
| | - Marilea Furtado Feira
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Instituto de Biociências, Departamento de Genética, Laboratório de Imunogenética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
- Instituto Nacional de Genética Médica Populacional (INaGeMP), Porto Alegre, RS, Brazil
| | - Nathan Araujo Cadore
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Instituto de Biociências, Departamento de Genética, Laboratório de Imunogenética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
- Instituto Nacional de Genética Médica Populacional (INaGeMP), Porto Alegre, RS, Brazil
| | - Renan Sbruzzi
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Instituto de Biociências, Departamento de Genética, Laboratório de Imunogenética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
- Instituto Nacional de Genética Médica Populacional (INaGeMP), Porto Alegre, RS, Brazil
| | - Tábita Hünemeier
- Universidade de São Paulo, Instituto de Biociências, Departamento de Genética e Biologia Evolutiva, São Paulo, SP, Brazil
- Institut de Biologia Evolutiva (Consejo Superior de Investigaciones Científicas/Universitat Pompeu Fabra), Barcelona, Spain
| | - Laurent Abel
- The Rockefeller University, Rockefeller Branch, St. Giles Laboratory of Human Genetics of Infectious Diseases, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Imagine Institute, Paris, France
- University Paris Cité, Imagine Institute, Paris, France
| | - Qian Zhang
- The Rockefeller University, Rockefeller Branch, St. Giles Laboratory of Human Genetics of Infectious Diseases, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| | - Jean-Laurent Casanova
- The Rockefeller University, Rockefeller Branch, St. Giles Laboratory of Human Genetics of Infectious Diseases, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Imagine Institute, Paris, France
- University Paris Cité, Imagine Institute, Paris, France
- Necker Hospital for Sick Children, Department of Pediatrics, Paris, France
| | - Fernanda Sales Luiz Vianna
- Hospital de Clínicas de Porto Alegre, Centro de Pesquisa Experimental, Laboratório de Medicina Genômica, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul, Instituto de Biociências, Departamento de Genética, Laboratório de Imunogenética, Programa de Pós-Graduação em Genética e Biologia Molecular, Porto Alegre, RS, Brazil
- Instituto Nacional de Genética Médica Populacional (INaGeMP), Porto Alegre, RS, Brazil
| |
Collapse
|
9
|
Lemmens V, Kelchtermans L, Debaveye S, Chiu W, Vercruysse T, Ma J, Thibaut HJ, Neyts J, Sanchez-Felipe L, Dallmeier K. YF17D-vectored Ebola vaccine candidate protects mice against lethal surrogate Ebola and yellow fever virus challenge. NPJ Vaccines 2023; 8:99. [PMID: 37433816 PMCID: PMC10336040 DOI: 10.1038/s41541-023-00699-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 06/27/2023] [Indexed: 07/13/2023] Open
Abstract
Ebola virus (EBOV) and related filoviruses such as Sudan virus (SUDV) threaten global public health. Effective filovirus vaccines are available only for EBOV, yet restricted to emergency use considering a high reactogenicity and demanding logistics. Here we present YF-EBO, a live YF17D-vectored dual-target vaccine candidate expressing EBOV glycoprotein (GP) as protective antigen. Safety of YF-EBO in mice was further improved over that of parental YF17D vaccine. A single dose of YF-EBO was sufficient to induce high levels of EBOV GP-specific antibodies and cellular immune responses, that protected against lethal infection using EBOV GP-pseudotyped recombinant vesicular stomatitis virus (rVSV-EBOV) in interferon-deficient (Ifnar-/-) mice as surrogate challenge model. Concomitantly induced yellow fever virus (YFV)-specific immunity protected Ifnar-/- mice against intracranial YFV challenge. YF-EBO could thus help to simultaneously combat both EBOV and YFV epidemics. Finally, we demonstrate how to target other highly pathogenic filoviruses such as SUDV at the root of the 2022 outbreak in Uganda.
Collapse
Affiliation(s)
- Viktor Lemmens
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
| | - Lara Kelchtermans
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
| | - Sarah Debaveye
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
| | - Winston Chiu
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
| | - Thomas Vercruysse
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), BE-3000, Leuven, Belgium
- AstriVax, BE-3001, Heverlee, Belgium
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), BE-3000, Leuven, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium
- GVN, Global Virus Network, Baltimore, MD, USA
| | - Lorena Sanchez-Felipe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium.
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, BE-3000, Leuven, Belgium.
| |
Collapse
|
10
|
Development of a Bicistronic Yellow Fever Live Attenuated Vaccine with Reduced Neurovirulence and Viscerotropism. Microbiol Spectr 2022; 10:e0224622. [PMID: 35980184 PMCID: PMC9602263 DOI: 10.1128/spectrum.02246-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The yellow fever (YF) live attenuated vaccine strain 17D (termed 17D) has been widely used for the prevention and control of YF disease. However, 17D retains significant neurovirulence and viscerotropism in mice, which is probably linked to the increased occurrences of serious adverse events following 17D vaccination. Thus, the development of an updated version of the YF vaccine with an improved safety profile is of high priority. Here, we generated a viable bicistronic YF virus (YFV) by incorporating the internal ribosome entry site (IRES) from Encephalomyocarditis virus into an infectious clone of YFV 17D. The resulting recombinant virus, 17D-IRES, exhibited similar replication efficiency to its parental virus (17D) in mammalian cell lines, while it was highly restricted in mosquito cells. Serial passage of 17D-IRES in BHK-21 cells showed good genetic stability. More importantly, in comparison with the parental 17D, 17D-IRES displayed significantly decreased mouse neurovirulence and viscerotropism in type I interferon (IFN)-signaling-deficient and immunocompetent mouse models. Interestingly, 17D-IRES showed enhanced sensitivity to type I IFN compared with 17D. Moreover, immunization with 17D-IRES provided solid protection for mice against a lethal challenge with YFV. These preclinical data support further development of 17D-IRES as an updated version for the approved YF vaccine. This IRES-based attenuation strategy could be also applied to the design of live attenuated vaccines against other mosquito-borne flaviviruses. IMPORTANCE Yellow fever (YF) continually spreads and causes epidemics around the world, posing a great threat to human health. The YF live attenuated vaccine 17D is considered the most efficient vaccine available and helps to successfully control disease epidemics. However, side effects may occur after vaccination, such as viscerotropic disease (YEL-AVD) and neurotropic adverse disease (YEL-AND). Thus, there is an urgent need for a safer YF vaccine. Here, an IRES strategy was employed, and a bicistronic YFV was successfully developed (named 17D-IRES). 17D-IRES showed effective replication and genetic stability in vitro and high attenuation in vivo. Importantly, 17D-IRES induced humoral and cellular immune responses and conferred full protection against lethal YFV challenge. Our study provides data suggesting that 17D-IRES, with its prominent advantages, could be a vaccine candidate against YF. Moreover, this IRES-based bicistronic technology platform represents a promising strategy for developing other live attenuated vaccines against emerging viruses.
Collapse
|
11
|
Rasulova M, Vercruysse T, Paulissen J, Coun C, Suin V, Heyndrickx L, Ma J, Geerts K, Timmermans J, Mishra N, Li LH, Kum DB, Coelmont L, Van Gucht S, Karimzadeh H, Thorn-Seshold J, Rothenfußer S, Ariën KK, Neyts J, Dallmeier K, Thibaut HJ. A High-Throughput Yellow Fever Neutralization Assay. Microbiol Spectr 2022; 10:e0254821. [PMID: 35670599 PMCID: PMC9241659 DOI: 10.1128/spectrum.02548-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 05/19/2022] [Indexed: 11/20/2022] Open
Abstract
Quick and accurate detection of neutralizing antibodies (nAbs) against yellow fever is essential in serodiagnosis during outbreaks for surveillance and to evaluate vaccine efficacy in population-wide studies. All of this requires serological assays that can process a large number of samples in a highly standardized format. Albeit being laborious, time-consuming, and limited in throughput, the classical plaque reduction neutralization test (PRNT) is still considered the gold standard for the detection and quantification of nAbs due to its sensitivity and specificity. Here, we report the development of an alternative fluorescence-based serological assay (SNTFLUO) with an equally high sensitivity and specificity that is fit for high-throughput testing with the potential for automation. Finally, our novel SNTFLUO was cross-validated in several reference laboratories and against international WHO standards, showing its potential to be implemented in clinical use. SNTFLUO assays with similar performance are available for the Japanese encephalitis, Zika, and dengue viruses amenable to differential diagnostics. IMPORTANCE Fast and accurate detection of neutralizing antibodies (nAbs) against yellow fever virus (YFV) is key in yellow fever serodiagnosis, outbreak surveillance, and monitoring of vaccine efficacy. Although classical PRNT remains the gold standard for measuring YFV nAbs, this methodology suffers from inherent limitations such as low throughput and overall high labor intensity. We present a novel fluorescence-based serum neutralization test (SNTFLUO) with equally high sensitivity and specificity that is fit for processing a large number of samples in a highly standardized manner and has the potential to be implemented for clinical use. In addition, we present SNTFLUO assays with similar performance for Japanese encephalitis, Zika, and dengue viruses, opening new avenues for differential diagnostics.
Collapse
Affiliation(s)
- Madina Rasulova
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Thomas Vercruysse
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Jasmine Paulissen
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Catherina Coun
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Vanessa Suin
- Sciensano, Viral Diseases Service, Scientific Directorate of Infectious Diseases in Humans, Brussels, Belgium
| | - Leo Heyndrickx
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Katrien Geerts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Jolien Timmermans
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| | - Niraj Mishra
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Li-Hsin Li
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Dieudonné Buh Kum
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Lotte Coelmont
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Steven Van Gucht
- Sciensano, Viral Diseases Service, Scientific Directorate of Infectious Diseases in Humans, Brussels, Belgium
| | - Hadi Karimzadeh
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany
- Unit Clinical Pharmacology (EKliP), Helmholtz Center for Environmental Health, Munich, Germany
| | - Julia Thorn-Seshold
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany
- Unit Clinical Pharmacology (EKliP), Helmholtz Center for Environmental Health, Munich, Germany
| | - Simon Rothenfußer
- Division of Clinical Pharmacology, University Hospital, LMU Munich, Munich, Germany
- Unit Clinical Pharmacology (EKliP), Helmholtz Center for Environmental Health, Munich, Germany
| | - Kevin K. Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- Global Virus Network (GVN), Baltimore, Maryland, USA
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Virology and Chemotherapy, Molecular Vaccinology & Vaccine Discovery, Leuven, Belgium
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Translational Platform Virology and Chemotherapy (TPVC), Leuven, Belgium
| |
Collapse
|
12
|
de Abreu ADJL, Cavalcante JR, de Araújo Lagos LW, Caetano R, Braga JU. A Systematic Review and a Meta-Analysis of the Yellow Fever Vaccine in the Elderly Population. Vaccines (Basel) 2022; 10:711. [PMID: 35632466 PMCID: PMC9147422 DOI: 10.3390/vaccines10050711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022] Open
Abstract
We conducted a systematic review and a meta-analysis to assess the risk of serious adverse events in the elderly after yellow fever vaccination compared to the non-elderly population. We searched multiple databases and grey literature, and we selected research without language and publication date restrictions. Studies were analyzed in a descriptive way and meta-analyzed and expressed in terms of prevalence ratio and risk ratio with a 95% confidence interval, depending on the degree of heterogeneity found. A total of 18 studies were included and 11 were meta-analyzed. The results obtained through the meta-analysis showed a risk of serious adverse events after yellow fever vaccination three times higher for the elderly when compared to the non-elderly population and five times higher for persons > 70 years. In relation to adverse event types, viscerotropic disease associated with the yellow fever vaccine had a risk that was six times higher when compared to the population < 60 years. The evidence found supports that the vaccine indication in individuals > 60 years of age should be based on a careful analysis of individual benefit-risk assessments. The results found suggest a higher risk of events for individuals > 70 years, especially for viscerotropic and neurotropic disease associated with YFV contraindicating the use of the YFV in this age group.
Collapse
Affiliation(s)
| | - João Roberto Cavalcante
- Instituto de Medicina Social da Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20550-013, Brazil; (J.R.C.); (R.C.); (J.U.B.)
| | | | - Rosângela Caetano
- Instituto de Medicina Social da Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20550-013, Brazil; (J.R.C.); (R.C.); (J.U.B.)
| | - José Ueleres Braga
- Instituto de Medicina Social da Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20550-013, Brazil; (J.R.C.); (R.C.); (J.U.B.)
- Escola Nacional de Saúde Publica Sergio Arouca-FIOCRUZ, Rio de Janeiro 21041-21, Brazil
| |
Collapse
|
13
|
[Vaccinations for the immunologic memory-Repeatedly or once only?]. Internist (Berl) 2022; 63:476-483. [PMID: 35376975 PMCID: PMC8978485 DOI: 10.1007/s00108-022-01324-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2022] [Indexed: 12/02/2022]
Abstract
Dank der Wirksamkeit von Impfstoffen sind einige besonders bedrohliche Infektionskrankheiten selten geworden. Allerdings sind Impfstoffe inzwischen Opfer ihres eigenen Erfolgs. Aufgrund unzureichender Zustimmung und unzureichender Impfquoten besteht die Gefahr, dass die Wirksamkeit von Impfungen als Präventionsmaßnahme immer mehr schwindet. Im Jahr 2019 stufte die Weltgesundheitsorganisation daher Zweifel an der Wirksamkeit von Impfstoffen als eine der zehn größten Gesundheitsgefahren weltweit ein. Im vorliegenden Beitrag wird auf wichtige Fragen zu Impfungen und Impfstoffen sowie zu ihren Wirkungen im Wechselspiel mit dem Immunsystem eingegangen. Behandelt werden folgende Themen: Vergleich der natürlichen und der durch Impfung erworbenen Immunität; Faktoren, die eine Auffrischungsimpfung erforderlich machen; Rolle der Herdenimmunität; Voraussetzungen für das Gelingen einer Erkrankungseradikation; Einfluss verschiedener T‑Zellen auf die Impfwirkung; Rolle des Immungedächtnisses; Faktoren, die den Impfschutz beeinflussen; Impfungen bei Immundefizienz; Potenzial und Einsatzbereiche der passiven Immunisierung. Im Angesicht der Coronapandemie und der laufenden Impfkampagne ist zu hoffen, dass eine allgemeine Renaissance der Impfungen gegen Infektionskrankheiten einsetzt.
Collapse
|
14
|
Casanova JL, Zhang Q, Bastard P, Jouanguy E. In memoriam: Stephen J Seligman, MD : Adverse reactions to the yellow fever vaccine: from epidemiological risk factors to causes and mechanisms. J Clin Immunol 2022; 42:437-440. [PMID: 35028800 PMCID: PMC8758354 DOI: 10.1007/s10875-021-01204-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 12/19/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA. .,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France. .,University of Paris, Imagine Institute, Paris, France. .,Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, Paris, France. .,Howard Hughes Medical Institute, New York, NY, USA.
| | - Qian Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France
| | - Paul Bastard
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France.,Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Emmanuelle Jouanguy
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France
| |
Collapse
|
15
|
Ledlie S, Ricci C, Pan C, Rojas A, Khromava A, Li L. Yellow fever vaccine usage in the United States and risk of neurotropic and viscerotropic disease: A retrospective cohort study using three healthcare databases. Vaccine 2022; 40:742-751. [PMID: 34996642 DOI: 10.1016/j.vaccine.2021.12.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/10/2021] [Accepted: 12/20/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Yellow fever (YF) vaccines are highly effective and have a well-established safety profile despite the risk of rare serious adverse events (SAEs), vaccine-associated neurotropic (YEL-AND) and viscerotropic disease (YEL-AVD). This study aimed to describe US civilian YF vaccine usage, the population characteristics and pre-existing immunosuppressive medical conditions among those vaccinated, and to provide updated risk estimates of neurotropic and viscerotropic disease post-vaccination. METHODS A retrospective cohort study was conducted using de-identified patient information from Optum Electronic Healthcare Record (EHR) (2007-2019), Optum Clinformatics Data Mart (CDM) (2004-2019) and IBM MarketScan (2007-2019) databases. YF vaccine recipients were identified using relevant vaccination and procedural codes. Demographic characteristics and pre-existing medical conditions were described. Incidence proportions with 95% confidence intervals (CI) of neurotropic and viscerotropic diseases occurring ≤ 30 days post-vaccination, after exclusion of unlikely cases based on current clinical guidelines of YEL-AND and YEL-AVD, were calculated. RESULTS A total of 92,205, 46,539 and 125,235 YF vaccine recipients were retrieved from Optum EHR, Optum CDM and IBM MarketScan databases, respectively. The majority of vaccine recipients were aged < 60 years (highest proportion aged 18-29 years) with a higher proportion of females overall. Few vaccine recipients (<1%) had conditions predisposing them to immunosuppression. Four non-fatal cases of neurotropic disease and zero cases of viscerotropic disease were identified. The incidence proportion of post-vaccination neurotropic disease was 1.41 (95% CI: 0.15-6.61) and 3.04 (95% CI: 0.86-8.11) per 100,000 vaccine recipients in Optum EHR and IBM MarketScan, respectively, with no events identified in Optum CDM. CONCLUSIONS This study provides updated insights into current YF vaccine usage in US civilian recipients and supports the safety profile of YF vaccines in US practice. The low frequency of pre-existing immunosuppressive medical conditions among vaccine recipients suggests good adherence to vaccination guidelines by healthcare practitioners. The risk of developing neurotropic and viscerotropic disease post-vaccination remains rare.
Collapse
Affiliation(s)
- Shaleesa Ledlie
- Epidemiology and Benefit Risk, Sanofi Pasteur, North York, Ontario, Canada.
| | - Christina Ricci
- Epidemiology and Benefit Risk, Sanofi Pasteur, North York, Ontario, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada.
| | - Chunshen Pan
- Epidemiology and Benefit Risk, Sanofi US, Bridgewater, NJ, USA.
| | - Andrey Rojas
- Global Pharmacovigilance, Sanofi Pasteur, Bogota, Colombia.
| | - Alena Khromava
- Epidemiology and Benefit Risk, Sanofi Pasteur, North York, Ontario, Canada.
| | - Lin Li
- Epidemiology and Benefit Risk, Sanofi US, Bridgewater, NJ, USA.
| |
Collapse
|
16
|
Gao Z, Zhang X, Zhang L, Wu S, Ma J, Wang F, Zhou Y, Dai X, Bullitt E, Du Y, Guo JT, Chang J. A yellow fever virus NS4B inhibitor not only suppresses viral replication, but also enhances the virus activation of RIG-I-like receptor-mediated innate immune response. PLoS Pathog 2022; 18:e1010271. [PMID: 35061864 PMCID: PMC8809586 DOI: 10.1371/journal.ppat.1010271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 02/02/2022] [Accepted: 01/11/2022] [Indexed: 12/24/2022] Open
Abstract
Flavivirus infection of cells induces massive rearrangements of the endoplasmic reticulum (ER) membrane to form viral replication organelles (ROs) which segregates viral RNA replication intermediates from the cytoplasmic RNA sensors. Among other viral nonstructural (NS) proteins, available evidence suggests for a prominent role of NS4B, an ER membrane protein with multiple transmembrane domains, in the formation of ROs and the evasion of the innate immune response. We previously reported a benzodiazepine compound, BDAA, which specifically inhibited yellow fever virus (YFV) replication in cultured cells and in vivo in hamsters, with resistant mutation mapped to P219 of NS4B protein. In the following mechanistic studies, we found that BDAA specifically enhances YFV induced inflammatory cytokine response in association with the induction of dramatic structural alteration of ROs and exposure of double-stranded RNA (dsRNA) in virus-infected cells. Interestingly, the BDAA-enhanced cytokine response in YFV-infected cells is attenuated in RIG-I or MAD5 knockout cells and completely abolished in MAVS knockout cells. However, BDAA inhibited YFV replication at a similar extent in the parent cells and cells deficient of RIG-I, MDA5 or MAVS. These results thus provided multiple lines of biological evidence to support a model that BDAA interaction with NS4B may impair the integrity of YFV ROs, which not only inhibits viral RNA replication, but also promotes the release of viral RNA from ROs, which consequentially activates RIG-I and MDA5. Although the innate immune enhancement activity of BDAA is not required for its antiviral activity in cultured cells, its dual antiviral mechanism is unique among all the reported antiviral agents thus far and warrants further investigation in animal models in future. Emergence and re-emergence of yellow fever (YF) caused by the yellow fever virus (YFV) infection have posed a global public health threat in previously non-epidemic as well as endemic regions. The approximately 30% of mortality rate makes the outbreaks particularly devastating. In addition to the vaccination campaign and mosquito controls, antiviral drugs are important components in the toolbox for combating YF outbreaks. However, only two nucleotide analogue drugs developed for the treatment of other RNA virus infections are currently repurposed for the treatment of YF with uncertain clinical efficacy. BDAA is a benzodiazepine compound discovered as a potent YFV-specific antiviral agent in our laboratory. The work reported herein further demonstrates that BDAA interaction with the YFV NS4B protein may impair the integrity of viral RNA replication organelles, which not only inhibits viral RNA replication, but also results in the leakage of viral RNA into the cytoplasm to activate RIG-I-like RNA receptors and enhances the innate antiviral immune response. The unprecedented antiviral mechanism of BDAA highlights the essential role of the NS4B protein in viral RNA replication and the evasion of host cellular innate immunity.
Collapse
Affiliation(s)
- Zhao Gao
- Baruch S. Blumberg Institute, Doylestown, Pennsylvania, United States of America
| | - Xuexiang Zhang
- Baruch S. Blumberg Institute, Doylestown, Pennsylvania, United States of America
| | - Lin Zhang
- Baruch S. Blumberg Institute, Doylestown, Pennsylvania, United States of America
| | - Shuo Wu
- Baruch S. Blumberg Institute, Doylestown, Pennsylvania, United States of America
| | - Julia Ma
- Baruch S. Blumberg Institute, Doylestown, Pennsylvania, United States of America
| | - Fuxuan Wang
- Baruch S. Blumberg Institute, Doylestown, Pennsylvania, United States of America
| | - Yan Zhou
- Bioinformatics and Biostatistics Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Xinghong Dai
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, United States of America
| | - Esther Bullitt
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Yanming Du
- Baruch S. Blumberg Institute, Doylestown, Pennsylvania, United States of America
| | - Ju-Tao Guo
- Baruch S. Blumberg Institute, Doylestown, Pennsylvania, United States of America
| | - Jinhong Chang
- Baruch S. Blumberg Institute, Doylestown, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
17
|
Ecarnot F, Maggi S, Michel JP, Veronese N, Rossanese A. Vaccines and Senior Travellers. FRONTIERS IN AGING 2021; 2:677907. [PMID: 35822022 PMCID: PMC9261415 DOI: 10.3389/fragi.2021.677907] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/28/2021] [Indexed: 11/18/2022]
Abstract
Background: International tourist travel has been increasingly steadily in recent years, and looks set to reach unprecedented levels in the coming decades. Among these travellers, an increasing proportion is aged over 60 years, and is healthy and wealthy enough to be able to travel. However, senior travellers have specific risks linked to their age, health and travel patterns, as compared to their younger counterparts. Methods: We review here the risk of major vaccine-preventable travel-associated infectious diseases, and forms and efficacy of vaccination for these diseases. Results: Routine vaccinations are recommended for older persons, regardless of whether they travel or not (e.g., influenza, pneumococcal vaccines). Older individuals should be advised about the vaccines that are recommended for their age group in the framework of the national vaccination schedule. Travel-specific vaccines must be discussed in detail on a case-by-case basis, and the risk associated with the vaccine should be carefully weighed against the risk of contracting the disease during travel. Travel-specific vaccines reviewed here include yellow fever, hepatitis, meningococcal meningitis, typhoid fever, cholera, poliomyelitis, rabies, Japanese encephalitis, tick-borne encephalitis and dengue. Conclusion: The number of older people who have the good health and financial resources to travel is rising dramatically. Older travellers should be advised appropriately about routine and travel-specific vaccines, taking into account the destination, duration and purpose of the trip, the activities planned, the type of accommodation, as well as patient-specific characteristics, such as health status and current medications.
Collapse
Affiliation(s)
- Fiona Ecarnot
- University Hospital Besancon and University of Franche-Comté, Besancon, France
| | - Stefania Maggi
- CNR, Institute of Neuroscience – Aging Branch, Padua, Italy
| | - Jean-Pierre Michel
- Department of Rehabilitation and Geriatrics, University of Geneva, Geneva, Switzerland
| | - Nicola Veronese
- Geriatrics Section, Department of Medicine, University of Palermo, Palermo, Italy
| | - Andrea Rossanese
- Department of Infectious-Tropical Diseases and Microbiology, IRCCS “Sacro Cuore-Don Calabria,” Verona, Italy
| |
Collapse
|
18
|
Alvim RGF, Lima TM, Silva JL, de Oliveira GAP, Castilho LR. Process intensification for the production of yellow fever virus-like particles as potential recombinant vaccine antigen. Biotechnol Bioeng 2021; 118:3581-3592. [PMID: 34143442 DOI: 10.1002/bit.27864] [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: 10/06/2020] [Revised: 04/08/2021] [Accepted: 06/14/2021] [Indexed: 11/08/2022]
Abstract
Yellow fever (YF) is a life-threatening viral disease endemic in parts of Africa and Latin America. Although there is a very efficacious vaccine since the 1930s, YF still causes 29,000-60,000 annual deaths. During recent YF outbreaks there were issues of vaccine shortage of the current egg-derived vaccine; rare but fatal vaccine adverse effects occurred; and cases were imported to Asia, where the circulating mosquito vector could potentially start local transmission. Here we investigated the production of YF virus-like particles (VLPs) using stably transfected HEK293 cells. Process intensification was achieved by combining sequential FACS (fluorescence-activated cell sorting) rounds to enrich the stable cell pool in terms of high producers and the use of perfusion processes. At shaken-tube scale, FACS enrichment of cells allowed doubling VLP production, and pseudoperfusion cultivation (with daily medium exchange) further increased VLP production by 9.3-fold as compared to batch operation mode. At perfusion bioreactor scale, the use of an inclined settler as cell retention device showed operational advantages over an ATF system. A one-step steric exclusion chromatography purification allowed significant removal of impurities and is a promising technique for future integration of upstream and downstream operations. Characterization by different techniques confirmed the identity and 3D-structure of the purified VLPs.
Collapse
Affiliation(s)
- Renata G F Alvim
- COPPE, PEQ, Cell Culture Engineering Laboratory (LECC), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Túlio M Lima
- COPPE, PEQ, Cell Culture Engineering Laboratory (LECC), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.,School of Chemistry (EQ), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Jerson L Silva
- Institute of Medical Biochemistry Leopoldo de Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Guilherme A P de Oliveira
- Institute of Medical Biochemistry Leopoldo de Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Leda R Castilho
- COPPE, PEQ, Cell Culture Engineering Laboratory (LECC), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
19
|
Lara AN, Miyaji KT, Ibrahim KY, Lopes MH, Sartori AMC. Adverse events following yellow fever vaccination in immunocompromised persons. Rev Inst Med Trop Sao Paulo 2021; 63:e13. [PMID: 33656136 PMCID: PMC7924977 DOI: 10.1590/s1678-9946202163013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/15/2020] [Indexed: 12/15/2022] Open
Abstract
This observational retrospective study conducted during an yellow fever (YF)
outbreak in Sao Paulo, Brazil, in 2017-2018, describes adverse events (AE)
following YF vaccination of immunocompromised persons. Risks and benefits of
vaccination were individually evaluated by physicians. AE were assessed by phone
call or electronic mail, 14 to 90 days after vaccination. Three hundred and
eighty one immunocompromised persons received a full-dose of YF vaccine. Their
age ranged from 1.4 to 89.3 years (median 50.8 years); 53% were women; 178
(46.7%) had chronic kidney disease, 78 (20.5%) had immune-mediated inflammatory
diseases; 94 (24.7%) were using or had recently used immunosuppressive/
immunomodulatory drugs. All of them denied previous YF vaccination. We were able
to contact 341 (89.5%) vaccinees: 233 (68.3%) of them received the YF vaccine
from BioManguinhos and 108 (31.7%) received the vaccine from Sanofi-Pasteur; 130
(38.1%) vaccinees received other vaccines (up to 4) simultaneously with the the
YF vaccine, mostly hepatitis B (59 vaccinees), pneumococcal polysaccharide
23-valent (46), influenza (43) and diphtheria-tetanus (dT, 41). One hundred and
eleven vaccinees (32.6%) reported at least one AE: 79 (23.2%) presented systemic
AE, 44 (12.9%) had local AE and 12 had both, local and systemic AE. The most
common AE was pain at the injection site (41 persons, 12%), myalgia (34; 10%),
fever (25; 7.3%) and headache (16; 4.7%). There was no statistically significant
difference on the AE frequency according to the vaccine producer. There were
four severe AE: one hospitalization and three deaths, considered not related to
the YF vaccine.
Collapse
Affiliation(s)
- Amanda Nazareth Lara
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Centro de Referência para Imunobiológicos Especiais, São Paulo, São Paulo, Brazil
| | - Karina Takesaki Miyaji
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Centro de Referência para Imunobiológicos Especiais, São Paulo, São Paulo, Brazil
| | - Karim Yaqub Ibrahim
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Centro de Referência para Imunobiológicos Especiais, São Paulo, São Paulo, Brazil
| | - Marta Heloisa Lopes
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Centro de Referência para Imunobiológicos Especiais, São Paulo, São Paulo, Brazil
| | - Ana Marli Christovam Sartori
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas, Centro de Referência para Imunobiológicos Especiais, São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Bastard P, Michailidis E, Hoffmann HH, Chbihi M, Le Voyer T, Rosain J, Philippot Q, Seeleuthner Y, Gervais A, Materna M, de Oliveira PMN, Maia MDLS, Dinis Ano Bom AP, Azamor T, Araújo da Conceição D, Goudouris E, Homma A, Slesak G, Schäfer J, Pulendran B, Miller JD, Huits R, Yang R, Rosen LB, Bizien L, Lorenzo L, Chrabieh M, Erazo LV, Rozenberg F, Jeljeli MM, Béziat V, Holland SM, Cobat A, Notarangelo LD, Su HC, Ahmed R, Puel A, Zhang SY, Abel L, Seligman SJ, Zhang Q, MacDonald MR, Jouanguy E, Rice CM, Casanova JL. Auto-antibodies to type I IFNs can underlie adverse reactions to yellow fever live attenuated vaccine. J Exp Med 2021; 218:211761. [PMID: 33544838 PMCID: PMC7871457 DOI: 10.1084/jem.20202486] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/21/2020] [Accepted: 01/19/2021] [Indexed: 12/21/2022] Open
Abstract
Yellow fever virus (YFV) live attenuated vaccine can, in rare cases, cause life-threatening disease, typically in patients with no previous history of severe viral illness. Autosomal recessive (AR) complete IFNAR1 deficiency was reported in one 12-yr-old patient. Here, we studied seven other previously healthy patients aged 13 to 80 yr with unexplained life-threatening YFV vaccine–associated disease. One 13-yr-old patient had AR complete IFNAR2 deficiency. Three other patients vaccinated at the ages of 47, 57, and 64 yr had high titers of circulating auto-Abs against at least 14 of the 17 individual type I IFNs. These antibodies were recently shown to underlie at least 10% of cases of life-threatening COVID-19 pneumonia. The auto-Abs were neutralizing in vitro, blocking the protective effect of IFN-α2 against YFV vaccine strains. AR IFNAR1 or IFNAR2 deficiency and neutralizing auto-Abs against type I IFNs thus accounted for more than half the cases of life-threatening YFV vaccine-associated disease studied here. Previously healthy subjects could be tested for both predispositions before anti-YFV vaccination.
Collapse
Affiliation(s)
- Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | | | | | - Marwa Chbihi
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France
| | - Yoann Seeleuthner
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France
| | - Adrian Gervais
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France
| | - Marie Materna
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France
| | | | | | - Ana Paula Dinis Ano Bom
- Laboratory of Immunological Techniques, Bio-Manguinhos, Fiocruz, Ministry of Health, Rio de Janeiro, Brazil
| | - Tamiris Azamor
- Laboratory of Immunological Techniques, Bio-Manguinhos, Fiocruz, Ministry of Health, Rio de Janeiro, Brazil
| | | | | | - Akira Homma
- Bio-Manguinhos, Fiocruz, Ministry of Health, Rio de Janeiro, Brazil
| | - Günther Slesak
- Tropical Medicine Department, Tropenklinik Paul-Lechler-Krankenhaus, Tübingen, Germany
| | - Johannes Schäfer
- Tropical Medicine Department, Tropenklinik Paul-Lechler-Krankenhaus, Tübingen, Germany
| | - Bali Pulendran
- Emory Vaccine Center and the Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA.,Institute for Immunity, Transplantation and Infection, Department of Pathology, Department of Microbiology and Immunology, Stanford University, Stanford, CA
| | - Joseph D Miller
- Emory Vaccine Center and the Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA.,Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases, Division of Scientific Resources, Atlanta, GA
| | - Ralph Huits
- Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Rui Yang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Lindsey B Rosen
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Lucy Bizien
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France
| | - Lazaro Lorenzo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France
| | - Maya Chrabieh
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France
| | - Lucia V Erazo
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France
| | - Flore Rozenberg
- Laboratory of Virology, University of Paris, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Mohamed Maxime Jeljeli
- Laboratory of Immunology, University of Paris, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Rafi Ahmed
- Emory Vaccine Center and the Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Shen-Ying Zhang
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Laurent Abel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Stephen J Seligman
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY.,New York Medical College, Valhalla, NY
| | - Qian Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Necker Hospital for Sick Children, Paris, France.,University of Paris, Imagine Institute, Paris, France.,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY.,Howard Hughes Medical Institute, New York, NY
| |
Collapse
|
21
|
Pollard AJ, Bijker EM. A guide to vaccinology: from basic principles to new developments. Nat Rev Immunol 2020; 21:83-100. [PMID: 33353987 PMCID: PMC7754704 DOI: 10.1038/s41577-020-00479-7] [Citation(s) in RCA: 796] [Impact Index Per Article: 159.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2020] [Indexed: 12/17/2022]
Abstract
Immunization is a cornerstone of public health policy and is demonstrably highly cost-effective when used to protect child health. Although it could be argued that immunology has not thus far contributed much to vaccine development, in that most of the vaccines we use today were developed and tested empirically, it is clear that there are major challenges ahead to develop new vaccines for difficult-to-target pathogens, for which we urgently need a better understanding of protective immunity. Moreover, recognition of the huge potential and challenges for vaccines to control disease outbreaks and protect the older population, together with the availability of an array of new technologies, make it the perfect time for immunologists to be involved in designing the next generation of powerful immunogens. This Review provides an introductory overview of vaccines, immunization and related issues and thereby aims to inform a broad scientific audience about the underlying immunological concepts. This Review, aimed at a broad scientific audience, provides an introductory guide to the history, development and immunological basis of vaccines, immunization and related issues to provide insight into the challenges facing immunologists who are designing the next generation of vaccines.
Collapse
Affiliation(s)
- Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK. .,NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK.
| | - Else M Bijker
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
| |
Collapse
|
22
|
Schreiner D, Zepp F. [General principles of vaccination under immunosuppression]. Z Rheumatol 2020; 79:848-854. [PMID: 33034682 DOI: 10.1007/s00393-020-00891-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2020] [Indexed: 11/25/2022]
Abstract
The number of patients suffering from immunodeficiency is increasing; however, the vaccination rate of these patients is below average. Administration of inactivated vaccines is harmless but does not reliably trigger a persistent immune response. Live vaccines provide a reliable protection but can cause severe disease in immunocompromised patients. Live vaccines can be administered under defined levels of immunosuppression, e.g. against measles, mumps, rubella and varicella (MMRV). In addition, the immunization of the domestic environment plays an important role in preventing infectious diseases.
Collapse
Affiliation(s)
- D Schreiner
- Zentrum für Kinder- und Jugendmedizin, Universitätsmedizin Mainz, Langenbeckstr. 1, 55131, Mainz, Deutschland
| | - F Zepp
- Zentrum für Kinder- und Jugendmedizin, Universitätsmedizin Mainz, Langenbeckstr. 1, 55131, Mainz, Deutschland.
| |
Collapse
|
23
|
Valim V, Machado KLLL, Miyamoto ST, Pinto AD, Rocha PCM, Serrano EV, Dinis VG, Gouvêa SA, Dias JGF, Campi-Azevedo AC, Teixeira-Carvalho A, Peruhype-Magalhães V, da Costa-Rocha IA, de Lima SMB, Miranda EH, Trindade GF, Maia MDLDS, Gavi MBRDO, da Silva LB, Duque RH, Gianordoli APE, Casagrande TZ, Oliveira KG, Moura BCDM, Nicole-Batista F, Rodrigues LC, Clemente TB, Magalhães ES, Bissoli MDF, Gouvea MDPG, Pinto-Neto LFDS, Costa CZ, Giovelli RA, Brandão LR, Polito ETL, Koehlert IDO, Borjaille BP, Pereira DB, Dias LH, Merlo DL, Genelhu LFF, Pretti FZ, Giacomin MDS, Burian APN, Fantinato FFST, Pileggi GS, da Mota LMH, Martins-Filho OA. Planned Yellow Fever Primary Vaccination Is Safe and Immunogenic in Patients With Autoimmune Diseases: A Prospective Non-interventional Study. Front Immunol 2020; 11:1382. [PMID: 32765496 PMCID: PMC7379374 DOI: 10.3389/fimmu.2020.01382] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 05/29/2020] [Indexed: 12/16/2022] Open
Abstract
Yellow Fever (YF) vaccination is suggested to induce a large number of adverse events (AE) and suboptimal responses in patients with autoimmune diseases (AID); however, there have been no studies on 17DD-YF primary vaccination performance in patients with AID. This prospective non-interventional study conducted between March and July, 2017 assessed the safety and immunogenicity of planned 17DD-YF primary vaccination in patients with AID. Adult patients with AID (both sexes) were enrolled, along with healthy controls, at a single hospital (Vitória, Brazil). Included patients were referred for planned vaccination by a rheumatologist; in remission, or with low disease activity; and had low level immunosuppression or the attending physician advised interruption of immunosuppression for safety reasons. The occurrence of AE, neutralizing antibody kinetics, seropositivity rates, and 17DD-YF viremia were evaluated at various time points (day 0 (D0), D3, D4, D5, D6, D14, and D28). Individuals evaluated (n = 278), including patients with rheumatoid arthritis (RA; 79), spondyloarthritis (SpA; 59), systemic sclerosis (8), systemic lupus erythematosus (SLE; 27), primary Sjögren's syndrome (SS; 54), and healthy controls (HC; 51). Only mild AE were reported. The frequency of local and systemic AE in patients with AID and HC did not differ significantly (8 vs. 10% and 21 vs. 32%; p = 1.00 and 0.18, respectively). Patients with AID presented late seroconversion profiles according to kinetic timelines of the plaque reduction neutralization test (PRNT). PRNT-determined virus titers (copies/mL) [181 (95% confidence interval (CI), 144–228) vs. 440 (95% CI, 291–665), p = 0.004] and seropositivity rate (78 vs. 96%, p = 0.01) were lower in patients with AID after 28 days, particularly those with SpA (73%) and SLE (73%), relative to HC. The YF viremia peak (RNAnemia) was 5–6 days after vaccination in all groups. In conclusion, consistent seroconversion rates were observed in patients with AID and our findings support that planned 17DD-YF primary vaccination is safe and immunogenic in patients with AID.
Collapse
Affiliation(s)
- Valéria Valim
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Ketty Lysie Libardi Lira Machado
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Samira Tatiyama Miyamoto
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Arthur Dalmaso Pinto
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Priscila Costa Martins Rocha
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Erica Vieira Serrano
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Valquiria Garcia Dinis
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil.,Escola de Ciências da Saúde da Santa Casa de Misericórdia, Vitória, Brazil
| | - Sônia Alves Gouvêa
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - João Gabriel Fragoso Dias
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | | | | | | | | | - Sheila Maria Barbosa de Lima
- Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Emily Hime Miranda
- Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Gisela Freitas Trindade
- Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | | | - Lidia Balarini da Silva
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Ruben Horst Duque
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Ana Paula Espíndula Gianordoli
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Thays Zanon Casagrande
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Karine Gadioli Oliveira
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Bruna Costa da Mata Moura
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Fernanda Nicole-Batista
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Luiza Correa Rodrigues
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Thalles Brandão Clemente
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Enan Sales Magalhães
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Maria de Fatima Bissoli
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | - Maria da Penha Gomes Gouvea
- Divisão de Reumatologia do Hospital Universitário Cassiano Antônio de Moraes, Universidade Federal do Espírito Santo (UFES), Vitória, Brazil
| | | | | | | | | | | | | | | | | | - Laiza Hombre Dias
- Sociedade de Reumatologia do Espírito Santo (SORES), Vitória, Brazil
| | | | | | - Flavia Zon Pretti
- Sociedade de Reumatologia do Espírito Santo (SORES), Vitória, Brazil
| | | | - Ana Paula Neves Burian
- Centro de Referências para Imunobiológicos Especiais (CRIE) da Secretaria de Saúde do Estado do Espírito Santo, Vitória, Brazil
| | | | | | - Lícia Maria Henrique da Mota
- Divisão de Reumatologia do Hospital Universitário de Brasília, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil
| | | |
Collapse
|
24
|
Huttner A, Eperon G, Lascano AM, Roth S, Schwob JM, Siegrist CA, Lalive PH. Risk of MS relapse after yellow fever vaccination: A self-controlled case series. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:7/4/e726. [PMID: 32358223 PMCID: PMC7217657 DOI: 10.1212/nxi.0000000000000726] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 03/20/2020] [Indexed: 01/25/2023]
Abstract
Objective To determine whether live-attenuated yellow fever vaccine (YFV) was associated with MS relapse, we evaluated the clinical courses of 23 patients in the year before and the year after immunization at the university hospital of Geneva, Switzerland. Methods This self-controlled retrospective cohort included adult patients with MS receiving YFV between 2014 and 2018 and defined the year before vaccination, the 3 months thereafter, and the 9 months following as the pre-exposure (PEP), exposure-risk (ERP), and postrisk (PRP) periods, respectively. The primary outcome was the relative incidence of relapse in the ERP vs the PEP. Secondary end points included the presence of new T2-weighted (T2) or T1-weighted gadolinium-positive (T1Gd+) MRI lesions. Results Of 23 patients with MS receiving YFV (20 relapsing MS and 3 primary progressive MS), 17 (74%) were women; mean age was 34 years (SD ±10); and 10 of 23 (40%) were treated with disease-modifying therapies (DMTs). Although 9 patients experienced 12 relapses in the PEP, only one experienced a relapse in the ERP; 3 other patients experienced one relapse each in the PRP. None of the 8 patients receiving natalizumab at the time of vaccination experienced relapse thereafter. In the PEP, ERP, and PRP, 18, 2, and 9 patients had new brain and/or spinal cord lesions on T2 or T1Gd + MRI, respectively. Conclusions In this cohort, YF vaccination was associated with neither an increase in MS relapse nor emergence of brain and/or spinal lesions. Further studies are warranted to confirm these findings. Classification of evidence This study provides Class IV evidence that for persons with MS, YFV may not increase relapse risk.
Collapse
Affiliation(s)
- Angela Huttner
- From the Center for Vaccinology (A.H., C.-A.S.), University of Geneva; Division of Infectious Diseases (A.H.), Geneva University Hospitals; Division of Tropical and Humanitarian Medicine (G.E., J.-M.S.), Geneva University Hospitals; Department of Neurosciences (A.M.L., S.R., P.H.L.), Division of Neurology, Unit of Neuroimmunology and Neuromuscular Diseases, Geneva University Hospitals; Department of Pathology and Immunology (C.-A.S., P.H.L.), Faculty of Medicine, University of Geneva; and Division of Laboratory Medicine (P.H.L.), Department of Diagnostic, Geneva University Hospitals, Switzerland
| | - Gilles Eperon
- From the Center for Vaccinology (A.H., C.-A.S.), University of Geneva; Division of Infectious Diseases (A.H.), Geneva University Hospitals; Division of Tropical and Humanitarian Medicine (G.E., J.-M.S.), Geneva University Hospitals; Department of Neurosciences (A.M.L., S.R., P.H.L.), Division of Neurology, Unit of Neuroimmunology and Neuromuscular Diseases, Geneva University Hospitals; Department of Pathology and Immunology (C.-A.S., P.H.L.), Faculty of Medicine, University of Geneva; and Division of Laboratory Medicine (P.H.L.), Department of Diagnostic, Geneva University Hospitals, Switzerland
| | - Agustina M Lascano
- From the Center for Vaccinology (A.H., C.-A.S.), University of Geneva; Division of Infectious Diseases (A.H.), Geneva University Hospitals; Division of Tropical and Humanitarian Medicine (G.E., J.-M.S.), Geneva University Hospitals; Department of Neurosciences (A.M.L., S.R., P.H.L.), Division of Neurology, Unit of Neuroimmunology and Neuromuscular Diseases, Geneva University Hospitals; Department of Pathology and Immunology (C.-A.S., P.H.L.), Faculty of Medicine, University of Geneva; and Division of Laboratory Medicine (P.H.L.), Department of Diagnostic, Geneva University Hospitals, Switzerland
| | - Serge Roth
- From the Center for Vaccinology (A.H., C.-A.S.), University of Geneva; Division of Infectious Diseases (A.H.), Geneva University Hospitals; Division of Tropical and Humanitarian Medicine (G.E., J.-M.S.), Geneva University Hospitals; Department of Neurosciences (A.M.L., S.R., P.H.L.), Division of Neurology, Unit of Neuroimmunology and Neuromuscular Diseases, Geneva University Hospitals; Department of Pathology and Immunology (C.-A.S., P.H.L.), Faculty of Medicine, University of Geneva; and Division of Laboratory Medicine (P.H.L.), Department of Diagnostic, Geneva University Hospitals, Switzerland
| | - Jean-Marc Schwob
- From the Center for Vaccinology (A.H., C.-A.S.), University of Geneva; Division of Infectious Diseases (A.H.), Geneva University Hospitals; Division of Tropical and Humanitarian Medicine (G.E., J.-M.S.), Geneva University Hospitals; Department of Neurosciences (A.M.L., S.R., P.H.L.), Division of Neurology, Unit of Neuroimmunology and Neuromuscular Diseases, Geneva University Hospitals; Department of Pathology and Immunology (C.-A.S., P.H.L.), Faculty of Medicine, University of Geneva; and Division of Laboratory Medicine (P.H.L.), Department of Diagnostic, Geneva University Hospitals, Switzerland
| | - Claire-Anne Siegrist
- From the Center for Vaccinology (A.H., C.-A.S.), University of Geneva; Division of Infectious Diseases (A.H.), Geneva University Hospitals; Division of Tropical and Humanitarian Medicine (G.E., J.-M.S.), Geneva University Hospitals; Department of Neurosciences (A.M.L., S.R., P.H.L.), Division of Neurology, Unit of Neuroimmunology and Neuromuscular Diseases, Geneva University Hospitals; Department of Pathology and Immunology (C.-A.S., P.H.L.), Faculty of Medicine, University of Geneva; and Division of Laboratory Medicine (P.H.L.), Department of Diagnostic, Geneva University Hospitals, Switzerland
| | - Patrice H Lalive
- From the Center for Vaccinology (A.H., C.-A.S.), University of Geneva; Division of Infectious Diseases (A.H.), Geneva University Hospitals; Division of Tropical and Humanitarian Medicine (G.E., J.-M.S.), Geneva University Hospitals; Department of Neurosciences (A.M.L., S.R., P.H.L.), Division of Neurology, Unit of Neuroimmunology and Neuromuscular Diseases, Geneva University Hospitals; Department of Pathology and Immunology (C.-A.S., P.H.L.), Faculty of Medicine, University of Geneva; and Division of Laboratory Medicine (P.H.L.), Department of Diagnostic, Geneva University Hospitals, Switzerland.
| |
Collapse
|
25
|
Kum DB, Mishra N, Vrancken B, Thibaut HJ, Wilder-Smith A, Lemey P, Neyts J, Dallmeier K. Limited evolution of the yellow fever virus 17d in a mouse infection model. Emerg Microbes Infect 2020; 8:1734-1746. [PMID: 31797751 PMCID: PMC6896426 DOI: 10.1080/22221751.2019.1694394] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
By infecting mice with the yellow fever virus vaccine strain 17D (YFV-17D; Stamaril®), the dose dependence and evolutionary consequences of neurotropic yellow fever infection was assessed. Highly susceptible AG129 mice were used to allow for a maximal/unlimited expansion of the viral populations. Infected mice uniformly developed neurotropic disease; the virus was isolated from their brains, plaque purified and sequenced. Viral RNA populations were overall rather homogenous [Shannon entropies 0−0.15]. The remaining, yet limited intra-host population diversity (0−11 nucleotide exchanges per genome) appeared to be a consequence of pre-existing clonal heterogeneities (quasispecies) of Stamaril®. In parallel, mice were infected with a molecular clone of YFV-17D which was in vivo launched from a plasmid. Such plasmid-launched YFV-17D had a further reduced and almost clonal evolution. The limited intra-host evolution during unrestricted expansion in a highly susceptible host is relevant for vaccine and drug development against flaviviruses in general. Firstly, a propensity for limited evolution even upon infection with a (very) low inoculum suggests that fractional dosing as implemented in current YF-outbreak control may pose only a limited risk of reversion to pathogenic vaccine-derived virus variants. Secondly, it also largely lowers the chance of antigenic drift and development of resistance to antivirals.
Collapse
Affiliation(s)
- Dieudonné Buh Kum
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium.,Aligos Belgium, Leuven, Belgium
| | - Niraj Mishra
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Bram Vrancken
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory for Clinical and Epidemiological Virology, Leuven, Belgium
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Annelies Wilder-Smith
- Department of Disease Control, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Heidelberg Institute of Global Health, University of Heidelberg, Heidelberg, Germany
| | - Philippe Lemey
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory for Clinical and Epidemiological Virology, Leuven, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| |
Collapse
|
26
|
Dudley MZ, Halsey NA, Omer SB, Orenstein WA, O'Leary ST, Limaye RJ, Salmon DA. The state of vaccine safety science: systematic reviews of the evidence. THE LANCET. INFECTIOUS DISEASES 2020; 20:e80-e89. [PMID: 32278359 DOI: 10.1016/s1473-3099(20)30130-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 11/24/2022]
Abstract
This Review updates the scientific evidence assessing possible causal associations of adverse events following immunisation (AEFI) compiled in the 2012 report from the Institute of Medicine and the 2014 report from the Agency for Healthcare Research and Quality. For 12 of 46 AEFI examined, a causal relationship has been established with at least one vaccine currently routinely recommended to the general USA population: anaphylaxis, arthralgia or arthritis (mild, acute, and transient, not chronic), deltoid bursitis (when vaccine is administered improperly), disseminated varicella infection (in immune deficient individuals for whom the varicella vaccine is contraindicated), encephalitis, febrile seizures, Guillain-Barré syndrome, hepatitis (in immune deficient individuals for whom the varicella vaccine is contraindicated), herpes zoster, immune thrombocytopenic purpura, meningitis, and syncope. Other than mild acute and transient arthralgia or arthritis, which is very common in adult women after rubella vaccine, these adverse reactions are rare or very rare. Vaccines have an excellent safety profile overall and provide protection against infectious diseases to individuals and the general population.
Collapse
Affiliation(s)
- Matthew Z Dudley
- Department of International Health, Johns Hopkins University, Baltimore, MD, USA; Institute of Vaccine Safety, Johns Hopkins University, Baltimore, MD, USA
| | - Neal A Halsey
- Department of International Health, Johns Hopkins University, Baltimore, MD, USA; Institute of Vaccine Safety, Johns Hopkins University, Baltimore, MD, USA; Department of Pediatrics, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Saad B Omer
- Epidemiology of Microbial Diseases, Yale School of Public Health, Yale School for Global Health, and Yale School of Medicine, New Haven, CT, USA
| | - Walter A Orenstein
- Department of Pediatrics and Emory Vaccine Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - Sean T O'Leary
- Adult and Child Consortium for Health Outcomes Research and Delivery Science and Department of Pediatrics, Anschutz Medical Campus and Children's Hospital, University of Colorado, Aurora, CO, USA
| | - Rupali J Limaye
- Department of International Health, Johns Hopkins University, Baltimore, MD, USA; Institute of Vaccine Safety, Johns Hopkins University, Baltimore, MD, USA; Department of Health, Behavior & Society, Johns Hopkins University, Baltimore, MD, USA; Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA; International Vaccine Access Center, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Daniel A Salmon
- Department of International Health, Johns Hopkins University, Baltimore, MD, USA; Institute of Vaccine Safety, Johns Hopkins University, Baltimore, MD, USA; Department of Health, Behavior & Society, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
27
|
Burkhard J, Ciurea A, Gabay C, Hasler P, Müller R, Niedrig M, Fehr J, Villiger P, Visser LG, de Visser AW, Walker UA, Hatz C, Bühler S. Long-term immunogenicity after yellow fever vaccination in immunosuppressed and healthy individuals. Vaccine 2020; 38:3610-3617. [PMID: 31911033 DOI: 10.1016/j.vaccine.2019.12.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/11/2019] [Accepted: 12/19/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND The live-attenuated yellow fever vaccine (YFV) is generally contraindicated in immunosuppressed patients. Our aim was to investigate if immunosuppressive therapy impairs the long-term protection against yellow fever virus in patients who had received YFV prior to the start of their immunosuppressive therapy. METHODS Our study examined 35 healthy individuals and 40 immunosuppressed patients with autoimmune diseases or organ transplants. All individuals had received YFV prior to the onset of their immunosuppression. We analysed the long-term influence of the immunosuppressive therapy on the YFV protective immunity by measuring neutralising antibodies (NA) with the Plaque Reduction Neutralisation Test (PRNT). We assessed risk factors for a negative PRNT result (titre below 1: 10) and their influence on the magnitude of the NA. RESULTS A median time interval of 21.1 years (interquartile range 14.4-31.3 years) after the YFV in all patients, a total of 35 immunosuppressed patients (88%) were seropositive (PRNT ≥ 1:10) compared to 31 patients (89%) in the control group. The geometric mean titres of NA did not differ between the groups. The duration of an underlying rheumatic disease was the only risk factor found for a lower magnitude of NA. An insufficient level of NA was found in nine subjects (12%) who had received a single dose of YFV (in one subject, the number of YFV doses was unknown). CONCLUSION The use of an immunosuppressive drug started after the administration of the YFV did not affect long-term persistence of NA. A second dose of YFV may be necessary to secure long-term immunity.
Collapse
Affiliation(s)
- J Burkhard
- Department of Public Health / Division of Infectious Diseases, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - A Ciurea
- Department of Rheumatology, University Hospital of Zurich, Zurich, Switzerland
| | - C Gabay
- Division of Rheumatology, University Hospital of Geneva, Geneva, Switzerland
| | - P Hasler
- Department of Rheumatology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - R Müller
- Division of Rheumatology, Department of Internal Medicine, Cantonal Hospital St. Gallen, St. Gallen, Switzerland; Division of Rheumatology and Clinical Immunology, Department of Internal Medicine Ludwig-Maximilians-University Munich, Germany
| | - M Niedrig
- Robert Koch-Institut (RKI), Berlin, Germany
| | - J Fehr
- Department of Public Health / Division of Infectious Diseases, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - P Villiger
- Department of Rheumatology and Clinical Immunology/Allergology, University Hospital of Bern, Bern, Switzerland
| | - L G Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - A W de Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - U A Walker
- Department of Rheumatology, University Hospital Basel, Basel, Switzerland
| | - C Hatz
- Department of Public Health / Division of Infectious Diseases, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland; Department of Medicine and Diagnostics, Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Switzerland; Division of Infectious Diseases & Hospital Epidemiology, Kantonsspital St. Gallen, Switzerland
| | - S Bühler
- Department of Public Health / Division of Infectious Diseases, Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland; Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I. Department of Medicine University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
28
|
Disease Resurgence, Production Capability Issues and Safety Concerns in the Context of an Aging Population: Is There a Need for a New Yellow Fever Vaccine? Vaccines (Basel) 2019; 7:vaccines7040179. [PMID: 31717289 PMCID: PMC6963298 DOI: 10.3390/vaccines7040179] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/28/2019] [Accepted: 11/05/2019] [Indexed: 12/19/2022] Open
Abstract
Yellow fever is a potentially fatal, mosquito-borne viral disease that appears to be experiencing a resurgence in endemic areas in Africa and South America and spreading to non-endemic areas despite an effective vaccine. This trend has increased the level of concern about the disease and the potential for importation to areas in Asia with ecological conditions that can sustain yellow fever virus transmission. In this article, we provide a broad overview of yellow fever burden of disease, natural history, treatment, vaccine, prevention and control initiatives, and vaccine and therapeutic agent development efforts.
Collapse
|
29
|
|
30
|
Vaccination Recommendations for Adults With Autoimmune Inflammatory Rheumatic Diseases in Latin America. J Clin Rheumatol 2019; 24:138-147. [PMID: 29232324 DOI: 10.1097/rhu.0000000000000624] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND/OBJECTIVE Patients with autoimmune inflammatory rheumatic diseases (AIRDs) are at increased risk of contracting severe infections and suffering complications, particularly when they are receiving immunomodulating therapy. Vaccination is an important means to prevent many potential infections and thereby reduce the morbidity and mortality associated with AIRD. The purpose of this consensus document is to provide health care professionals with recommendations for the vaccination of AIRD patients who reside in Latin America. The recommendations were developed by an expert committee from the region based on a review of the literature and their clinical experience. METHODS The Americas Health Foundation (AHF) used PubMed and EMBASE to identify clinicians and scientists with an academic or hospital affiliation and who had published in the field of adult vaccination and rheumatic diseases since 2010. As a result of this effort, AHF convened an 8-member panel of clinical and scientific experts from Latin America. Both the AHF and panel members conducted a careful literature review to identify relevant publications in the areas of adult vaccination and rheumatology, and the sum of the articles identified was provided to the entire panel. Prior to the conference, panelists were each asked to prepare a written response to a salient issue on the subject, identified by AHF. RESULTS AND CONCLUSIONS During the conference, each response was edited by the entire group, through numerous drafts and rounds of discussion until a complete consensus on vaccination recommendations for adult patients with AIRDs was obtained, including 7 key recommendations.
Collapse
|
31
|
|
32
|
Cohen C, Moreira ED, Nañez H, Nachiappan JP, Arvinder-Singh HS, Huoi C, Nealon J, Sarti E, Puentes-Rosas E, Moureau A, Khromava A. Incidence rates of neurotropic-like and viscerotropic-like disease in three dengue-endemic countries: Mexico, Brazil, and Malaysia. Vaccine 2019; 37:1868-1875. [PMID: 30826144 DOI: 10.1016/j.vaccine.2019.01.087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 11/25/2022]
Abstract
BACKGROUND The background incidence of viscerotropic- (VLD) and neurotropic-like disease (NLD) unrelated to immunization in dengue-endemic countries is currently unknown. METHODS This retrospective population-based analysis estimated crude and standardized incidences of VLD and NLD in twelve hospitals in Brazil (n = 3), Mexico (n = 3), and Malaysia (n = 6) over a 1-year period before the introduction of the tetravalent dengue vaccine. Catchment areas were estimated using publicly available population census information and administrative data. The denominator population for incidence rates was calculated, and sensitivity analyses assessed the impact of important assumptions. RESULTS Total cases adjudicated as definite VLD were 5, 57, and 56 in Brazil, Mexico, and Malaysia, respectively. Total cases adjudicated as definite NLD were 103, 29, and 26 in Brazil, Mexico, and Malaysia, respectively. Crude incidence rates of cases adjudicated as definite VLD in Brazil, Mexico, and Malaysia were 1.17, 2.60, and 1.48 per 100,000 person-years, respectively. Crude incidence rates of cases adjudicated as definite NLD in Brazil, Mexico, and Malaysia were 4.45, 1.32, and 0.69 per 100,000 person-years, respectively. CONCLUSIONS Background incidence estimates of VLD and NLD obtained in Mexico, Brazil, and Malaysia could provide context for cases occurring after the introduction of the tetravalent dengue vaccine.
Collapse
Affiliation(s)
| | - Edson D Moreira
- Associação Obras Sociais Irmã Dulce and Oswaldo Cruz Foundation, Brazilian Ministry of Health, Bahia, Brazil.
| | - Homero Nañez
- University Hospital Dr. José E. González, Faculty of Medicine Universidad Autonoma de Nuevo León, Monterrey N.L., Mexico.
| | | | | | | | | | - Elsa Sarti
- Sanofi Pasteur LATAM, Coyoacán, CDMX, Mexico.
| | | | - Annick Moureau
- Clinical Development, Sanofi Pasteur, Marcy l'Etoile, Lyon, France.
| | | | | |
Collapse
|
33
|
Kum DB, Mishra N, Boudewijns R, Gladwyn-Ng I, Alfano C, Ma J, Schmid MA, Marques RE, Schols D, Kaptein S, Nguyen L, Neyts J, Dallmeier K. A yellow fever-Zika chimeric virus vaccine candidate protects against Zika infection and congenital malformations in mice. NPJ Vaccines 2018; 3:56. [PMID: 30564463 PMCID: PMC6292895 DOI: 10.1038/s41541-018-0092-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 11/05/2018] [Indexed: 12/27/2022] Open
Abstract
The recent Zika virus (ZIKV) epidemic in the Americas led to an intense search for therapeutics and vaccines. Here we report the engineering of a chimeric virus vaccine candidate (YF-ZIKprM/E) by replacing the antigenic surface glycoproteins and the capsid anchor of YFV-17D with those of a prototypic Asian lineage ZIKV isolate. By intracellular passaging, a variant with adaptive mutations in the E protein was obtained. Unlike YFV-17D, YF-ZIKprM/E replicates poorly in mosquito cells. Also, YF-ZIKprM/E does not cause disease nor mortality in interferon α/β, and γ receptor KO AG129 mice nor following intracranial inoculation of BALB/c pups. A single dose as low as 1 × 102 PFU results, as early as 7 days post vaccination, in seroconversion to neutralizing antibodies and confers full protection in AG129 mice against stringent challenge with a lethal inoculum (105 LD50) of either homologous or heterologous ZIKV strains. Induction of multi-functional CD4+ and CD8+ T cell responses against ZIKV structural and YFV-17D non-structural proteins indicates that cellular immunity may also contribute to protection. Vaccine immunogenicity and protection was confirmed in other mouse strains, including after temporal blockade of interferon-receptors in wild-type mice to facilitate ZIKV replication. Vaccination of wild-type NMRI dams with YF-ZIKprM/E results in complete protection of foetuses against brain infections and malformations following a stringent intraplacental challenge with an epidemic ZIKV strain. The particular characteristic of YF-ZIKprM/E in terms of efficacy and its marked attenuation in mice warrants further exploration as a vaccine candidate. Zika virus (ZIKV) infection generally results in mild symptoms but can cause serious developmental abnormalities in infants born to ZIKV infected mothers. Kai Dallmeier and colleagues at the KU Leuven in Belgium, engineered a chimeric live-attenuated vaccine (YF-ZIKprM/E) by swapping the glycoprotein from the Yellow Fever vaccine YFV-17D with that of a pre-epidemic ZIKV strain. YF-ZIKprM/E is very well tolerated with no adverse effects even following high dose intracranial infection. Mice highly susceptible to ZIKV infection—including AG129 and type I interferon receptor deficient strains—vaccinated with a single dose of YF-ZIKprM/E are fully protected from lethal ZIKV challenge. Protection can be achieved within 7 days and by low doses of YF-ZIKprM/E, is durable and generally results in sterilizing immunity. YF-ZIKprM/E elicits both neutralizing antibodies and robust cellular immunity. Finally, YF-ZIKprM/E can also prevent vertical transmission of ZIKV and achieve efficient protection of pups from neurological defects following intraplacental challenge.
Collapse
Affiliation(s)
- Dieudonné B Kum
- KU Leuven Department of Microbiology and Immunology, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Niraj Mishra
- KU Leuven Department of Microbiology and Immunology, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Robbert Boudewijns
- KU Leuven Department of Microbiology and Immunology, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Ivan Gladwyn-Ng
- 2GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, C.H.U. Sart Tilman, Liège, Belgium
| | - Christian Alfano
- 2GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, C.H.U. Sart Tilman, Liège, Belgium
| | - Ji Ma
- KU Leuven Department of Microbiology and Immunology, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Michael A Schmid
- KU Leuven Department of Microbiology and Immunology, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Rafael E Marques
- 3Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Sao Paulo, Brazil
| | - Dominique Schols
- KU Leuven Department of Microbiology and Immunology, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Suzanne Kaptein
- KU Leuven Department of Microbiology and Immunology, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Laurent Nguyen
- 2GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, C.H.U. Sart Tilman, Liège, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology and Immunology, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Kai Dallmeier
- KU Leuven Department of Microbiology and Immunology, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| |
Collapse
|
34
|
Robinson HL, Marzi A. Ebola virus - prospects for a novel virus-like-particle-expressing modified vaccinia Ankara-based vaccine. Expert Rev Vaccines 2018; 17:769-771. [PMID: 30092691 PMCID: PMC9491071 DOI: 10.1080/14760584.2018.1510775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
| | - Andrea Marzi
- b Laboratory of Virology, Division of Intramural Research , National Institute of Allergy and Infectious Diseases, National Institutes of Health , Hamilton , MT , USA
| |
Collapse
|
35
|
Abstract
Biological sex is a determinant of both susceptibility to and pathogenesis of multiple infections, including HIV. These differences have effects on the spectrum of HIV disease from acquisition to eradication, with diverse mechanisms including distinct chromosomal complements, variation in microbiota composition, hormonal effects on transcriptional profiles, and expression of different immunoregulatory elements. With a comparative biology approach, these sex differences can be used to highlight protective and detrimental immune activation pathways, to identify strategies for effective prevention, treatment, and curative interventions.
Collapse
Affiliation(s)
- Eileen P Scully
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine
| |
Collapse
|
36
|
The value of Autoimmune Syndrome Induced by Adjuvant (ASIA) - Shedding light on orphan diseases in autoimmunity. Autoimmun Rev 2018. [DOI: 10.1016/j.autrev.2017.11.037] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
37
|
Abstract
PURPOSE OF REVIEW This review will outline the multilevel effects of biological sex on HIV acquisition, pathogenesis, treatment response, and prospects for cure. Potential mechanisms will be discussed along with future research directions. RECENT FINDINGS HIV acquisition risk is modified by sex hormones and the vaginal microbiome, with the latter acting through both inflammation and local metabolism of pre-exposure prophylaxis drugs. Female sex associates with enhanced risk for non-AIDS morbidities including cardiovascular and cerebrovascular disease, suggesting different inflammatory profiles in men and women. Data from research on HIV cure points to sex differences in viral reservoir dynamics and a direct role for sex hormones in latency maintenance. Biological sex remains an important variable in determining the risk of HIV infection and subsequent viral pathogenesis, and emerging data suggest sex differences relevant to curative interventions. Recruitment of women in HIV clinical research is a pathway to both optimize care for women and to identify novel therapeutics for use in both men and women.
Collapse
Affiliation(s)
- Eileen P Scully
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Pre-Clinical Teaching Building, Suite 211, 725 N Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
38
|
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: 2.8] [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
|
39
|
Abstract
Recognition that the live yellow fever vaccine may rarely be associated with viscerotropic disease (YEL-AVD) has diminished its safety status. However, the vaccine remains the principal tool for limiting the occurrence of yellow fever, making large portions of Africa and South America more habitable. The subject has previously been exhaustively reviewed. Novel concepts in the current report include the description of a systematic method for deciding whom to vaccinate, recommendations for obtaining data helpful in making that decision, and suggestions for additional study. The vaccine is indeed a worthy friend, but its adverse reactions need to be recognized.
Collapse
Affiliation(s)
- Stephen J Seligman
- a Department of Microbiology and Immunology , New York Medical College , Valhalla , NY , USA.,b St. Giles Laboratory of Human Genetics of Infectious Diseases , The Rockefeller University , New York , NY , USA
| | - Jean-Laurent Casanova
- b St. Giles Laboratory of Human Genetics of Infectious Diseases , The Rockefeller University , New York , NY , USA.,c Howard Hughes Medical Institute , The Rockefeller University , New York , NY , USA
| |
Collapse
|
40
|
Holbrook MR. Historical Perspectives on Flavivirus Research. Viruses 2017; 9:E97. [PMID: 28468299 PMCID: PMC5454410 DOI: 10.3390/v9050097] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/13/2017] [Accepted: 04/21/2017] [Indexed: 12/21/2022] Open
Abstract
The flaviviruses are small single-stranded RNA viruses that are typically transmitted by mosquito or tick vectors. These "arboviruses" are found around the world and account for a significant number of cases of human disease. The flaviviruses cause diseases ranging from mild or sub-clinical infections to lethal hemorrhagic fever or encephalitis. In many cases, survivors of neurologic flavivirus infections suffer long-term debilitating sequelae. Much like the emergence of West Nile virus in the United States in 1999, the recent emergence of Zika virus in the Americas has significantly increased the awareness of mosquito-borne viruses. The diseases caused by several flaviviruses have been recognized for decades, if not centuries. However, there is still a lot that is unknown about the flaviviruses as the recent experience with Zika virus has taught us. The objective of this review is to provide a general overview and some historical perspective on several flaviviruses that cause significant human disease. In addition, available medical countermeasures and significant gaps in our understanding of flavivirus biology are also discussed.
Collapse
Affiliation(s)
- Michael R Holbrook
- NIAID Integrated Research Facility, 8200 Research Plaza, Ft. Detrick, Frederick, MD 21702, USA.
| |
Collapse
|
41
|
Beirão P, Pereira P, Nunes A, Antunes P. Yellow fever vaccine-associated neurological disease, a suspicious case. BMJ Case Rep 2017; 2017:bcr-2016-218706. [PMID: 28254833 DOI: 10.1136/bcr-2016-218706] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
A 70-year-old man with known cardiovascular risk factors, presented with acute onset expression aphasia, agraphia, dyscalculia, right-left disorientation and finger agnosia, without fever or meningeal signs. Stroke was thought to be the cause, but cerebrovascular disease investigation was negative. Interviewing the family revealed he had undergone yellow fever vaccination 18 days before. Lumbar puncture revealed mild protein elevation. Cultural examinations, Coxiella burnetti, and neurotropic virus serologies were negative. Regarding the yellow fever virus, IgG was identified in serum and cerebrospinal fluid (CSF), with negative IgM and virus PCR in CSF. EEG showed an encephalopathic pattern. The patient improved gradually and a week after discharge was his usual self. Only criteria for suspect neurotropic disease were met, but it's possible the time spent between symptom onset and lumbar puncture prevented a definite diagnosis of yellow fever vaccine-associated neurological disease. This gap would have been smaller if the vaccination history had been collected earlier.
Collapse
Affiliation(s)
- Pedro Beirão
- Department of Internal Medicine, Hospital Garcia de Orta, Almada, Portugal
| | - Patrícia Pereira
- Department of Internal Medicine, Hospital Garcia de Orta, Almada, Portugal
| | - Andreia Nunes
- Department of Internal Medicine, Hospital Garcia de Orta, Almada, Portugal
| | - Pedro Antunes
- Endocrinology department of the Portuguese Oncology Institute, Lisbon, Portugal
| |
Collapse
|
42
|
Amanna IJ, Slifka MK. Questions regarding the safety and duration of immunity following live yellow fever vaccination. Expert Rev Vaccines 2016; 15:1519-1533. [PMID: 27267203 PMCID: PMC5171234 DOI: 10.1080/14760584.2016.1198259] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 06/02/2016] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The World Health Organization (WHO) and other health agencies have concluded that yellow fever booster vaccination is unnecessary since a single dose of vaccine confers lifelong immunity. Areas covered: We reviewed the clinical studies cited by health authorities in their investigation of both the safety profile and duration of immunity for the YFV-17D vaccine and examined the position that booster vaccination is no longer needed. We found that antiviral immunity may be lost in 1-in-3 to 1-in-5 individuals within 5 to 10 years after a single vaccination and that children may be at greater risk for primary vaccine failure. The safety profile of YFV-17D was compared to other licensed vaccines including oral polio vaccine (OPV) and the rotavirus vaccine, RotaShield, which have subsequently been withdrawn from the US and world market, respectively. Expert commentary: Based on these results and recent epidemiological data on vaccine failures (particularly evident at >10 years after vaccination), we believe that current recommendations to no longer administer YFV-17D booster vaccination be carefully re-evaluated, and that further development of safer vaccine approaches should be considered.
Collapse
Affiliation(s)
- Ian J. Amanna
- Najít Technologies, Inc., 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Mark K. Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| |
Collapse
|
43
|
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: 3.2] [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
|
44
|
Abstract
INTRODUCTION To assure the highest safety of immunization programs, detect adverse events following immunization (AEFIs), eliminate concerns, and reduce the risk of low vaccination coverage, authorities in industrialized countries have collected years of reports of suspected AEFIs and have systematically assessed their clinical importance. AREAS COVERED In this paper, the methods used to assess vaccine safety and the results obtained by the analysis of reports, studies, and meta-analyses are discussed. EXPERT OPINION Severe AEFIs are rare, and all evaluations of safety of vaccines recommended for both children and adults have demonstrated that the advantages of vaccines are always significantly higher than the problems that they cause, and there is no need to modify recommendations. However, the definition of AEFI is dependent on the vaccines themselves, complicating the definition of an AEFI and explaining why doubts and concerns have been raised. Presently, disease epidemiology data collected in healthy people and in subjects with underlying disease, general vaccine coverage, and the vaccination status of subjects with AEFIs are managed by many independent institutions. Only strict co-operation between these institutions will lead to the successful identification of AEFIs and to a reduction of the weight of anti-vaccine arguments.
Collapse
Affiliation(s)
- Nicola Principi
- a Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| | - Susanna Esposito
- a Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation , Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico , Milan , Italy
| |
Collapse
|
45
|
Leung WS, Chan MC, Chik SH, Tsang TY. First case of yellow fever vaccine-associated viscerotropic disease (YEL-AVD) in Hong Kong. J Travel Med 2016; 23:taw020. [PMID: 27087559 DOI: 10.1093/jtm/taw020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/14/2016] [Indexed: 11/13/2022]
Abstract
Yellow fever is an important and potentially fatal infection in tropical regions of Africa, South America, eastern Panama in Central America and Trinidad in the Caribbean. Yellow fever vaccination is not only crucial to reduce the disease risk and mortality in individuals travelling to these areas, but also an important public health measure to prevent the spread of the disease. Despite generally considered as a safe vaccine, yellow fever vaccine can rarely be associated with severe adverse reactions including yellow fever vaccine-associated viscerotropic disease (YEL-AVD). Here, we report the first case of YEL-AVD in Hong Kong. Clinicians should alert to the possibility of YEL-AVD in vaccinees presenting with compatible symptoms after yellow fever vaccination, particularly in people at higher risk of adverse events.
Collapse
Affiliation(s)
- Wai Shing Leung
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, China
| | - Man Chun Chan
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, China
| | - Shiu Hong Chik
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, China
| | - Tak Yin Tsang
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong, China
| |
Collapse
|
46
|
Tanizaki R, Ujiie M, Hori N, Kanagawa S, Kutsuna S, Takeshita N, Hayakawa K, Kato Y, Ohmagari N. Comparative study of adverse events after yellow fever vaccination between elderly and non-elderly travellers: questionnaire survey in Japan over a 1-year period. J Travel Med 2016; 23:taw012. [PMID: 27021495 DOI: 10.1093/jtm/taw012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2016] [Indexed: 11/14/2022]
Abstract
BACKGROUND A live attenuated yellow fever (YF) vaccination is required of all travellers visiting countries where YF virus is endemic. Although the risk of serious adverse events (AEs) after YF vaccination is known to be greater in elderly people than in younger people, information about other AEs among elderly travellers is lacking. METHODS A prospective observational questionnaire study was conducted to investigate the occurrence of AEs after YF vaccination in travellers who attended a designated YF vaccination centre in Tokyo, Japan, from 1 November 2011 to 31 October 2012. A questionnaire enquiring about any AEs experienced in the 2 weeks following YF vaccination was distributed to all vaccinees enrolled in this study, and responses were collected subsequently by mail or phone. For child vaccinees, their parents were allowed to respond in their stead. RESULTS Of the 1298 vaccinees who received the YF vaccine, 1044 (80.4%) were enrolled in the present study and 666 (63.8%) responded to the questionnaire. Of these 666 respondents, 370 (55.6%) reported AEs, of which 258 (38.7%) were systemic and 230 (34.5%) were local. No severe AEs associated with YF vaccination were reported. Elderly vaccinees (aged ≥60 years) reported fewer total AEs than those aged <60 years (42.9% vs 60.3%;P < 0.001). CONCLUSION Our study showed that fewer general AEs after yellow vaccination reported among elderly vaccinees than among non-elderly vaccinees. These results could provide supplementary information for judging the adaptation of vaccination in elderly travellers.
Collapse
Affiliation(s)
- Ryutaro Tanizaki
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mugen Ujiie
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Narumi Hori
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shuzo Kanagawa
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Satoshi Kutsuna
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Nozomi Takeshita
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kayoko Hayakawa
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yasuyuki Kato
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
47
|
Van Hoeven N, Joshi SW, Nana GI, Bosco-Lauth A, Fox C, Bowen RA, Clements DE, Martyak T, Parks DE, Baldwin S, Reed SG, Coler RN. A Novel Synthetic TLR-4 Agonist Adjuvant Increases the Protective Response to a Clinical-Stage West Nile Virus Vaccine Antigen in Multiple Formulations. PLoS One 2016; 11:e0149610. [PMID: 26901122 PMCID: PMC4762984 DOI: 10.1371/journal.pone.0149610] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/02/2016] [Indexed: 01/27/2023] Open
Abstract
West Nile virus (WNV) is a mosquito-transmitted member of the Flaviviridae family that has emerged in recent years to become a serious public health threat. Given the sporadic nature of WNV epidemics both temporally and geographically, there is an urgent need for a vaccine that can rapidly provide effective immunity. Protection from WNV infection is correlated with antibodies to the viral envelope (E) protein, which encodes receptor binding and fusion functions. Despite many promising E-protein vaccine candidates, there are currently none licensed for use in humans. This study investigates the ability to improve the immunogenicity and protective capacity of a promising clinical-stage WNV recombinant E-protein vaccine (WN-80E) by combining it with a novel synthetic TLR-4 agonist adjuvant. Using the murine model of WNV disease, we find that inclusion of a TLR-4 agonist in either a stable oil-in-water emulsion (SE) or aluminum hydroxide (Alum) formulation provides both dose and dosage sparing functions, whereby protection can be induced after a single immunization containing only 100 ng of WN-80E. Additionally, we find that inclusion of adjuvant with a single immunization reduced viral titers in sera to levels undetectable by viral plaque assay. The enhanced protection provided by adjuvanted immunization correlated with induction of a Th1 T-cell response and the resultant shaping of the IgG response. These findings suggest that inclusion of a next generation adjuvant may greatly enhance the protective capacity of WNV recombinant subunit vaccines, and establish a baseline for future development.
Collapse
Affiliation(s)
- Neal Van Hoeven
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle, WA 98103, United States of America
- * E-mail:
| | - Sharvari Waghmare Joshi
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle, WA 98103, United States of America
| | - Ghislain Ismael Nana
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle, WA 98103, United States of America
| | - Angela Bosco-Lauth
- Colorado State University Department of Biomedical Sciences, Foothills Campus, Fort Collins, CO 80523, United States of America
| | - Christopher Fox
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle, WA 98103, United States of America
| | - Richard A. Bowen
- Colorado State University Department of Biomedical Sciences, Foothills Campus, Fort Collins, CO 80523, United States of America
| | - David E. Clements
- Hawaii Biotech Inc. 99-193 Aiea Heights Drive, Aiea, Hawaii 96701, United States of America
| | - Timothy Martyak
- Hawaii Biotech Inc. 99-193 Aiea Heights Drive, Aiea, Hawaii 96701, United States of America
| | - D. Elliot Parks
- Hawaii Biotech Inc. 99-193 Aiea Heights Drive, Aiea, Hawaii 96701, United States of America
| | - Susan Baldwin
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle, WA 98103, United States of America
| | - Steven G. Reed
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle, WA 98103, United States of America
| | - Rhea N. Coler
- Infectious Disease Research Institute, 1616 Eastlake Ave E., Seattle, WA 98103, United States of America
| |
Collapse
|
48
|
Vaccination against Viruses. ENCYCLOPEDIA OF IMMUNOBIOLOGY 2016. [PMCID: PMC7152391 DOI: 10.1016/b978-0-12-374279-7.14016-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Most vaccines in use today are the result of empirical development. The mechanism of action of many vaccines in common use remains incompletely understood. Understanding how such vaccines protect is an ongoing subject of study using increasingly sophisticated immunological tools, such as B cell and T cell repertoire and transcriptome analysis. Such tools are also being applied to the design of vaccines against those viral targets that have evaded vaccine-mediated protection thus far. As basic immunological science intersects with the practicalities of assuring vaccine safety, tolerability, efficacy, and consistency in the clinic, the practical utility of more sophisticated immunological measures for vaccine development may be determined by whether they can be reduced to simply executed, highly standardized, reproducible assays with outcomes that have clear interpretations for vaccine development and use. Basic immunology, empirical vaccine testing, and regulatory science are all necessary contributors to developing the next generation of vaccines, including vaccines effective against the pathogens for which vaccines are not currently available.
Collapse
|
49
|
Observational study on immune response to yellow fever and measles vaccines in 9 to 15-month old children. Is it necessary to wait 4 weeks between two live attenuated vaccines? Vaccine 2015; 33:2301-6. [DOI: 10.1016/j.vaccine.2015.03.069] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Revised: 03/20/2015] [Accepted: 03/23/2015] [Indexed: 11/24/2022]
|
50
|
de Menezes Martins R, da Luz Fernandes Leal M, Homma A. Serious adverse events associated with yellow fever vaccine. Hum Vaccin Immunother 2015; 11:2183-7. [PMID: 26090855 PMCID: PMC4635904 DOI: 10.1080/21645515.2015.1022700] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 02/20/2015] [Indexed: 01/27/2023] Open
Abstract
Yellow fever vaccine was considered one of the safest vaccines, but in recent years it was found that it could rarely cause invasive and disseminated disease in some otherwise healthy individuals, with high lethality. After extensive studies, although some risk factors have been identified, the real cause of causes of this serious adverse event are largely unknown, but findings point to individual host factors. Meningoencephalitis, once considered to happen only in children less than 6 months of age, has also been identified in older children and adults, but with good prognosis. Efforts are being made to develop a safer yellow fever vaccine, and an inactivated vaccine or a vaccine prepared with the vaccine virus envelope produced in plants are being tested. Even with serious and rare adverse events, yellow fever vaccine is the best way to avoid yellow fever, a disease of high lethality and should be used routinely in endemic areas, and on people from non-endemic areas that could be exposed, according to a careful risk-benefit analysis.
Collapse
Affiliation(s)
| | | | - Akira Homma
- Bio-Manguinhos/Fiocruz; Rio de Janeiro, Brazil
| |
Collapse
|