1
|
Guo W, Jiang T, Rao J, Zhang Z, Zhang X, Su J, Yin C, Lu M, Hu X, Shan C. A safer cell-based yellow fever live attenuated vaccine protects mice against YFV infection. iScience 2024; 27:110972. [PMID: 39398246 PMCID: PMC11470684 DOI: 10.1016/j.isci.2024.110972] [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] [Received: 05/02/2024] [Revised: 07/07/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
The live attenuated yellow fever vaccine (YF17D) has caused controversial safety issues in history with low-yield problems, which has led to a large population being unable to be vaccinated and vaccine shortage in facing recent outbreaks. Here, we report a safer live attenuated vaccine candidate, YF17D-Δ77, which contains 77 nucleotides deletion in the 3' untranslated region (3' UTR) of the YF17D genome. YF17D-Δ77 exhibited no neurotropism and decreased viscerotropism and caused significantly lower lethality in mice compared to YF17D. Mechanistically, the deletion enhanced the sensitivity of the virus to type I and type II interferon responses, which hindered viral replication. Encouragingly, YF17D-Δ77 provided comparable immune protection in mice as did YF17D. Even 10 PFU of YF17D-Δ77 completely protected mice against YFV-Asibi challenge. In addition, the Δ77 mutation showed excellent stability after successive passages in Vero cells. Collectively, the data suggest that further development of YF17D-Δ77 as vaccine candidate is warranted.
Collapse
Affiliation(s)
- Weiwei Guo
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Tingting Jiang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Juhong Rao
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Zihan Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Xuekai Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Jiaoling Su
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Chunhong Yin
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Mingqing Lu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Xue Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Chao Shan
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
- Hubei Jiangxia Laboratory, Wuhan 430200, China
| |
Collapse
|
2
|
Pagnon A, Carre C, Aguirre M, Chautard E, Gimenez S, Raynal F, Feroldi E, Scott P, Modjarrad K, Vangelisti M, Mantel N. Next generation yellow fever vaccine induces an equivalent immune and transcriptomic profile to the current vaccine: observations from a phase I randomised clinical trial. EBioMedicine 2024; 108:105332. [PMID: 39293214 PMCID: PMC11424963 DOI: 10.1016/j.ebiom.2024.105332] [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: 04/10/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Yellow fever (YF), a mosquito-borne acute viral haemorrhagic illness, is endemic to many tropical and subtropical areas of Africa and Central and South America. Vaccination remains the most effective prevention strategy; however, as repeated outbreaks have exhausted vaccine stockpiles, there is a need for improved YF vaccines to meet global demand. A live-attenuated YF vaccine candidate (referred to as vYF) cloned from a YF-17D vaccine (YF-VAX®) sub-strain, adapted for growth in Vero cells cultured in serum-free media, is in clinical development. We report the innate and adaptive immune responses and the transcriptome profile of selected genes induced by vYF. METHODS Healthy adults aged 18-60 years were randomised at a 1:1:1:1 ratio to receive one dose of vYF at 4, 5 or 6 Log CCID50 or YF-VAX (reference vaccine), administered subcutaneously in the upper arm (ClinicalTrials.gov identifier: NCT04142086). Blood/serum samples were obtained at scheduled time points through 180 days (D180) post-vaccination. The surrogate endpoints assessed were: serum cytokine/chemokine concentrations, measured by bead-based Multiplex assay; peripheral blood vYF-specific IgG and IgM memory B cell frequencies, measured by FluoroSpot assay; and expression of genes involved in the immune response to YF-17D vaccination by RT-qPCR. FINDINGS There was no increase in any of the cytokine/chemokine concentrations assessed through D14 following vaccination with vYF or YF-VAX, except for a slight increase in IP-10 (CXCL10) levels. The gene expression profiles and kinetics following vaccination with vYF and YF-VAX were similar, inclusive of innate (antiviral responses [type-1 interferon, IFN signal transduction; interferon-stimulated genes], activated dendritic cells, viral sensing pattern recognition receptors) and adaptive (cell division in stimulated CD4+ T cells, B cell and antibody) immune signatures, which peaked at D7 and D14, respectively. Increases in vYF-specific IgG and IgM memory B cell frequencies at D28 and D180 were similar across the study groups. INTERPRETATION vYF-induced strong innate and adaptive immune responses comparable to those induced by YF-VAX, with similar transcriptomic and kinetic profiles observed. FUNDING Sanofi.
Collapse
Affiliation(s)
- Anke Pagnon
- Vaccine Research and Development, Sanofi, Marcy l'Etoile, France
| | - Christophe Carre
- Vaccine Research and Development, Sanofi, Marcy l'Etoile, France
| | - Marion Aguirre
- Vaccine Research and Development, Sanofi, Marcy l'Etoile, France
| | - Emilie Chautard
- Vaccine Research and Development, Sanofi, Marcy l'Etoile, France
| | - Sophie Gimenez
- Vaccine Research and Development, Sanofi, Marcy l'Etoile, France
| | - Franck Raynal
- Vaccine Research and Development, Sanofi, Marcy l'Etoile, France
| | - Emmanuel Feroldi
- Vaccine Research and Development, Sanofi, Marcy l'Etoile, France
| | - Paul Scott
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | | | - Nathalie Mantel
- Vaccine Research and Development, Sanofi, Marcy l'Etoile, France.
| |
Collapse
|
3
|
Modjarrad K, Scott PT, McCauley M, Ober-Shepherd B, Sondergaard E, Amare MF, Parikh AP, Omar B, Minutello AM, Adhikarla H, Wu Y, P AR, Delore V, Mantel N, Morrison MN, Kourbanova KS, Martinez ME, Guzman I, Greenleaf ME, Darden JM, Koren MA, Hamer MJ, Lee CE, Hutter JN, Peel SA, Robb ML, Vangelisti M, Feroldi E. Safety and immunogenicity of a next-generation live-attenuated yellow fever vaccine produced in a Vero cell line in the USA: a phase 1 randomised, observer-blind, active-controlled, dose-ranging clinical trial. THE LANCET. INFECTIOUS DISEASES 2024:S1473-3099(24)00406-7. [PMID: 39153488 DOI: 10.1016/s1473-3099(24)00406-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Recent outbreaks between 2015-17 and production delays have led to a yellow fever vaccine shortage. Therefore, there is an urgent need for new yellow fever vaccines with improved production scalability. A next-generation live-attenuated yellow fever vaccine candidate (vYF), produced in a Vero cell line has shown similar immunogenicity to licensed yellow fever vaccines in preclinical studies. In this study, we aimed to report the safety and immunogenicity of vYF in human clinical trial participants. METHODS In this first in-human, phase 1 randomised, observer-blind, active-controlled, dose-ranging clinical trial conducted at a single centre in the USA (Walter Reed Army Institute of Research, Silver Spring, MD, USA), 72 healthy adults (aged 18-60 years), without a known history of flavivirus infection or vaccination were randomly assigned (1:1:1:1) using interactive response technology to receive one dose of either vYF at 4, 5 or 6 Log CCID50 or the licensed YF-VAX (18 individuals per group). The primary outcomes were safety, neutralising antibody (NAb) titres through D180 post-vaccination in the per-protocol analysis set (comprised of yellow fever-naive participants who received their intended vaccine and provided a valid post-vaccination blood sample), and occurrence, and level of yellow fever viraemia in each vaccine group through D14 post-vaccination. FINDINGS All vYF doses had a safety and tolerability profile similar to YF-VAX. The most frequently reported solicited injection site reactions (vYF groups vs YF-VAX group) were pain (22% [12 of 54 participants, 95% CI 12-36] vs 28% [five of 18 participants, 10-54]), and erythema (13% [seven of 54 participants, 5-25] vs 39% [seven of 18 participants, 17-64]), with headache (32% [17 of 54 participants, 20-46] vs 44% [eight of 18 participants, 22-69]) and malaise (26% [14 of 54 participants, 15-40] vs 33% [six of 18 participants, 13-59]) as the most frequently reported solicited systemic reactions. One grade 3 solicited reaction (erythema) reported in the YF-VAX group resolved spontaneously. No serious unsolicited adverse events or deaths were reported. Viraemia was transiently detected in 50 participants between D4 and D10 in all groups and was observed in more participants or for a longer time in the vYF 6 Log CCID50 and YF-VAX groups. All yellow fever-naive vaccine recipients across the study groups seroconverted yielding four-fold increase from baseline in yellow fever NAb titres measured by yellow fever microneutralisation assay by D28 and were seroprotected with yellow fever NAb titres of at least 10 [1/dil]). Overall, 100% (18 of 18 participants, 95% CI 82-100), 89% (16 participants, 65-99), 100% (18 participants, 82-100), and 94% (17 participants, 73-100) of participants in the vYF 4 Log, vYF 5 Log, vYF 6 Log CCID50 groups, and YF-VAX group, respectively, remained seroprotected through D180. INTERPRETATION vYF has a similar safety and immunogenicity profile to YF-VAX. In general, the vYF 5 Log CCID50 dose appeared to show optimal viraemia, safety, and immunogenicity, and was chosen for subsequent development. FUNDING Sanofi.
Collapse
Affiliation(s)
- Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Paul T Scott
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Melanie McCauley
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Brittany Ober-Shepherd
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Erica Sondergaard
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Mihret F Amare
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Ajay P Parikh
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Badryah Omar
- Emerging Infectious Diseases Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | | | | | | | | | | | | | - Meshell N Morrison
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Kamila S Kourbanova
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Melissa E Martinez
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Ivelese Guzman
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Melissa E Greenleaf
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Janice M Darden
- Diagnostics and Countermeasures Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Michael A Koren
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Melinda J Hamer
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA; Department of Military and Emergency Medicine, Uniformed Services University, Bethesda, MD, USA; Department of Emergency Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Christine E Lee
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Jack N Hutter
- Clinical Trials Center, Walter Reed Army Institute of Research, Bethesda, MD, USA
| | - Sheila A Peel
- Diagnostics and Countermeasures Branch, Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Merlin L Robb
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | | | | |
Collapse
|
4
|
Liang Y, Dai X. The global incidence and trends of three common flavivirus infections (Dengue, yellow fever, and Zika) from 2011 to 2021. Front Microbiol 2024; 15:1458166. [PMID: 39206366 PMCID: PMC11349664 DOI: 10.3389/fmicb.2024.1458166] [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] [Received: 07/02/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Background Flavivirus pose a continued threat to global health, yet their worldwide burden and trends remain poorly quantified. We aimed to evaluate the global, regional, and national incidence of three common flavivirus infections (Dengue, yellow fever, and Zika) from 2011 to 2021. Methods Data on the number and rate of incidence for the three common flavivirus infection in 204 countries and territories were retrieved from the Global Burden of Diseases, Injuries, and Risk Factors Study (GBD) 2021. The estimated annual percent change (EAPC) was calculated to quantify the temporal trend during 2011-2016, 2016-2019, and 2019-2021, respectively. Results In 2021, an estimated 59,220,428 individuals were infected globally, comprising 58,964,185 cases of dengue, 86,509 cases of yellow fever, and 169,734 cases of Zika virus infection. The age-standardized incidence rate (ASIR) of the three common flavivirus infections increased by an annual average of 5.08% (95% CI 4.12 to 6.05) globally from 2011 to 2016, whereas decreased by an annual average of -8.37% (95% CI -12.46 to -4.08) per year between 2016 to 2019. The ASIR remained stable during 2019-2021, with an average change of 0.69% (95% CI -0.96 to 2.37) per year globally for the three common flavivirus infections. Regionally, the burden of the three common flavivirus infections was primarily concentrated in those regions with middle income, such as South Asia, Southeast Asia, and Tropical Latin America. Additionally, at the country level, there was an inverted "U" relationship between the SDI level and the ASI. Notably, an increase in the average age of infected cases has been observed worldwide, particularly in higher-income regions. Conclusion Flavivirus infections are an expanding public health concern worldwide, with considerable regional and demographic variation in the incidence. Policymakers and healthcare providers must stay vigilant regarding the impact of COVID-19 and other environmental factors on the risk of flavivirus infection and be prepared for potential future outbreaks.
Collapse
Affiliation(s)
- Yuanhao Liang
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Central Hospital, Jiangmen, China
| | - Xingzhu Dai
- Department of Stomatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Kelley M, Holmes CJ, Herbert C, Rayhan A, Joves J, Uhran M, Klaus L, Frigard R, Singh K, Limbach PA, Addepalli B, Benoit JB. Tyrosine transfer RNA levels and modifications during blood-feeding and vitellogenesis in the mosquito, Aedes aegypti. INSECT MOLECULAR BIOLOGY 2024. [PMID: 39105593 DOI: 10.1111/imb.12950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024]
Abstract
Mosquitoes such as Aedes aegypti must consume a blood meal for the nutrients necessary for egg production. Several transcriptome and proteome changes occur post-blood meal that likely corresponds with codon usage alterations. Transfer RNA (tRNA) is the adapter molecule that reads messenger RNA codons to add the appropriate amino acid during protein synthesis. Chemical modifications to tRNA enhance codon decoding, improving the accuracy and efficiency of protein synthesis. Here, we examined tRNA modifications and transcripts associated with the blood meal and subsequent periods of vitellogenesis in A. aegypti. More specifically, we assessed tRNA transcript abundance and modification levels in the fat body at critical times post blood-feeding. Based on a combination of alternative codon usage and identification of particular modifications, we discovered that increased transcription of tyrosine tRNAs is likely critical during the synthesis of egg yolk proteins in the fat body following a blood meal. Altogether, changes in both the abundance and modification of tRNA are essential factors in the process of vitellogenin production after blood-feeding in mosquitoes.
Collapse
Affiliation(s)
- Melissa Kelley
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Christopher J Holmes
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Cassandra Herbert
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| | - Asif Rayhan
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| | - Judd Joves
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Melissa Uhran
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lucas Klaus
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ronja Frigard
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Khwahish Singh
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| | - Patrick A Limbach
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio, USA
| | | | - Joshua B Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
6
|
Amanna IJ, Thomas A, Engelmann F, Hammarlund E, Raué HP, Bailey AL, Poore EA, Quintel BK, Lewis AD, Axthelm MK, Johnson AL, Colgin LMA, Diamond MS, Messaoudi I, Slifka MK. Development of a hydrogen peroxide-inactivated vaccine that protects against viscerotropic yellow fever in a non-human primate model. Cell Rep Med 2024; 5:101655. [PMID: 39019010 PMCID: PMC11293362 DOI: 10.1016/j.xcrm.2024.101655] [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: 11/13/2023] [Revised: 03/06/2024] [Accepted: 06/19/2024] [Indexed: 07/19/2024]
Abstract
Yellow fever virus (YFV) is endemic in >40 countries and causes viscerotropic disease with up to 20%-60% mortality. Successful live-attenuated yellow fever (YF) vaccines were developed in the mid-1930s, but their use is restricted or formally contraindicated in vulnerable populations including infants, the elderly, and people with compromised immune systems. In these studies, we describe the development of a next-generation hydrogen peroxide-inactivated YF vaccine and determine immune correlates of protection based on log neutralizing index (LNI) and neutralizing titer-50% (NT50) studies. In addition, we compare neutralizing antibody responses and protective efficacy of hydrogen peroxide-inactivated YF vaccine candidates to live-attenuated YFV-17D (YF-VAX) in a rhesus macaque model of viscerotropic YF. Our results indicate that an optimized, inactivated YF vaccine elicits protective antibody responses that prevent viral dissemination and lethal infection in rhesus macaques and may be a suitable alternative for vaccinating vulnerable populations who are not eligible to receive replicating live-attenuated YF vaccines.
Collapse
Affiliation(s)
- Ian J Amanna
- Najít Technologies, Inc., Beaverton, OR 97006, USA
| | - Archana Thomas
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Flora Engelmann
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, KY 40506, USA
| | - Erika Hammarlund
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Hans-Peter Raué
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Adam L Bailey
- Department of Pathology & Laboratory Medicine, University of Wisconsin - Madison, Madison, WI 53706, USA
| | | | | | - Anne D Lewis
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael K Axthelm
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, and The Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Amanda L Johnson
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Lois M A Colgin
- Division of Comparative Medicine, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, KY 40506, USA
| | - Mark K Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
7
|
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
|
8
|
Huebl L, Nnyombi A, Kihumuro A, Lukwago D, Walakira E, Kutalek R. Perceptions of yellow fever emergency mass vaccinations among vulnerable groups in Uganda: A qualitative study. PLoS Negl Trop Dis 2024; 18:e0012173. [PMID: 38739650 PMCID: PMC11115279 DOI: 10.1371/journal.pntd.0012173] [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: 02/10/2023] [Revised: 05/23/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Yellow fever (YF), a mosquito-borne viral hemorrhagic fever, is endemic in Uganda and causes frequent outbreaks. A total of 1.6 million people were vaccinated during emergency mass immunization campaigns in 2011 and 2016. This study explored local perceptions of YF emergency mass immunization among vulnerable groups to inform future vaccination campaigns. METHODOLOGY In this qualitative study, we conducted 43 semi-structured interviews, 4 focus group discussions, and 10 expert interviews with 76 participants. Data were collected in six affected districts with emergency mass vaccination. We included vulnerable groups (people ≥ 65 years and pregnant women) who are typically excluded from YF vaccination except during mass immunization. Data analysis was conducted using grounded theory. Inductive coding was utilized, progressing through open, axial, and selective coding. PRINCIPAL FINDINGS Participants relied on community sources for information about the YF mass vaccination. Information was disseminated door-to-door, in community spaces, during religious gatherings, and on the radio. However, most respondents had no knowledge of the vaccine, and it was unclear to them whether a booster dose was required. In addition, the simultaneous presidential election during the mass vaccination campaign led to suspicion and resistance to vaccination. The lack of reliable and trustworthy information and the politicization of vaccination campaigns reinforced mistrust of YF vaccines. CONCLUSIONS/SIGNIFICANCE People in remote areas affected by YF outbreaks rely on community sources of information. We therefore recommend improving health education, communication, and engagement through respected and trusted community members. Vaccination campaigns can never be seen as detached from political systems and power relations.
Collapse
Affiliation(s)
- Lena Huebl
- Unit Medical Anthropology and Global Health, Department of Social and Preventive Medicine, Center for Public Health, Medical University of Vienna, Vienna, Austria
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Aloysious Nnyombi
- Department of Social Work and Social Administration, Makerere University, Kampala, Uganda
| | - Aban Kihumuro
- Department of Nursing and Health Sciences, Bishop Stuart University, Mbarara, Uganda
| | - Denis Lukwago
- Cluster Monitoring and Evaluation Lead, Rakai Health Sciences Program, Masaka, Uganda
| | - Eddy Walakira
- Department of Social Work and Social Administration, Makerere University, Kampala, Uganda
| | - Ruth Kutalek
- Unit Medical Anthropology and Global Health, Department of Social and Preventive Medicine, Center for Public Health, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
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
|
10
|
Dong HL, Chen ZL, He MJ, Cui JZ, Cheng H, Wang QY, Xiong XH, Liu G, Chen HP. The Chimeric Chaoyang-Zika Vaccine Candidate Is Safe and Protective in Mice. Vaccines (Basel) 2024; 12:215. [PMID: 38400198 PMCID: PMC10893063 DOI: 10.3390/vaccines12020215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/06/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Zika virus (ZIKV) is an emerging flavivirus that causes congenital syndromes including microcephaly and fetal demise in pregnant women. No commercial vaccines against ZIKV are currently available. We previously generated a chimeric ZIKV (ChinZIKV) based on the Chaoyang virus (CYV) by replacing the prME protein of CYV with that of a contemporary ZIKV strain GZ01. Herein, we evaluated this vaccine candidate in a mouse model and showed that ChinZIKV was totally safe in both adult and suckling immunodeficient mice. No viral RNA was detected in the serum of mice inoculated with ChinZIKV. All of the mice inoculated with ChinZIKV survived, while mice inoculated with ZIKV succumbed to infection in 8 days. A single dose of ChinZIKV partially protected mice against lethal ZIKV challenge. In contrast, all the control PBS-immunized mice succumbed to infection after ZIKV challenge. Our results warrant further development of ChinZIKV as a vaccine candidate in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Gang Liu
- Academy of Military Medical Sciences, Beijing 100071, China
| | - Hui-Peng Chen
- Academy of Military Medical Sciences, Beijing 100071, China
| |
Collapse
|
11
|
Gabiane G, Bohers C, Mousson L, Obadia T, Dinglasan RR, Vazeille M, Dauga C, Viglietta M, Yébakima A, Vega-Rúa A, Gutiérrez Bugallo G, Gélvez Ramírez RM, Sonor F, Etienne M, Duclovel-Pame N, Blateau A, Smith-Ravin J, De Lamballerie X, Failloux AB. Evaluating vector competence for Yellow fever in the Caribbean. Nat Commun 2024; 15:1236. [PMID: 38336944 PMCID: PMC10858021 DOI: 10.1038/s41467-024-45116-2] [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: 09/22/2023] [Accepted: 01/16/2024] [Indexed: 02/12/2024] Open
Abstract
The mosquito-borne disease, Yellow fever (YF), has been largely controlled via mass delivery of an effective vaccine and mosquito control interventions. However, there are warning signs that YF is re-emerging in both Sub-Saharan Africa and South America. Imported from Africa in slave ships, YF was responsible for devastating outbreaks in the Caribbean. In Martinique, the last YF outbreak was reported in 1908 and the mosquito Aedes aegypti was incriminated as the main vector. We evaluated the vector competence of fifteen Ae. aegypti populations for five YFV genotypes (Bolivia, Ghana, Nigeria, Sudan, and Uganda). Here we show that mosquito populations from the Caribbean and the Americas were able to transmit the five YFV genotypes, with YFV strains for Uganda and Bolivia having higher transmission success. We also observed that Ae. aegypti populations from Martinique were more susceptible to YFV infection than other populations from neighboring Caribbean islands, as well as North and South America. Our vector competence data suggest that the threat of re-emergence of YF in Martinique and the subsequent spread to Caribbean nations and beyond is plausible.
Collapse
Affiliation(s)
- Gaelle Gabiane
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
- Université des Antilles, Ecole Doctorale 589, Schœlcher, Martinique, Marseille, France
| | - Chloé Bohers
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | - Laurence Mousson
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | - Thomas Obadia
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Marseille, France
- Institut Pasteur, Université Paris Cité, G5 Infectious Disease Epidemiology and Analytics, Paris, France
| | - Rhoel R Dinglasan
- University of Florida, Department of Infectious Diseases & Immunology and Emerging Pathogens Institute, College of Veterinary Medicine, Gainesville, FL, USA
| | - Marie Vazeille
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | - Catherine Dauga
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | - Marine Viglietta
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | | | - Anubis Vega-Rúa
- Institut Pasteur de Guadeloupe, Laboratory of Vector Control Research, Unit Transmission Reservoir and Pathogens Diversity, Les Abymes, Guadeloupe, Marseille, France
| | - Gladys Gutiérrez Bugallo
- Institut Pasteur de Guadeloupe, Laboratory of Vector Control Research, Unit Transmission Reservoir and Pathogens Diversity, Les Abymes, Guadeloupe, Marseille, France
- Department of Vector Control, Center for Research, Diagnostic, and Reference, Institute of Tropical Medicine Pedro Kouri, Havana, Cuba
| | - Rosa Margarita Gélvez Ramírez
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas, Fundación INFOVIDA, Bucaramanga, Colombia
- Unité des Virus Emergents (UVE), Aix Marseille Université, IRD 190, Inserm 1207, IHU Méditerranée Infection, Marseille, France
| | - Fabrice Sonor
- Centre de Démoustication et de Recherches Entomologiques, Lutte antivectorielle, Martinique, Marseille, France
- Agence Régionale de Santé, Direction de la Santé Publique, Martinique, Marseille, France
| | - Manuel Etienne
- Centre de Démoustication et de Recherches Entomologiques, Lutte antivectorielle, Martinique, Marseille, France
| | - Nathalie Duclovel-Pame
- Agence Régionale de Santé, Direction de la Santé Publique, Martinique, Marseille, France
| | - Alain Blateau
- Agence Régionale de Santé, Direction de la Santé Publique, Martinique, Marseille, France
| | - Juliette Smith-Ravin
- Groupe de recherche Biospheres Université des Antilles, Campus de Schœlcher, Martinique, Marseille, France
| | - Xavier De Lamballerie
- Unité des Virus Emergents (UVE), Aix Marseille Université, IRD 190, Inserm 1207, IHU Méditerranée Infection, Marseille, France
| | - Anna-Bella Failloux
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France.
| |
Collapse
|
12
|
Judson SD, Kenu E, Fuller T, Asiedu-Bekoe F, Biritwum-Nyarko A, Schroeder LF, Dowdy DW. Yellow fever in Ghana: Predicting emergence and ecology from historical outbreaks. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.29.24301911. [PMID: 38352600 PMCID: PMC10862978 DOI: 10.1101/2024.01.29.24301911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/25/2024]
Abstract
Understanding the epidemiology and ecology of yellow fever in endemic regions is critical for preventing future outbreaks. Ghana is a high-risk country for yellow fever. In this study we estimate the epidemiology, ecological cycles, and areas at risk for yellow fever in Ghana based on historical outbreaks. We identify 2371 cases and 887 deaths (case fatality rate 37.4%) from yellow fever reported in Ghana from 1910 to 2022. Since implementation of routine childhood vaccination in 1992, the estimated mean annual number of cases decreased by 81% and the geographic distribution of yellow fever cases also changed. While there have been multiple large historical outbreaks of yellow fever in Ghana from the urban cycle, recent outbreaks have originated among unvaccinated nomadic groups in rural areas with the sylvatic/savanna cycles. Using machine learning and an ecological niche modeling framework, we predict areas in Ghana that are similar to where prior yellow fever outbreaks have originated based on temperature, precipitation, landcover, elevation, and human population density. We find differences in predictions depending on the ecological cycles of outbreaks. Ultimately, these findings and methods could be used to inform further subnational risk assessments for yellow fever in Ghana and other high-risk countries.
Collapse
Affiliation(s)
- Seth D. Judson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ernest Kenu
- Department of Epidemiology, University of Ghana School of Public Health, Accra, Ghana
| | - Trevon Fuller
- Institute of the Environment and Sustainability, University of California Los Angeles, USA
| | | | | | - Lee F. Schroeder
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - David W. Dowdy
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Athni TS, Childs ML, Glidden CK, Mordecai EA. Temperature dependence of mosquitoes: comparing mechanistic and machine learning approaches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.04.569955. [PMID: 38105988 PMCID: PMC10723351 DOI: 10.1101/2023.12.04.569955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Mosquito vectors of pathogens (e.g., Aedes , Anopheles , and Culex spp. which transmit dengue, Zika, chikungunya, West Nile, malaria, and others) are of increasing concern for global public health. These vectors are geographically shifting under climate and other anthropogenic changes. As small-bodied ectotherms, mosquitoes are strongly affected by temperature, which causes unimodal responses in mosquito life history traits (e.g., biting rate, adult mortality rate, mosquito development rate, and probability of egg-to-adult survival) that exhibit upper and lower thermal limits and intermediate thermal optima in laboratory studies. However, it remains unknown how mosquito thermal responses measured in laboratory experiments relate to the realized thermal responses of mosquitoes in the field. To address this gap, we leverage thousands of global mosquito occurrences and geospatial satellite data at high spatial resolution to construct machine-learning based species distribution models, from which vector thermal responses are estimated. We apply methods to restrict models to the relevant mosquito activity season and to conduct ecologically-plausible spatial background sampling centered around ecoregions for comparison to mosquito occurrence records. We found that thermal minima estimated from laboratory studies were highly correlated with those from the species distributions (r = 0.90). The thermal optima were less strongly correlated (r = 0.69). For most species, we did not detect thermal maxima from their observed distributions so were unable to compare to laboratory-based estimates. The results suggest that laboratory studies have the potential to be highly transportable to predicting lower thermal limits and thermal optima of mosquitoes in the field. At the same time, lab-based models likely capture physiological limits on mosquito persistence at high temperatures that are not apparent from field-based observational studies but may critically determine mosquito responses to climate warming.
Collapse
|
14
|
Kelley M, Holmes CJ, Herbert C, Rayhan A, Joves J, Uhran M, Frigard R, Singh K, Limbach PA, Addepalli B, Benoit JB. Tyrosine transfer RNA levels and modifications during blood-feeding and vitellogenesis in the mosquito, Aedes aegypti. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569187. [PMID: 38076852 PMCID: PMC10705485 DOI: 10.1101/2023.11.29.569187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Mosquitoes such as Aedes aegypti must consume a blood meal for the nutrients necessary for egg production. Several transcriptome and proteome changes occur post blood meal that likely corresponds with codon usage alterations. Transfer RNA (tRNA) is the adapter molecule that reads messenger RNA (mRNA) codons to add the appropriate amino acid during protein synthesis. Chemical modifications to tRNA enhance codons' decoding, improving the accuracy and efficiency of protein synthesis. Here, we examined tRNA modifications and transcripts associated with the blood meal and subsequent periods of vitellogenesis in A. aegypti. More specifically, we assessed tRNA transcript abundance and modification levels in the fat body at critical times post blood-feeding. Based on a combination of alternative codon usage and identification of particular modifications, we identified that increased transcription of tyrosine tRNAs is likely critical during the synthesis of egg yolk proteins in the fat body following a blood meal. Altogether, changes in both the abundance and modification of tRNA are essential factors in the process of vitellogenin production after blood-feeding in mosquitoes.
Collapse
Affiliation(s)
- Melissa Kelley
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45211
| | | | - Cassandra Herbert
- Department of Chemistry, University of Cincinnati, Cincinnati, OH 45211
| | - Asif Rayhan
- Department of Chemistry, University of Cincinnati, Cincinnati, OH 45211
| | - Judd Joves
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45211
| | - Melissa Uhran
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45211
| | - Ronja Frigard
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45211
| | - Khwahish Singh
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45211
| | | | | | - Joshua B. Benoit
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45211
| |
Collapse
|
15
|
Zhang Y, Yang R, Yuan G, Li W, Cui Z, Xiao Z, Dong X, Yang H, Liu X, Zhang L, Hou Y, Liu M, Liu S, Hao Y, Zhang Y, Zheng X. Enhancing Inactivated Yellow Fever 17D Vaccine-Induced Immune Responses in Balb/C Mice Using Alum/CpG. Vaccines (Basel) 2023; 11:1744. [PMID: 38140149 PMCID: PMC10747526 DOI: 10.3390/vaccines11121744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/24/2023] Open
Abstract
There are some concerns about the safety of live attenuated yellow fever vaccines (YF-live), particularly viscerotropic adverse events, which have a high mortality rate. The cellular production of the vaccine will not cause these adverse effects and has the potential to extend applicability to those who have allergic reactions, immunosuppression, and age. In this study, inactivated yellow fever (YF) was prepared and adsorbed with Alum/CpG. The cellular and humoral immunities were investigated in a mouse model. The results showed that Alum/CpG (20 μg/mL) could significantly increase the binding and neutralizing activities of the antibodies against YF. Moreover, the antibody level at day 28 after one dose was similar to that of the attenuated vaccine, but significantly higher after two doses. At the same time, Alum/CpG significantly increased the levels of IFN-γ and IL-4 cytokines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yuntao Zhang
- Beijing Institute of Biological Products Company Limited, Beijing 100170, China; (Y.Z.); (R.Y.); (G.Y.); (W.L.); (Z.C.); (Z.X.); (X.D.); (H.Y.); (X.L.); (L.Z.); (Y.H.); (M.L.); (S.L.); (Y.H.)
| | - Xiaotong Zheng
- Beijing Institute of Biological Products Company Limited, Beijing 100170, China; (Y.Z.); (R.Y.); (G.Y.); (W.L.); (Z.C.); (Z.X.); (X.D.); (H.Y.); (X.L.); (L.Z.); (Y.H.); (M.L.); (S.L.); (Y.H.)
| |
Collapse
|
16
|
Kazakova E, Lane TR, Jones T, Puhl AC, Riabova O, Makarov V, Ekins S. 1-Sulfonyl-3-amino-1 H-1,2,4-triazoles as Yellow Fever Virus Inhibitors: Synthesis and Structure-Activity Relationship. ACS OMEGA 2023; 8:42951-42965. [PMID: 38024733 PMCID: PMC10653066 DOI: 10.1021/acsomega.3c06106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023]
Abstract
Yellow fever virus (YFV) transmitted by infected mosquitoes causes an acute viral disease for which there are no approved small-molecule therapeutics. Our recently developed machine learning models for YFV inhibitors led to the selection of a new pyrazolesulfonamide derivative RCB16003 with acceptable in vitro activity. We report that the N-phenyl-1-(phenylsulfonyl)-1H-1,2,4-triazol-3-amine class, which was recently identified as active non-nucleoside reverse transcriptase inhibitors against HIV-1, can also be repositioned as inhibitors of yellow fever virus replication. As compared to other Flaviviridae or Togaviridae family viruses tested, both compounds RCB16003 and RCB16007 demonstrate selectivity for YFV over related viruses, with only RCB16007 showing some inhibition of the West Nile virus (EC50 7.9 μM, CC50 17 μM, SI 2.2). We also describe the absorption, distribution, metabolism, and excretion (ADME) in vitro and pharmacokinetics (PK) for RCB16007 in mice. This compound had previously been shown to not inhibit hERG, and we now describe that it has good metabolic stability in mouse and human liver microsomes, low levels of CYP inhibition, high protein binding, and no indication of efflux in Caco-2 cells. A single-dose oral PK study in mice has a T1/2 of 3.4 h and Cmax of 1190 ng/mL, suggesting good availability and stability. We now propose that the N-phenyl-1-(phenylsulfonyl)-1H-1,2,4-triazol-3-amine class may be prioritized for in vivo efficacy testing against YFV.
Collapse
Affiliation(s)
- Elena Kazakova
- Federal
Research Centre “Fundamentals of Biotechnology” of the
Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
| | - Thomas R. Lane
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Thane Jones
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Ana C. Puhl
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Olga Riabova
- Federal
Research Centre “Fundamentals of Biotechnology” of the
Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
| | - Vadim Makarov
- Federal
Research Centre “Fundamentals of Biotechnology” of the
Russian Academy of Sciences (Research Centre of Biotechnology RAS), 33-2 Leninsky Prospect, 119071 Moscow, Russia
| | - Sean Ekins
- Collaborations
Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
17
|
Hansen CA, Staples JE, Barrett ADT. Fractional Dosing of Yellow Fever Live Attenuated 17D Vaccine: A Perspective. Infect Drug Resist 2023; 16:7141-7154. [PMID: 38023411 PMCID: PMC10640814 DOI: 10.2147/idr.s370013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/20/2023] [Indexed: 12/01/2023] Open
Abstract
Yellow fever virus (YFV) is a mosquito-borne flavivirus that causes over 109,000 severe infections and over 51,000 deaths annually in endemic areas of sub-Saharan Africa and tropical South America. The virus has a transmission cycle involving mosquitoes and humans or non-human primates (NHPs) as the vertebrate hosts. Although yellow fever (YF) is prevented by a live attenuated vaccine (strain 17D), recent epidemics in Angola, the Democratic Republic of the Congo (DRC), and Brazil put great pressure on vaccine stockpiles. This resulted in the World Health Organization (WHO) and Pan American Health Organization (PAHO) implementing, on an emergency basis only, off-label dose-sparing techniques and policies during 2016-2018 to protect as many people in DRC and Brazil as possible from disease during unexpected large outbreaks of YF. Subsequently non-inferiority studies involving full doses compared to fractional doses indicated promising results, leading some policy-makers and scientists to consider utilizing YF vaccine fractional doses in non-emergency scenarios. Although the additional data on the immunogenicity and safety of fractional doses are promising, there are several questions and considerations that remain regarding the use of fractional doses, including differences in the initial antibody kinetics, differences in the immune response in certain populations, and durability of the immune response to fractional doses compared to full doses. Until the remaining knowledge gaps are addressed, full doses instead of fractional doses should continue to be used unless there are insufficient doses of the vaccine available to control outbreaks of YF.
Collapse
Affiliation(s)
- Clairissa A Hansen
- Department of Pathology and Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555-4036, USA
| | - J Erin Staples
- Arboviral Diseases Branch, U.S. Centers for Disease Control and Prevention, Fort Collins, CO, USA
| | - Alan D T Barrett
- Department of Pathology and Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555-4036, USA
| |
Collapse
|
18
|
Lim AY, Jafari Y, Caldwell JM, Clapham HE, Gaythorpe KAM, Hussain-Alkhateeb L, Johansson MA, Kraemer MUG, Maude RJ, McCormack CP, Messina JP, Mordecai EA, Rabe IB, Reiner RC, Ryan SJ, Salje H, Semenza JC, Rojas DP, Brady OJ. A systematic review of the data, methods and environmental covariates used to map Aedes-borne arbovirus transmission risk. BMC Infect Dis 2023; 23:708. [PMID: 37864153 PMCID: PMC10588093 DOI: 10.1186/s12879-023-08717-8] [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: 06/14/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Aedes (Stegomyia)-borne diseases are an expanding global threat, but gaps in surveillance make comprehensive and comparable risk assessments challenging. Geostatistical models combine data from multiple locations and use links with environmental and socioeconomic factors to make predictive risk maps. Here we systematically review past approaches to map risk for different Aedes-borne arboviruses from local to global scales, identifying differences and similarities in the data types, covariates, and modelling approaches used. METHODS We searched on-line databases for predictive risk mapping studies for dengue, Zika, chikungunya, and yellow fever with no geographical or date restrictions. We included studies that needed to parameterise or fit their model to real-world epidemiological data and make predictions to new spatial locations of some measure of population-level risk of viral transmission (e.g. incidence, occurrence, suitability, etc.). RESULTS We found a growing number of arbovirus risk mapping studies across all endemic regions and arboviral diseases, with a total of 176 papers published 2002-2022 with the largest increases shortly following major epidemics. Three dominant use cases emerged: (i) global maps to identify limits of transmission, estimate burden and assess impacts of future global change, (ii) regional models used to predict the spread of major epidemics between countries and (iii) national and sub-national models that use local datasets to better understand transmission dynamics to improve outbreak detection and response. Temperature and rainfall were the most popular choice of covariates (included in 50% and 40% of studies respectively) but variables such as human mobility are increasingly being included. Surprisingly, few studies (22%, 31/144) robustly tested combinations of covariates from different domains (e.g. climatic, sociodemographic, ecological, etc.) and only 49% of studies assessed predictive performance via out-of-sample validation procedures. CONCLUSIONS Here we show that approaches to map risk for different arboviruses have diversified in response to changing use cases, epidemiology and data availability. We identify key differences in mapping approaches between different arboviral diseases, discuss future research needs and outline specific recommendations for future arbovirus mapping.
Collapse
Affiliation(s)
- Ah-Young Lim
- Department of Infectious Disease Epidemiology and Dynamics, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK.
- Centre for Mathematical Modelling of Infectious Diseases, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK.
| | - Yalda Jafari
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jamie M Caldwell
- High Meadows Environmental Institute, Princeton University, Princeton, NJ, USA
| | - Hannah E Clapham
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Katy A M Gaythorpe
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Laith Hussain-Alkhateeb
- School of Public Health and Community Medicine, Sahlgrenska Academy, Institute of Medicine, Global Health, University of Gothenburg, Gothenburg, Sweden
- Population Health Research Section, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Michael A Johansson
- Dengue Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan, Puerto Rico, USA
| | | | - Richard J Maude
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Clare P McCormack
- MRC Centre for Global Infectious Disease Analysis, School of Public Health, Imperial College London, London, UK
| | - Jane P Messina
- School of Geography and the Environment, University of Oxford, Oxford, UK
- Oxford School of Global and Area Studies, University of Oxford, Oxford, UK
| | - Erin A Mordecai
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Ingrid B Rabe
- Department of Epidemic and Pandemic Preparedness and Prevention, World Health Organization, Geneva, Switzerland
| | - Robert C Reiner
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
- Department of Health Metrics Sciences, School of Medicine, University of Washington, Seattle, WA, USA
| | - Sadie J Ryan
- Department of Geography and Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Henrik Salje
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Jan C Semenza
- Department of Public Health and Clinical Medicine, Section of Sustainable Health, Umeå University, Umeå, Sweden
| | - Diana P Rojas
- Department of Epidemic and Pandemic Preparedness and Prevention, World Health Organization, Geneva, Switzerland
| | - Oliver J Brady
- Department of Infectious Disease Epidemiology and Dynamics, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
- Centre for Mathematical Modelling of Infectious Diseases, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
19
|
Kulkarni A, Delgadillo FM, Gayathrinathan S, Grajeda BI, Roy S. Current Status of Omics Studies Elucidating the Features of Reproductive Biology in Blood-Feeding Insects. INSECTS 2023; 14:802. [PMID: 37887814 PMCID: PMC10607566 DOI: 10.3390/insects14100802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/28/2023]
Abstract
Female insects belonging to the genera Anopheles, Aedes, Glossina, and Rhodnius account for the majority of global vector-borne disease mortality. In response to mating, these female insects undergo several molecular, physiological, and behavioral changes. Studying the dynamic post-mating molecular responses in these insects that transmit human diseases can lead to the identification of potential targets for the development of novel vector control methods. With the continued advancements in bioinformatics tools, we now have the capability to delve into various physiological processes in these insects. Here, we discuss the availability of multiple datasets describing the reproductive physiology of the common blood-feeding insects at the molecular level. Additionally, we compare the male-derived triggers transferred during mating to females, examining both shared and species-specific factors. These triggers initiate post-mating genetic responses in female vectors, affecting not only their reproductive success but also disease transmission.
Collapse
Affiliation(s)
- Aditi Kulkarni
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA; (A.K.); (F.M.D.); (S.G.); (B.I.G.)
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Frida M. Delgadillo
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA; (A.K.); (F.M.D.); (S.G.); (B.I.G.)
- Environmental Science and Engineering Ph.D. Program, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Sharan Gayathrinathan
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA; (A.K.); (F.M.D.); (S.G.); (B.I.G.)
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Brian I. Grajeda
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA; (A.K.); (F.M.D.); (S.G.); (B.I.G.)
- Biosciences Ph.D. Program, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Sourav Roy
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA; (A.K.); (F.M.D.); (S.G.); (B.I.G.)
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
20
|
Hills SL, Wong JM, Staples JE. Arboviral vaccines for use in pregnant travelers. Travel Med Infect Dis 2023; 55:102624. [PMID: 37517630 DOI: 10.1016/j.tmaid.2023.102624] [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: 05/12/2023] [Revised: 06/20/2023] [Accepted: 07/26/2023] [Indexed: 08/01/2023]
Abstract
Pregnant women traveling abroad can be exposed to a variety of arboviruses, primarily spread by mosquitoes or ticks. Some arboviral infections can be of particular concern for pregnant women or their fetuses. Vaccination is one preventive measure that can reduce the risk for infection. Several arboviral vaccines have been licensed for many years and can be used to prevent infection in travelers, namely Japanese encephalitis, yellow fever, and tick-borne encephalitis vaccines. Recommendations on use of these vaccines in pregnancy vary. Other arboviral vaccines have been licensed but are not indicated for use in pregnant travelers (e.g., dengue vaccines) or are in development (e.g., chikungunya, Zika vaccines). This review describes arboviral vaccines for travelers, focusing on women who are pregnant and those planning travel during pregnancy.
Collapse
Affiliation(s)
- S L Hills
- Arboviral Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, USA.
| | - J M Wong
- Dengue Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, San Juan, PR, USA
| | - J E Staples
- Arboviral Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO, USA
| |
Collapse
|
21
|
Arjomand T, Herrán OF, Mantilla LC, Estupiñán MI, Lozano-Parra A, Gelvez RM, Herrera VM, Villar LA, Villamor E. Dietary Patterns and Anti-Flavivirus IgG Seroconversion in Colombian Children. Am J Trop Med Hyg 2023; 109:397-403. [PMID: 37339760 PMCID: PMC10397447 DOI: 10.4269/ajtmh.22-0619] [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: 09/22/2022] [Accepted: 05/21/2023] [Indexed: 06/22/2023] Open
Abstract
The burden of flaviviral infections, especially dengue and Zika, is high in the Americas. Malnutrition affects the risk and response to infections, but the role of diet on flaviviral infection risk is uncertain. The objective of this study was to investigate the relations between dietary patterns adherence and anti-flavivirus IgG seroconversion in children during a Zika epidemic in a dengue-endemic area of Colombia. In 2015-2016, we followed 424 anti-flavivirus IgG seronegative children aged 2 to 12 years for 1 year. Baseline data included children's sociodemographic, anthropometric, and dietary information collected through a 38-item food frequency questionnaire (FFQ). IgG testing was repeated at the end of follow-up. The primary exposure was adherence to each of four dietary patterns (animal foods, traditional, ultraprocessed foods, and prudent) that were identified from the FFQ through principal component analysis. Secondary exposures were intake frequencies of foods contributing to relevant patterns. We estimated risk of seroconversion by quartiles of adherence scores and compared them using relative risks (RR) and 95% CI from Poisson regression adjusted for sex, age, and socioeconomic status indicators. Seroconversion risk was 32.1%. Adherence to the traditional pattern was positively related to seroconversion. RR comparing fourth versus first quartiles of adherence was 1.52 (95% CI: 1.04-2.21; P trend = 0.02). Of the most representative foods in this pattern, potato and sugarcane water intake frequencies were related to increased seroconversion risk. In conclusion, adherence to a traditional foods pattern, including potatoes and sugarcane water, was positively associated with anti-flavivirus IgG seroconversion.
Collapse
Affiliation(s)
- Tina Arjomand
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Oscar F. Herrán
- Centro de Investigaciones Epidemiológicas, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Laura C. Mantilla
- Centro de Investigaciones Epidemiológicas, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga, Colombia
| | | | - Anyela Lozano-Parra
- Centro de Investigaciones Epidemiológicas, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga, Colombia
| | | | - Víctor M. Herrera
- Centro de Investigaciones Epidemiológicas, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga, Colombia
| | - Luis A. Villar
- Centro de Investigaciones Epidemiológicas, Facultad de Salud, Universidad Industrial de Santander, Bucaramanga, Colombia
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas, Bucaramanga, Colombia
| | - Eduardo Villamor
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan
| |
Collapse
|
22
|
Dong HL, He MJ, Wang QY, Cui JZ, Chen ZL, Xiong XH, Zhang LC, Cheng H, Xiong GQ, Hu A, Lu YY, Cheng CL, Meng ZX, Zhu C, Zhao G, Liu G, Chen HP. Rapid Generation of Recombinant Flaviviruses Using Circular Polymerase Extension Reaction. Vaccines (Basel) 2023; 11:1250. [PMID: 37515065 PMCID: PMC10383701 DOI: 10.3390/vaccines11071250] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The genus Flavivirus is a group of arthropod-borne single-stranded RNA viruses, which includes important human and animal pathogens such as Japanese encephalitis virus (JEV), Zika virus (ZIKV), Dengue virus (DENV), yellow fever virus (YFV), West Nile virus (WNV), and Tick-borne encephalitis virus (TBEV). Reverse genetics has been a useful tool for understanding biological properties and the pathogenesis of flaviviruses. However, the conventional construction of full-length infectious clones for flavivirus is time-consuming and difficult due to the toxicity of the flavivirus genome to E. coli. Herein, we applied a simple, rapid, and bacterium-free circular polymerase extension reaction (CPER) method to synthesize recombinant flaviviruses in vertebrate cells as well as insect cells. We started with the de novo synthesis of the JEV vaccine strain SA-14-14-2 in Vero cells using CPER, and then modified the CPER method to recover insect-specific flaviviruses (ISFs) in mosquito C6/36 cells. Chimeric Zika virus (ChinZIKV) based on the Chaoyang virus (CYV) backbone and the Culex flavivirus reporter virus expressing green fluorescent protein (CxFV-GFP) were subsequently rescued in C6/36 cells. CPER is a simple method for the rapid generation of flaviviruses and other potential RNA viruses. A CPER-based recovery system for flaviviruses of different host ranges was established, which would facilitate the development of countermeasures against flavivirus outbreaks in the future.
Collapse
Affiliation(s)
- Hao-Long Dong
- Academy of Military Medical Sciences, Beijing 100071, China
| | - Mei-Juan He
- Academy of Military Medical Sciences, Beijing 100071, China
| | - Qing-Yang Wang
- Academy of Military Medical Sciences, Beijing 100071, China
| | - Jia-Zhen Cui
- Academy of Military Medical Sciences, Beijing 100071, China
| | - Zhi-Li Chen
- Academy of Military Medical Sciences, Beijing 100071, China
| | | | | | - Hao Cheng
- Academy of Military Medical Sciences, Beijing 100071, China
| | - Guo-Qing Xiong
- Institutes of Physical Science and Information Technology, Anhui University, Hefei 230000, China
| | - Ao Hu
- Institutes of Physical Science and Information Technology, Anhui University, Hefei 230000, China
| | - Yuan-Yuan Lu
- Institutes of Physical Science and Information Technology, Anhui University, Hefei 230000, China
| | - Chun-Lin Cheng
- School of Life Science, Hebei University, Baoding 071000, China
| | - Zhi-Xin Meng
- School of Life Science, Hebei University, Baoding 071000, China
| | - Chen Zhu
- Academy of Military Medical Sciences, Beijing 100071, China
| | - Guang Zhao
- Academy of Military Medical Sciences, Beijing 100071, China
| | - Gang Liu
- Academy of Military Medical Sciences, Beijing 100071, China
| | - Hui-Peng Chen
- Academy of Military Medical Sciences, Beijing 100071, China
| |
Collapse
|
23
|
Ribeiro YP, Falcão LFM, Smith VC, de Sousa JR, Pagliari C, Franco ECS, Cruz ACR, Chiang JO, Martins LC, Nunes JAL, Vilacoert FSDS, Santos LCD, Furlaneto MP, Fuzii HT, Bertonsin Filho MV, da Costa LD, Duarte MIS, Furlaneto IP, Martins Filho AJ, Aarão TLDS, Vasconcelos PFDC, Quaresma JAS. Comparative Analysis of Human Hepatic Lesions in Dengue, Yellow Fever, and Chikungunya: Revisiting Histopathological Changes in the Light of Modern Knowledge of Cell Pathology. Pathogens 2023; 12:pathogens12050680. [PMID: 37242350 DOI: 10.3390/pathogens12050680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Arboviruses, such as yellow fever virus (YFV), dengue virus (DENV), and chikungunya virus (CHIKV), present wide global dissemination and a pathogenic profile developed in infected individuals, from non-specific clinical conditions to severe forms, characterised by the promotion of significant lesions in different organs of the harbourer, culminating in multiple organ dysfunction. An analytical cross-sectional study was carried out via the histopathological analysis of 70 samples of liver patients, collected between 2000 and 2017, with confirmed laboratory diagnoses, who died due to infection and complications due to yellow fever (YF), dengue fever (DF), and chikungunya fever (CF), to characterise, quantify, and compare the patterns of histopathological alterations in the liver between the samples. Of the histopathological findings in the human liver samples, there was a significant difference between the control and infection groups, with a predominance of alterations in the midzonal area of the three cases analysed. Hepatic involvement in cases of YF showed a greater intensity of histopathological changes. Among the alterations evaluated, cell swelling, microvesicular steatosis, and apoptosis were classified according to the degree of tissue damage from severe to very severe. Pathological abnormalities associated with YFV, DENV, and CHIKV infections showed a predominance of changes in the midzonal area. We also noted that, among the arboviruses studied, liver involvement in cases of YFV infection was more intense.
Collapse
Affiliation(s)
- Yasmin Pacheco Ribeiro
- Center for Biological and Health Sciences, State University of Pará, Belém 66087-662, PA, Brazil
| | - Luiz Fabio Magno Falcão
- Center for Biological and Health Sciences, State University of Pará, Belém 66087-662, PA, Brazil
| | - Vanessa Cavaleiro Smith
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | - Jorge Rodrigues de Sousa
- Center for Biological and Health Sciences, State University of Pará, Belém 66087-662, PA, Brazil
| | - Carla Pagliari
- School of Medicine, São Paulo University, São Paulo 01246-903, SP, Brazil
| | | | - Ana Cecília Ribeiro Cruz
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | - Janniffer Oliveira Chiang
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | - Livia Carício Martins
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | - Juliana Abreu Lima Nunes
- Section of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | | | - Lais Carneiro Dos Santos
- Section of Pathology, Evandro Chagas Institute, Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | | | - Hellen Thais Fuzii
- Tropical Medicine Center, Federal University of Pará, Belém 66055-240, PA, Brazil
| | | | - Luccas Delgado da Costa
- Section of Pathology, Evandro Chagas Institute, Ministry of Health, Ananindeua 67030-000, PA, Brazil
| | | | - Ismari Perini Furlaneto
- Center for Biological and Health Sciences, State University of Pará, Belém 66087-662, PA, Brazil
| | | | | | | | - Juarez Antônio Simões Quaresma
- Center for Biological and Health Sciences, State University of Pará, Belém 66087-662, PA, Brazil
- School of Medicine, São Paulo University, São Paulo 01246-903, SP, Brazil
- Tropical Medicine Center, Federal University of Pará, Belém 66055-240, PA, Brazil
| |
Collapse
|
24
|
Kampango A, Pinto J, Abílio AP, Machoe E, Matusse J, McCall PJ. Characterisation of human exposure to nocturnal biting by malaria and arbovirus vectors in a rural community in Chókwè district, southern Mozambique. Wellcome Open Res 2023; 8:193. [PMID: 37484481 PMCID: PMC10357080 DOI: 10.12688/wellcomeopenres.19278.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2023] [Indexed: 07/25/2023] Open
Abstract
Background: Understanding the magnitude of human exposure to mosquito biting is fundamental to reduce pathogen transmission. Here we report on a study quantifying the levels of mosquitoes attacking humans throughout the night in a rural area of Southern Mozambique. Methods: Surveys were carried out in Massavasse village, southern Mozambique. The abundance and composition of host-seeking mosquito communities at night were assessed by human-landing catches (HLC) at one-hour intervals. Periods when people were located predominantly outdoors or indoors were used to estimate the amount of residents' exposure to mosquito bites in either location, to explore the potential impact a bed net could have had in reducing biting by each vector species. Results: A total of 69,758 host-seeking female mosquitoes comprising 23 species in four genera were collected. The exposure to biting by virtually all vector species was consistently high outdoors, typically at early evening and morning, with exception of An. gambiae s.l which was likely of biting a person with nearly same intensity indoors and outdoors throughout the night. Bed nets use could have reduced biting by An. gambiae s.l (dominated by An. arabiensis), Ma. africana, Ma. uniformis, Cx. pipiens, Cx. antennatus, and Cx. poicilipes by 53%, 47%, 46%, 38%, 31%, and 28% respectively, compared to non-users. Conversely, a bed net user would have had little protection against An. pharoensis, An. ziemanni, An. tenebrosus, and Cx. tritaeniorhynchus biting exposures. Conclusions: This study showed that Massavasse residents were exposed to high levels of outdoor biting by malaria and arbovirus vectors that abound in the village. The findings help to identify entomological drivers of persistent malaria transmission in Mozambique and identify a wide range of arbovirus vectors nocturnally active in rural areas, many with outbreak potential. The study highlights the need for a surveillance system for monitoring arboviral diseases vectors in Mozambique.
Collapse
Affiliation(s)
- Ayubo Kampango
- Sector de Estudo de Vectores, Instituto Nacional de Saúde (INS), Maputo, Villa de Marracuene EN1, Plot 3943, Mozambique
- Department of Zoology and Entomology, University of Pretoria, Pretoria, Hatfield, 0028, South Africa
| | - João Pinto
- Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine (IHMT), Lisbon, Rua da Junqueira, 100 1349-008, Portugal
| | - Ana Paula Abílio
- Sector de Estudo de Vectores, Instituto Nacional de Saúde (INS), Maputo, Villa de Marracuene EN1, Plot 3943, Mozambique
| | - Elias Machoe
- Sector de Estudo de Vectores, Instituto Nacional de Saúde (INS), Maputo, Villa de Marracuene EN1, Plot 3943, Mozambique
| | - Júlio Matusse
- Sector de Estudo de Vectores, Instituto Nacional de Saúde (INS), Maputo, Villa de Marracuene EN1, Plot 3943, Mozambique
| | - Philip J. McCall
- Vector Biology Department, Liverpool School of Tropical Medicine (LSTM), Liverpool, Pembroke Place, L3 5QA, UK
| |
Collapse
|
25
|
Li D, Lu HT, Ding YZ, Wang HJ, Ye JL, Qin CF, Liu ZY. Specialized cis-Acting RNA Elements Balance Genome Cyclization to Ensure Efficient Replication of Yellow Fever Virus. J Virol 2023; 97:e0194922. [PMID: 37017533 PMCID: PMC10134800 DOI: 10.1128/jvi.01949-22] [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: 12/19/2022] [Accepted: 03/13/2023] [Indexed: 04/06/2023] Open
Abstract
Genome cyclization is essential for viral RNA (vRNA) replication of the vertebrate-infecting flaviviruses, and yet its regulatory mechanisms are not fully understood. Yellow fever virus (YFV) is a notorious pathogenic flavivirus. Here, we demonstrated that a group of cis-acting RNA elements in YFV balance genome cyclization to govern efficient vRNA replication. It was shown that the downstream of the 5'-cyclization sequence hairpin (DCS-HP) is conserved in the YFV clade and is important for efficient YFV propagation. By using two different replicon systems, we found that the function of the DCS-HP is determined primarily by its secondary structure and, to a lesser extent, by its base-pair composition. By combining in vitro RNA binding and chemical probing assays, we found that the DCS-HP orchestrates the balance of genome cyclization through two different mechanisms, as follows: the DCS-HP assists the correct folding of the 5' end in a linear vRNA to promote genome cyclization, and it also limits the overstabilization of the circular form through a potential crowding effect, which is influenced by the size and shape of the DCS-HP structure. We also provided evidence that an A-rich sequence downstream of the DCS-HP enhances vRNA replication and contributes to the regulation of genome cyclization. Interestingly, diversified regulatory mechanisms of genome cyclization, involving both the downstream of the 5'-cyclization sequence (CS) and the upstream of the 3'-CS elements, were identified among different subgroups of the mosquito-borne flaviviruses. In summary, our work highlighted how YFV precisely controls the balance of genome cyclization to ensure viral replication. IMPORTANCE Yellow fever virus (YFV), the prototype of the Flavivirus genus, can cause devastating yellow fever disease. Although it is preventable by vaccination, there are still tens of thousands of yellow fever cases per year, and no approved antiviral medicine is available. However, the understandings about the regulatory mechanisms of YFV replication are obscure. In this study, by a combination of bioinformatics, reverse genetics, and biochemical approaches, it was shown that the downstream of the 5'-cyclization sequence hairpin (DCS-HP) promotes efficient YFV replication by modulating the conformational balance of viral RNA. Interestingly, we found specialized combinations for the downstream of the 5'-cyclization sequence (CS) and upstream of the 3'-CS elements in different groups of the mosquito-borne flaviviruses. Moreover, possible evolutionary relationships among the various downstream of the 5'-CS elements were implied. This work highlighted the complexity of RNA-based regulatory mechanisms in the flaviviruses and will facilitate the design of RNA structure-targeted antiviral therapies.
Collapse
Affiliation(s)
- Dan Li
- The Centre for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Hai-Tao Lu
- The Centre for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yu-Zhen Ding
- The Centre for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Hong-Jiang Wang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- The Chinese People’s Liberation Army Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Jing-Long Ye
- The Centre for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Zhong-Yu Liu
- The Centre for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, China
| |
Collapse
|
26
|
Ricciardi MJ, Rust LN, Pedreño-Lopez N, Yusova S, Biswas S, Webb GM, Gonzalez-Nieto L, Voigt TB, Louw JJ, Laurino FD, DiBello JR, Raué HP, Barber-Axthelm AM, Chun K, Uttke S, Raphael LMS, Yrizarry-Medina A, Rosen BC, Agnor R, Gao L, Labriola C, Axthelm M, Smedley J, Julander JG, Bonaldo MC, Walker LM, Messaoudi I, Slifka MK, Burton DR, Kallas EG, Sacha JB, Watkins DI, Burwitz BJ. Therapeutic neutralizing monoclonal antibody administration protects against lethal yellow fever virus infection. Sci Transl Med 2023; 15:eade5795. [PMID: 36989376 PMCID: PMC10617428 DOI: 10.1126/scitranslmed.ade5795] [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: 08/25/2022] [Accepted: 03/10/2023] [Indexed: 03/31/2023]
Abstract
Yellow fever virus (YFV) is a reemerging global health threat, driven by several factors, including increased spread of the mosquito vector and rapid urbanization. Although a prophylactic vaccine exists, vaccine hesitancy, supply deficits, and distribution difficulties leave specific populations at risk of severe YFV disease, as evidenced by recent outbreaks in South America. To establish a treatment for patients with severe YFV infection, we tested 37 YFV-specific monoclonal antibodies isolated from vaccinated humans and identified two capable of potently neutralizing multiple pathogenic primary YFV isolates. Using both hamster and nonhuman primate models of lethal YFV infection, we demonstrate that a single administration of either of these two potently neutralizing antibodies during acute infection fully controlled viremia and prevented severe disease and death in treated animals. Given the potential severity of YFV-induced disease, our results show that these antibodies could be effective in saving lives and fill a much-needed void in managing YFV cases during outbreaks.
Collapse
Affiliation(s)
- Michael J. Ricciardi
- Mabloc LLC, 725 21st St. NW, Suite 301, Washington, DC 20052, USA
- George Washington University, 2121 I St. NW, Washington, DC 20052, USA
| | - Lauren N. Rust
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Nuria Pedreño-Lopez
- George Washington University, 2121 I St. NW, Washington, DC 20052, USA
- IrsiCaixa AIDS Research Institute, Ctra. del Canyet SN, Badalona 08916, Barcelona, Spain
| | - Sofiya Yusova
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Sreya Biswas
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Gabriela M. Webb
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
| | | | - Thomas B. Voigt
- Mabloc LLC, 725 21st St. NW, Suite 301, Washington, DC 20052, USA
- George Washington University, 2121 I St. NW, Washington, DC 20052, USA
| | - Johan J. Louw
- George Washington University, 2121 I St. NW, Washington, DC 20052, USA
| | | | - John R. DiBello
- Mabloc LLC, 725 21st St. NW, Suite 301, Washington, DC 20052, USA
| | - Hans-Peter Raué
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Aaron M. Barber-Axthelm
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Kimberly Chun
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Samantha Uttke
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Lidiane M. S. Raphael
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Brandon C. Rosen
- Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Rebecca Agnor
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Lina Gao
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Caralyn Labriola
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Michael Axthelm
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Justin G. Julander
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Myrna C. Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | | | - Ilhem Messaoudi
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Mark K. Slifka
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - Dennis R. Burton
- Mabloc LLC, 725 21st St. NW, Suite 301, Washington, DC 20052, USA
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Esper G. Kallas
- Mabloc LLC, 725 21st St. NW, Suite 301, Washington, DC 20052, USA
- Department of Infectious and Parasitic Diseases, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Jonah B. Sacha
- Mabloc LLC, 725 21st St. NW, Suite 301, Washington, DC 20052, USA
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| | - David I. Watkins
- Mabloc LLC, 725 21st St. NW, Suite 301, Washington, DC 20052, USA
- George Washington University, 2121 I St. NW, Washington, DC 20052, USA
| | - Benjamin J. Burwitz
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR 97006, USA
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, USA
| |
Collapse
|
27
|
Immunogenicity and protective activity of mRNA vaccine candidates against yellow fever virus in animal models. NPJ Vaccines 2023; 8:31. [PMID: 36871059 PMCID: PMC9984760 DOI: 10.1038/s41541-023-00629-7] [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: 07/15/2022] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Despite the success of the widely used attenuated yellow fever (YF) vaccine, its global supply remains a substantial barrier to implementing vaccination campaigns in endemic regions and combating emerging epidemics. In A129 mice and rhesus macaques, we evaluated the immunogenicity and protective activity of messenger RNA (mRNA) vaccine candidates encapsulated in lipid nanoparticles, expressing the pre-membrane and envelope proteins or the non-structural protein 1 of YF virus. Vaccine constructs induced humoral and cell-mediated immune responses in mice, resulting in protection against lethal YF virus infection after passive administration of serum or splenocytes from vaccinated mice. Vaccination of macaques induced sustained high humoral and cellular immune responses for at least 5 months after the second dose. Our data demonstrate that these mRNA vaccine candidates can be considered an attractive addition to the licensed YF vaccine supply based on the induction of functional antibodies correlating with protection and T-cell responses; they could alleviate the limited supply of current YF vaccines, mitigating future YF epidemics.
Collapse
|
28
|
Saito K, Shimasaki K, Fukasawa M, Suzuki R, Okemoto-Nakamura Y, Katoh K, Takasaki T, Hanada K. Establishment of Vero cell lines persistently harboring a yellow fever virus 17D subgenomic replicon. Virus Res 2022; 322:198935. [PMID: 36152929 DOI: 10.1016/j.virusres.2022.198935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022]
Abstract
Yellow fever virus (YFV), a member of the genus Flavivirus, family Flaviviridae, is the etiological agent for an acute viral hemorrhagic disease, yellow fever. Although effective live attenuated vaccines based on the strain YFV 17D are currently available, no specific antiviral drug is available, and the disease remains a major public health concern. Hence, the discovery and development of antiviral drugs should lead to great benefits in controlling the disease. To provide a screening platform for antiviral agents targeting YFV RNA translation/replication, we have established and characterized two Vero cell lines that persistently harbor a subgenomic replicon derived from YFV 17D-204 (referred to as replicon cells). The replicon carries YFV nucleotides (1 - 176 and 2382-10,862) and a green fluorescent protein (GFP)-Zeocin resistance fusion gene as a selection marker and indicator of persistent replication. Immunofluorescence analysis revealed that both replicon cells and YFV 17D-infected cells showed similar distribution patterns of viral NS4B protein and replication intermediate, double-stranded RNA. Sequencing analysis of persistent replicons from the two replicon cell lines suggested that their nucleotide sequences did not vary greatly following multiple passages. We examined the effect of five agents, the antiviral cytokines interferon-β and -γ, the nucleoside analog ribavirin, the squalene synthase inhibitor zaragozic acid A, and the antibiotic rifapentine, a recently reported entry and replication inhibitor against YFV, on the persistent replication in the two replicon cell lines. These agents were selected because they inhibited both production of YFV 17D and transient replication of a luciferase-expressing replicon in Vero cells, without greatly affecting cell viability. We found that each of the agents decreased GFP fluorescence in the replicon cells, albeit to varying degrees. The agents other than rifapentine also showed a decrease in viral RNA levels in the replicon cells comparable to that seen for GFP fluorescence. These results indicate that persistent replication is susceptible to each of these five agents, although their mechanisms of action may differ. Taken together, these results provide evidence that translation/replication of the replicon in the replicon cells mimics that of the viral genome upon YFV 17D infection, indicating that the replicon cell lines can serve as a useful tool for high-throughput antiviral drug screening.
Collapse
Affiliation(s)
- Kyoko Saito
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan.
| | - Kentaro Shimasaki
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Masayoshi Fukasawa
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, Musashimurayama-shi, Tokyo, Japan
| | - Yuko Okemoto-Nakamura
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Kaoru Katoh
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba-shi, Ibaragi, Japan; AIRC, National Institute of Advanced Industrial Science and Technology (AIST), Koto-ku, Tokyo, Japan
| | - Tomohiko Takasaki
- Kanagawa Prefectural Institute of Public Health, Chigasaki-shi, Kanagawa, Japan
| | - Kentaro Hanada
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan; Department of Quality Assurance, Radiation Safety, and Information System, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
29
|
Friedman-Klabanoff DJ, Birkhold M, Short MT, Wilson TR, Meneses CR, Lacsina JR, Oliveira F, Kamhawi S, Valenzuela JG, Hunsberger S, Mateja A, Stoloff G, Pleguezuelos O, Memoli MJ, Laurens MB. Safety and immunogenicity of AGS-v PLUS, a mosquito saliva peptide vaccine against arboviral diseases: A randomized, double-blind, placebo-controlled Phase 1 trial. EBioMedicine 2022; 86:104375. [PMID: 36436281 PMCID: PMC9700263 DOI: 10.1016/j.ebiom.2022.104375] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Immunity to mosquito salivary proteins could provide protection against multiple mosquito-borne diseases and significantly impact public health. We evaluated the safety and immunogenicity of AGS-v PLUS, a mosquito salivary peptide vaccine, in healthy adults 18-50 years old. METHODS We conducted a randomized, double-blind, placebo-controlled Phase 1 study of AGS-v PLUS administered subcutaneously on Days 1 and 22 at the Center for Vaccine Development and Global Health, Baltimore, MD, USA. Participants were block randomized 1:1:1:1:1 to two doses saline placebo, two doses AGS-v PLUS, AGS-v PLUS/ISA-51 and saline placebo, two doses AGS-v PLUS/ISA-51, or two doses AGS-v PLUS/Alhydrogel. Primary endpoints were safety (all participants receiving ≥1 injection) and antibody and cytokine responses (all participants with day 43 samples), analysed by intention to treat. FINDINGS Between 26 August 2019 and 25 February 2020, 51 participants were enrolled and randomized, 11 into the single dose AGS-v PLUS/ISA-51 group and ten in other groups. Due to COVID-19, 15 participants did not return for day 43 samplings. Participants experienced no treatment-emergent or serious adverse events. All solicited symptoms in 2/10 placebo recipients and 22/41 AGS-v PLUS recipients after dose one and 1/10 placebo recipients and 22/41 AGS-v PLUS recipients after dose two were mild/moderate except for one severe fever the day after vaccination (placebo group). Only injection site pain was more common in vaccine groups (15/51 after dose 1 and 11/51 after dose 2) versus placebo. Compared to placebo, all vaccine groups had significantly greater fold change in anti-AGS-v PLUS IgG and IFN-ɣ from baseline. INTERPRETATION AGS-v PLUS had favourable safety profile and induced robust immune responses. Next steps will determine if findings translate into clinical efficacy against mosquito-borne diseases. FUNDING UK Department of Health and Social Care.
Collapse
Affiliation(s)
- DeAnna J Friedman-Klabanoff
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Megan Birkhold
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mara T Short
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Timothy R Wilson
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Claudio R Meneses
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Joshua R Lacsina
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Fabiano Oliveira
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Shaden Kamhawi
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Jesus G Valenzuela
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Sally Hunsberger
- Biostatistics Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Allyson Mateja
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | - Matthew J Memoli
- Clinical Studies Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Matthew B Laurens
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
30
|
Lindsey NP, Horton J, Barrett ADT, Demanou M, Monath TP, Tomori O, Van Herp M, Zeller H, Fall IS, Cibrelus L, Erin Staples J. Yellow fever resurgence: An avoidable crisis? NPJ Vaccines 2022; 7:137. [PMID: 36323723 PMCID: PMC9629880 DOI: 10.1038/s41541-022-00552-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/10/2022] [Indexed: 11/26/2022] Open
Affiliation(s)
- Nicole P. Lindsey
- Eliminate Yellow Fever Epidemic Risk Assessment Working Group, Geneva, Switzerland ,grid.416738.f0000 0001 2163 0069Arboviral Diseases Branch, U.S. Centers for Disease Control and Prevention, Fort Collins, CO USA
| | - Jennifer Horton
- Eliminate Yellow Fever Epidemic Risk Assessment Working Group, Geneva, Switzerland ,grid.3575.40000000121633745Eliminate Yellow Fever Epidemics Strategy, High Impact Epidemics, WHO Health Emergencies Programme, World Health Organization, Geneva, Switzerland
| | - Alan D. T. Barrett
- Eliminate Yellow Fever Epidemic Risk Assessment Working Group, Geneva, Switzerland ,grid.176731.50000 0001 1547 9964Department of Pathology and Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX USA
| | - Maurice Demanou
- Eliminate Yellow Fever Epidemic Risk Assessment Working Group, Geneva, Switzerland ,Regional Office for Africa, World Health Organization, Ouagadougou, Burkina Faso
| | - Thomas P. Monath
- Eliminate Yellow Fever Epidemic Risk Assessment Working Group, Geneva, Switzerland ,grid.511631.1Crozet BioPharma, Lexington, MA USA
| | - Oyewale Tomori
- Eliminate Yellow Fever Epidemic Risk Assessment Working Group, Geneva, Switzerland ,grid.442553.10000 0004 0622 6369African Center for Genomics of Infectious Diseases (ACEGID), Redeemer’s University, Ede, Nigeria
| | - Michel Van Herp
- Eliminate Yellow Fever Epidemic Risk Assessment Working Group, Geneva, Switzerland ,grid.452593.cMédecins Sans Frontières, Brussels, Belgium
| | - Herve Zeller
- Eliminate Yellow Fever Epidemic Risk Assessment Working Group, Geneva, Switzerland ,Consultant, Paris, France
| | - Ibrahima Soce Fall
- grid.3575.40000000121633745WHO Health Emergencies Programme, World Health Organization Headquarters, Geneva, Switzerland
| | - Laurence Cibrelus
- Eliminate Yellow Fever Epidemic Risk Assessment Working Group, Geneva, Switzerland ,grid.3575.40000000121633745Eliminate Yellow Fever Epidemics Strategy, High Impact Epidemics, WHO Health Emergencies Programme, World Health Organization, Geneva, Switzerland
| | - J. Erin Staples
- Eliminate Yellow Fever Epidemic Risk Assessment Working Group, Geneva, Switzerland ,grid.416738.f0000 0001 2163 0069Arboviral Diseases Branch, U.S. Centers for Disease Control and Prevention, Fort Collins, CO USA
| |
Collapse
|
31
|
Corliss L, Holliday M, Lennemann NJ. Dual-fluorescent reporter for live-cell imaging of the ER during DENV infection. Front Cell Infect Microbiol 2022; 12:1042735. [PMID: 36389173 PMCID: PMC9640912 DOI: 10.3389/fcimb.2022.1042735] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/11/2022] [Indexed: 08/25/2023] Open
Abstract
Infection by flaviviruses leads to dramatic remodeling of the endoplasmic reticulum (ER). Viral replication occurs within virus-induced vesicular invaginations in the ER membrane. A hallmark of flavivirus infection is expansion of the ER membrane which can be observed at specific time points post infection. However, this process has not been effectively visualized in living cells throughout the course of infection at the single cell resolution. In this study, we developed a plasmid-based reporter system to monitor flavivirus infection and simultaneous virus-induced manipulation of single cells throughout the course of infection in real-time. This system requires viral protease cleavage to release an ER-anchored fluorescent protein infection reporter that is fused to a nuclear localization signal (NLS). This proteolytic cleavage allows for the translocation of the infection reporter signal to the nucleus while an ER-specific fluorescent marker remains localized in the lumen. Thus, the construct allows for the visualization of virus-dependent changes to the ER throughout the course of infection. In this study, we show that our reporter was efficiently cleaved upon the expression of multiple flavivirus proteases, including dengue virus (DENV), Zika virus (ZIKV), and yellow fever virus (YFV). We also found that the DENV protease-dependent cleavage of our ER-anchored reporter exhibited more stringent cleavage sequence specificity than what has previously been shown with biochemical assays. Using this system for long term time-lapse imaging of living cells infected with DENV, we observed nuclear translocation of the reporter signal beginning approximately 8 hours post-infection, which continued to increase throughout the time course. Interestingly, we found that increased reporter signal translocation correlated with increased ER signal intensity, suggesting a positive association between DENV infection and ER expansion in a time-dependent manner. Overall, this report demonstrates that the FlavER platform provides a useful tool for monitoring flavivirus infection and simultaneously observing virus-dependent changes to the host cell ER, allowing for study of the temporal nature of virus-host interactions.
Collapse
Affiliation(s)
| | | | - Nicholas J. Lennemann
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
32
|
Ateutchia Ngouanet S, Wanji S, Yadouleton A, Demanou M, Djouaka R, Nanfack-Minkeu F. Factors enhancing the transmission of mosquito-borne arboviruses in Africa. Virusdisease 2022; 33:477-488. [PMID: 36278029 PMCID: PMC9579656 DOI: 10.1007/s13337-022-00795-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/19/2022] [Indexed: 11/28/2022] Open
Affiliation(s)
- Sandra Ateutchia Ngouanet
- International Institute of Tropical Agriculture (IITA), 08 Tri-Postal, P.O. Box 0932, Cotonou, Benin
- Department Microbiology and Parasitology, Faculty of Science, University of Buea, P.O. BOX 63, Buea, Cameroon
| | - Samuel Wanji
- Department Microbiology and Parasitology, Faculty of Science, University of Buea, P.O. BOX 63, Buea, Cameroon
| | - Anges Yadouleton
- Centre de Recherche Entomologique de Cotonou (CREC), Cotonou, Benin
| | - Maurice Demanou
- Regional Yellow Fever Laboratory Coordinator World Health Organization, Inter-Country Support Team West Africa, 03 P.O. Box 7019, Ouagadougou 03, Burkina Faso
| | - Rousseau Djouaka
- International Institute of Tropical Agriculture (IITA), 08 Tri-Postal, P.O. Box 0932, Cotonou, Benin
| | - Ferdinand Nanfack-Minkeu
- International Institute of Tropical Agriculture (IITA), 08 Tri-Postal, P.O. Box 0932, Cotonou, Benin
- Department of Biology, The College of Wooster, Wooster, OH USA
| |
Collapse
|
33
|
Continuous Circulation of Yellow Fever among Rural Populations in the Central African Republic. Viruses 2022; 14:v14092014. [PMID: 36146820 PMCID: PMC9503741 DOI: 10.3390/v14092014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Yellow fever remains a public-health threat in remote regions of Africa. Here, we report the identification and genetic characterisation of one yellow-fever case observed during the investigation of a cluster of nine suspected haemorrhagic fever cases in a village in the Central African Republic. Samples were tested using real-time RT-PCR targeting the main African haemorrhagic fever viruses. Following negative results, we attempted virus isolation on VERO E6 cells and new-born mice and rescreened the samples using rRT-PCR. The whole viral genome was sequenced using an Illumina NovaSeq 6000 sequencer. Yellow-fever virus (YFV) was isolated from one woman who reported farming activities in a forest setting several days before disease onset. Phylogenetic analysis shows that this strain belongs to the East–Central African YFV genotype, with an estimated emergence some 63 years ago. Finally, five unique amino-acid changes are present in the capsid, envelop, NS1A, NS3, and NS4B proteins. More efforts are required to control yellow-fever re-emergence in resource-limited settings.
Collapse
|
34
|
Lee WL, Gu X, Armas F, Leifels M, Wu F, Chandra F, Chua FJD, Syenina A, Chen H, Cheng D, Ooi EE, Wuertz S, Alm EJ, Thompson J. Monitoring human arboviral diseases through wastewater surveillance: Challenges, progress and future opportunities. WATER RESEARCH 2022; 223:118904. [PMID: 36007397 DOI: 10.1016/j.watres.2022.118904] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 05/21/2023]
Abstract
Arboviral diseases are caused by a group of viruses spread by the bite of infected arthropods. Amongst these, dengue, Zika, west nile fever and yellow fever cause the greatest economic and social impact. Arboviral epidemics have increased in frequency, magnitude and geographical extent over the past decades and are expected to continue increasing with climate change and expanding urbanisation. Arboviral prevalence is largely underestimated, as most infections are asymptomatic, nevertheless existing surveillance systems are based on passive reporting of loosely defined clinical syndromes with infrequent laboratory confirmation. Wastewater-based surveillance (WBS), which has been demonstrated to be useful for monitoring diseases with significant asymptomatic populations including COVID19 and polio, could be a useful complement to arboviral surveillance. We review the current state of knowledge and identify key factors that affect the feasibility of monitoring arboviral diseases by WBS to include viral shedding loads by infected persons, the persistence of shed arboviruses and the efficiency of their recovery from sewage. We provide a simple model on the volume of wastewater that needs to be processed for detection of arboviruses, in face of lower arboviral shedding rates. In all, this review serves to reflect on the key challenges that need to be addressed and overcome for successful implementation of arboviral WBS.
Collapse
Affiliation(s)
- Wei Lin Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Xiaoqiong Gu
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Federica Armas
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Mats Leifels
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551, Singapore
| | - Fuqing Wu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, Center for Infectious Disease, University of Texas School of Public Health, Houston, TX, USA
| | - Franciscus Chandra
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Feng Jun Desmond Chua
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551, Singapore
| | - Ayesa Syenina
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore; Viral Research and Experimental Medicine Centre (ViREMiCS), SingHealth Duke-NUS Academic Medical Centre, Singapore 169856, Singapore
| | - Hongjie Chen
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Dan Cheng
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551, Singapore
| | - Eng Eong Ooi
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore; Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore; Viral Research and Experimental Medicine Centre (ViREMiCS), SingHealth Duke-NUS Academic Medical Centre, Singapore 169856, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| | - Stefan Wuertz
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551, Singapore; School of Civil and Environmental Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Eric J Alm
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore; Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore; Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Janelle Thompson
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore; Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore 637551, Singapore; Asian School of the Environment, Nanyang Technological University, Singapore 637459, Singapore.
| |
Collapse
|
35
|
Gabiane G, Yen P, Failloux A. Aedes mosquitoes in the emerging threat of urban yellow fever transmission. Rev Med Virol 2022; 32:e2333. [PMID: 35124859 PMCID: PMC9541788 DOI: 10.1002/rmv.2333] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/05/2022] [Accepted: 01/18/2022] [Indexed: 01/04/2023]
Abstract
This last decade has seen a resurgence of yellow fever (YF) in historical endemic regions and repeated attempts of YF introduction in YF-free countries such as the Asia-Pacific region and the Caribbean. Infected travellers are the main entry routes in these regions where competent mosquito vectors proliferate in appropriate environmental conditions. With the discovery of the 17D vaccine, it was thought that YF would be eradicated. Unfortunately, it was not the case and, contrary to dengue, chikungunya and Zika, factors that cotribute to YF transmission remain under investigation. Today, all the signals are red and it is very likely that YF will be the next pandemic in the YF-free regions where millions of people are immunologically naïve. Unlike COVID-19, YF is associated with a high case-fatality rate and a high number of deaths are expected. This review gives an overview of global YF situation, including the non-endemic Asia-Pacific region and the Caribbean where Aedes aegypti is abundantly distributed, and also proposes different hypotheses on why YF outbreaks have not yet occurred despite high records of travellers importing YF into these regions and what role Aedes mosquitoes play in the emergence of urban YF.
Collapse
Affiliation(s)
- Gaelle Gabiane
- Institut PasteurUniversité de Paris, Unit of Arboviruses and Insect VectorsParisFrance
- Université des Antilles, Campus de SchoelcherSchoelcherMartinique
| | - Pei‐Shi Yen
- Institut PasteurUniversité de Paris, Unit of Arboviruses and Insect VectorsParisFrance
| | - Anna‐Bella Failloux
- Institut PasteurUniversité de Paris, Unit of Arboviruses and Insect VectorsParisFrance
| |
Collapse
|
36
|
Doyle MP, Genualdi JR, Bailey AL, Kose N, Gainza C, Rodriguez J, Reeder KM, Nelson CA, Jethva PN, Sutton RE, Bombardi RG, Gross ML, Julander JG, Fremont DH, Diamond MS, Crowe JE. Isolation of a Potently Neutralizing and Protective Human Monoclonal Antibody Targeting Yellow Fever Virus. mBio 2022; 13:e0051222. [PMID: 35420472 PMCID: PMC9239089 DOI: 10.1128/mbio.00512-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/07/2022] [Indexed: 01/23/2023] Open
Abstract
Yellow fever virus (YFV) causes sporadic outbreaks of infection in South America and sub-Saharan Africa. While live-attenuated yellow fever virus vaccines based on three substrains of 17D are considered some of the most effective vaccines in use, problems with production and distribution have created large populations of unvaccinated, vulnerable individuals in areas of endemicity. To date, specific antiviral therapeutics have not been licensed for human use against YFV or any other related flavivirus. Recent advances in monoclonal antibody (mAb) technology have allowed the identification of numerous candidate therapeutics targeting highly pathogenic viruses, including many flaviviruses. Here, we sought to identify a highly neutralizing antibody targeting the YFV envelope (E) protein as a therapeutic candidate. We used human B cell hybridoma technology to isolate mAbs from circulating memory B cells from human YFV vaccine recipients. These antibodies bound to recombinant YFV E protein and recognized at least five major antigenic sites on E. Two mAbs (designated YFV-136 and YFV-121) recognized a shared antigenic site and neutralized the YFV-17D vaccine strain in vitro. YFV-136 also potently inhibited infection by multiple wild-type YFV strains, in part, at a postattachment step in the virus replication cycle. YFV-136 showed therapeutic protection in two animal models of YFV challenge, including hamsters and immunocompromised mice engrafted with human hepatocytes. These studies define features of the antigenic landscape of the YFV E protein recognized by the human B cell response and identify a therapeutic antibody candidate that inhibits infection and disease caused by highly virulent strains of YFV. IMPORTANCE Yellow fever virus (YFV) is a mosquito-borne virus that occasionally causes outbreaks of severe infection and disease in South America and sub-Saharan Africa. There are very effective live-attenuated (weakened) yellow fever virus vaccines, but recent problems with their production and distribution have left many people in affected areas vulnerable. Here, we sought to isolate an antibody targeting the surface of the virus for possible use in the future as a biologic drug to prevent or treat YFV infection. We isolated naturally occurring antibodies from individuals who had received a YFV vaccine. We created antibodies and tested them. We found that the antibody with the most powerful antiviral activity was a beneficial treatment in two different small-animal models of human infection. These studies identified features of the virus that are recognized by the human immune system and generated a therapeutic antibody candidate that inhibits infection caused by highly virulent strains of YFV.
Collapse
Affiliation(s)
- Michael P. Doyle
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Joseph R. Genualdi
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Adam L. Bailey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nurgun Kose
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Christopher Gainza
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jessica Rodriguez
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kristen M. Reeder
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Christopher A. Nelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Prashant N. Jethva
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Rachel E. Sutton
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Robin G. Bombardi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael L. Gross
- Department of Chemistry, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Justin G. Julander
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, USA
| | - Daved H. Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael S. Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - James E. Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
37
|
Silva TMRD, Nogueira de Sá ACMG, Prates EJS, Rodrigues DE, Silva TPRD, Matozinhos FP, Vieira EWR. Yellow fever vaccination before and during the covid-19 pandemic in Brazil. Rev Saude Publica 2022; 56:45. [PMID: 35703600 PMCID: PMC9165638 DOI: 10.11606/s1518-8787.2022056004503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To analyze the number of yellow fever vaccine doses administered before and during the covid-19 pandemic in Brazil. METHODS This is an ecological, time series study based on data from the National Immunization Program. Differences between the median number of yellow fever vaccine doses administered in Brazil and in its regions before (from April/2019 to March/2020) and after (from April/2020 to March/2021) the implementation of social distancing measures in the country were assessed via the Mann-Whitney test. Prais-Winsten regression models were used for time series analyses. RESULTS We found a reduction in the median number of yellow fever vaccine doses administered in Brazil and in its regions: North (-34.71%), Midwest (-21.72%), South (-63.50%), and Southeast (-34.42%) (p < 0.05). Series showed stationary behavior in Brazil and in its five regions during the covid-19 pandemic (p > 0.05). Brazilian states also showed stationary trends, except for two states which recorded an increasing trend in the number of administered yellow fever vaccine doses, namely: Alagoas State (before: β = 64, p = 0.081; after: β = 897, p = 0.039), which became a yellow fever vaccine recommendation zone, and Roraima State (before: β = 68, p = 0.724; after: β = 150, p = 0.000), which intensified yellow fever vaccinations due to a yellow fever case confirmation in a Venezuelan State in 2020. CONCLUSION The reduced number of yellow fever vaccine doses administered during the covid-19 pandemic in Brazil may favor the reemergence of urban yellow fever cases in the country.
Collapse
Affiliation(s)
- Tércia Moreira Ribeiro da Silva
- Universidade Federal de Minas Gerais. Escola de Enfermagem. Departamento de Enfermagem Materno-Infantil e Saúde Pública. Belo Horizonte, MG, Brasil
| | | | - Elton Junio Sady Prates
- Universidade Federal de Minas Gerais. Escola de Enfermagem. Departamento de Enfermagem Materno-Infantil e Saúde Pública. Belo Horizonte, MG, Brasil
| | | | - Thales Philipe Rodrigues da Silva
- Universidade Federal de Minas Gerais. Escola de Enfermagem. Departamento de Enfermagem Materno-Infantil e Saúde Pública. Belo Horizonte, MG, Brasil
| | - Fernanda Penido Matozinhos
- Universidade Federal de Minas Gerais. Escola de Enfermagem. Departamento de Enfermagem Materno-Infantil e Saúde Pública. Belo Horizonte, MG, Brasil
| | - Ed Wilson Rodrigues Vieira
- Universidade Federal de Minas Gerais. Escola de Enfermagem. Departamento de Enfermagem Materno-Infantil e Saúde Pública. Belo Horizonte, MG, Brasil
| |
Collapse
|
38
|
Martin J, Barrett ADT, Lei D, Minor P. WHO working group meeting to amend WHO Recommendations to assure the quality, safety and efficacy of live attenuated yellow fever vaccines. Vaccine 2022; 40:3490-3494. [PMID: 35550848 PMCID: PMC9176314 DOI: 10.1016/j.vaccine.2022.04.095] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/03/2022] [Accepted: 04/28/2022] [Indexed: 01/13/2023]
Abstract
The current WHO Recommendations to assure the quality, safety and efficacy of live attenuated yellow fever vaccines were adopted in 2010. This document recommends that vaccine virus master and working seed lots should be tested for viscerotropism, immunogenicity and neurovirulence in monkeys. A vaccine manufacturer has reported, recently, discrepancies on the clinical scoring of monkeys during assessment of working seed lots and suggested aligning neurotropism assessment of yellow fever vaccines virus seed lots with that of neurovirulence testing of polio vaccines virus seed lots. In this approach, clinical signs are recorded but do not form part of the assessment or pass/fail criteria. At its 71st meeting in August 2020, the ECBS agreed to establish a drafting group and to consult with manufacturers and other stakeholders on the proposed amendment. Then a survey had been conducted to seek opinions of stakeholders on the neurotropism testing and revision of current WHO Recommendations for yellow fever vaccines. It was recognized from the answers of the survey that the test for neurovirulence in monkeys presents several technical challenges which could be addressed in the amended version of the Recommendations. On 18-19 March 2021, a virtual WHO working group meeting was held to discuss a proposed draft of the amended text with participants of yellow fever vaccine manufacturers and relevant regulators. Overall, there was a consensus among manufacturers and regulators that clinical evaluation provides important information and should be retained as part of the neurotropism test. However, there was also agreement that the test is somewhat subjective, and that analysis can be difficult. It was recognized that there was potential for improvement in both test execution and analysis to increase harmonization between manufacturers. Alternative tests to the non-human primates neurovirulence test would be useful but it was agreed that none seem to be sufficiently developed at present. Based on these working group discussions, it was proposed that the appendix on neurotropism test to be further revised by the WHO drafting group and submitted to ECBS for review and adoption. Issues other than neurotropism test were discussed in the meeting as well. There were a number of points identified during the meeting, such as new platform of production, animal models, deep sequencing, international standards, that are outside the current recommendations that are worthy of further discussion. Therefore, it is recommended that there would be a future meeting with various stakeholders to discuss the potential revision of the whole Recommendations for yellow fever vaccines in order to meet the current needs.
Collapse
Affiliation(s)
- Javier Martin
- National Institute for Biological Standards and Control, London, United Kingdom
| | | | | | | |
Collapse
|
39
|
Aliaga-Samanez A, Real R, Segura M, Marfil-Daza C, Olivero J. Yellow fever surveillance suggests zoonotic and anthroponotic emergent potential. Commun Biol 2022; 5:530. [PMID: 35654842 PMCID: PMC9163115 DOI: 10.1038/s42003-022-03492-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 05/11/2022] [Indexed: 11/09/2022] Open
Abstract
Yellow fever is transmitted by mosquitoes among human and non-human primates. In the last decades, infections are occurring in areas that had been free from yellow fever for decades, probably as a consequence of the rapid spread of mosquito vectors, and of the virus evolutionary dynamic in which non-human primates are involved. This research is a pathogeographic assessment of where enzootic cycles, based on primate assemblages, could be amplifying the risk of yellow fever infections, in the context of spatial changes shown by the disease since the late 20th century. In South America, the most relevant spread of disease cases affects parts of the Amazon basin and a wide area of southern Brazil, where forest fragmentation could be activating enzootic cycles next to urban areas. In Africa, yellow fever transmission is apparently spreading from the west of the continent, and primates could be contributing to this in savannas around rainforests. Our results are useful for identifying new areas that should be prioritised for vaccination, and suggest the need of deep yellow fever surveillance in primates of South America and Africa.
Collapse
Affiliation(s)
- Alisa Aliaga-Samanez
- Grupo de Biogeografía, Diversidad y Conservación, Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, 29071, Malaga, Spain.
| | - Raimundo Real
- Grupo de Biogeografía, Diversidad y Conservación, Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, 29071, Malaga, Spain
- Instituto IBYDA, Centro de Experimentación Grice-Hutchinson, Malaga, Spain
| | - Marina Segura
- Centro de Vacunación Internacional, Ministerio de Sanidad, Consumo y Bienestar Social, Estación Marítima, Recinto del Puerto, Muelle 3, 29001, Malaga, Spain
| | - Carlos Marfil-Daza
- Grupo de Biogeografía, Diversidad y Conservación, Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, 29071, Malaga, Spain
| | - Jesús Olivero
- Grupo de Biogeografía, Diversidad y Conservación, Departamento de Biología Animal, Facultad de Ciencias, Universidad de Málaga, 29071, Malaga, Spain
- Instituto IBYDA, Centro de Experimentación Grice-Hutchinson, Malaga, Spain
| |
Collapse
|
40
|
Vector-Borne Viral Diseases as a Current Threat for Human and Animal Health—One Health Perspective. J Clin Med 2022; 11:jcm11113026. [PMID: 35683413 PMCID: PMC9181581 DOI: 10.3390/jcm11113026] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022] Open
Abstract
Over the last decades, an increase in the emergence or re-emergence of arthropod-borne viruses has been observed in many regions. Viruses such as dengue, yellow fever, or zika are a threat for millions of people on different continents. On the other hand, some arboviruses are still described as endemic, however, they could become more important in the near future. Additionally, there is a group of arboviruses that, although important for animal breeding, are not a direct threat for human health. Those include, e.g., Schmallenberg, bluetongue, or African swine fever viruses. This review focuses on arboviruses and their major vectors: mosquitoes, ticks, biting midges, and sandflies. We discuss the current knowledge on arbovirus transmission, ecology, and methods of prevention. As arboviruses are a challenge to both human and animal health, successful prevention and control are therefore only possible through a One Health perspective.
Collapse
|
41
|
Monoclonal Antibodies and Flaviviruses: a Possible Option? mBio 2022; 13:e0082422. [PMID: 35575500 PMCID: PMC9239274 DOI: 10.1128/mbio.00824-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
M. P. Doyle, J. R. Genualdi, A. L. Bailey, N. Kose, et al. (mBio 13:e00512-22, 2022, https://doi.org/10.1128/mBio.00512-22), report on the cloning of a panel of fully human monoclonal antibodies (mAbs) directed against yellow fever virus (YFV). In particular, mAb YFV-136 is endowed with interesting cross-YFV substrain-neutralizing features. The importance of YFV-136 and other mAbs with similar characteristics is related not necessarily only to their possible future use in the clinic but also to their role in a better understanding of the biology of YFV (as well as of other flaviviruses) for the development of effective therapeutic and prophylactic strategies. The emergence and reemergence of different flaviviruses worldwide in the last decades certainly make this a compelling clinical priority.
Collapse
|
42
|
Kampango A, Hocke EF, Hansson H, Furu P, Haji KA, David JP, Konradsen F, Saleh F, Weldon CW, Schiøler KL, Alifrangis M. High DDT resistance without apparent association to kdr and Glutathione-S-transferase (GST) gene mutations in Aedes aegypti population at hotel compounds in Zanzibar. PLoS Negl Trop Dis 2022; 16:e0010355. [PMID: 35576233 PMCID: PMC9109918 DOI: 10.1371/journal.pntd.0010355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/25/2022] [Indexed: 11/18/2022] Open
Abstract
Global efforts to control Aedes mosquito-transmitted pathogens still rely heavily on insecticides. However, available information on vector resistance is mainly restricted to mosquito populations located in residential and public areas, whereas commercial settings, such as hotels are overlooked. This may obscure the real magnitude of the insecticide resistance problem and lead to ineffective vector control and resistance management. We investigated the profile of insecticide susceptibility of Aedes aegypti mosquitoes occurring at selected hotel compounds on Zanzibar Island. At least 100 adults Ae. aegypti females from larvae collected at four hotel compounds were exposed to papers impregnated with discriminant concentrations of DDT (4%), permethrin (0.75%), 0.05 deltamethrin (0.05%), propoxur (0.1%) and bendiocarb (0.1%) to determine their susceptibility profile. Allele-specific qPCR and sequencing analysis were applied to determine the possible association between observed resistance and presence of single nucleotide polymorphisms (SNPs) in the voltage-gated sodium channel gene (VGSC) linked to DDT/pyrethroid cross-resistance. Additionally, we explored the possible involvement of Glutathione-S-Transferase gene (GSTe2) mutations for the observed resistance profile. In vivo resistance bioassay indicated that Ae. aegypti at studied sites were highly resistant to DDT, mortality rate ranged from 26.3% to 55.3% and, moderately resistant to deltamethrin with a mortality rate between 79% to and 100%. However, genotyping of kdr mutations affecting the voltage-gated sodium channel only showed a low frequency of the V1016G mutation (n = 5; 0.97%). Moreover, for GSTe2, seven non-synonymous SNPs were detected (L111S, C115F, P117S, E132A, I150V, E178A and A198E) across two distinct haplotypes, but none of these were significantly associated with the observed resistance to DDT. Our findings suggest that cross-resistance to DDT/deltamethrin at hotel compounds in Zanzibar is not primarily mediated by mutations in VGSC. Moreover, the role of identified GSTe2 mutations in the resistance against DDT remains inconclusive. We encourage further studies to investigate the role of other potential insecticide resistance markers. Available information on mosquito resistance to insecticides is mainly restricted to residential and public areas, whereas commercial settings, such as hotels are overlooked. This may hide the real size of an insecticide resistance problem and lead to ineffective mosquito control. We investigated insecticide susceptibility of Aedes aegypti mosquitoes occurring at selected hotel compounds on Zanzibar Island. We also looked at whether resistance occurred in mosquitoes with gene mutations for two proteins (voltage-gated sodium channels and glutathione-S-transferase) that are known to cause resistance to insecticides in other parts of the world. The Ae. aegypti mosquitoes collected from hotels were highly resistant to DDT, and moderately and possibly resistant to deltamethrin and propoxur, respectively. However, resistance to these insecticides was not linked to mutations in either of the studied genes. The presence of insecticide resistance in Ae. aegypti in hotel compounds on Zanzibar is concerning and shows that these areas can act as sources of resistant mosquitoes. More needs to be done to establish the underlying causes for insecticide resistance in hotel Ae. aegypti populations, and this information can then be used to design measures that prevent resistance from becoming more widespread on Zanzibar.
Collapse
Affiliation(s)
- Ayubo Kampango
- Sector de Estudos de Vectores, Instituto Nacional de Saúde (INS), Vila de Marracuene, Província de Maputo, Mozambique
- Department of Zoology and Entomology, University of Pretoria (UP), Hatfield, South Africa
- * E-mail:
| | - Emma F. Hocke
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Helle Hansson
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Peter Furu
- Global Health Section, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Khamis A. Haji
- Zanzibar Malaria Elimination Programme (ZAMEP), Unguja Island, Zanzibar, Tanzania
| | - Jean-Philippe David
- Laboratoire d’Ecologie Alpine (LECA), UMR 5553, Centre National de la Recherche Scientifique (CNRS)—Université Grenoble-Alpes, Grenoble, France
| | - Flemming Konradsen
- Global Health Section, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Fatma Saleh
- Department of Allied Health Sciences, School of Health and Medical Sciences, The State University of Zanzibar, Unguja Island, Zanzibar, Tanzania
| | - Christopher W. Weldon
- Department of Zoology and Entomology, University of Pretoria (UP), Hatfield, South Africa
| | - Karin L. Schiøler
- Global Health Section, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Michael Alifrangis
- Center for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| |
Collapse
|
43
|
Nemg FBS, Abanda NN, Yonga MG, Ouapi D, Samme IE, Djoumetio MD, Endegue-Zanga MC, Demanou M, Njouom R. Sustained circulation of yellow fever virus in Cameroon: an analysis of laboratory surveillance data, 2010-2020. BMC Infect Dis 2022; 22:418. [PMID: 35488234 PMCID: PMC9055699 DOI: 10.1186/s12879-022-07407-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 04/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The re-emergence of yellow fever poses a serious public health risk to unimmunized communities in the tropical regions of Africa and South America and unvaccinated travelers visiting these regions. This risk is further accentuated by the likely spread of the virus to areas with potential for yellow fever transmission such as in Asia, Europe, and North America. To mitigate this risk, surveillance of yellow fever is pivotal. We performed an analysis of laboratory-based surveillance of yellow fever suspected cases in Cameroon during 2010-2020 to characterize the epidemiology of yellow fever cases and define health districts at high risk. METHOD We reviewed IgM capture ELISA and plaque reduction neutralization test (PRNT) test results of all suspected yellow fever patients analyzed at Centre Pasteur of Cameroon, the national yellow fever testing laboratory, during 2010-2020. RESULTS Of the 20,261 yellow fever suspected patient's samples that were tested, yellow fever IgM antibodies were detected in 360 patients representing an annual average of 33 cases/year. A major increase in YF IgM positive cases was observed in 2015 and in 2016 followed by a decrease in cases to below pre-2015 levels. The majority of the 2015 cases occurred during the latter part of the year while those in 2016, occurred between February and May. This trend may be due to an increase in transmission that began in late 2015 and continued to early 2016 or due to two separate transmission events. In 2016, where the highest number of cases were detected, 60 health districts in the 10 regions of Cameroon were affected with the Littoral, Northwest and, Far North regions being the most affected. After 2016, the number of detected yellow fever IgM positive cases dropped. CONCLUSION Our study shows that yellow fever transmission continues to persist and seems to be occurring all over Cameroon with all 10 regions under surveillance reporting a case. Preventive measures such as mass vaccination campaigns and routine childhood immunizations are urgently needed to increase population immunity. The diagnostic limitations in our analysis highlight the need to strengthen laboratory capacity and improve case investigations.
Collapse
Affiliation(s)
| | - Ngu Njei Abanda
- Virology Unit, Virology Service, Centre Pasteur of Cameroon, 451 Rue 2005, BP 1274, Yaounde, Cameroon.
| | - Martial Gide Yonga
- Virology Unit, Virology Service, Centre Pasteur of Cameroon, 451 Rue 2005, BP 1274, Yaounde, Cameroon
| | - Diane Ouapi
- Virology Unit, Virology Service, Centre Pasteur of Cameroon, 451 Rue 2005, BP 1274, Yaounde, Cameroon
| | - Ivis Ewang Samme
- Expanded Programme on Immunization, Ministry of Public Health, Yaoundé, Cameroon
| | | | | | - Maurice Demanou
- World Health Organization, Inter-Country Support Team West Africa, 03 BP 7019, Ouagadougou, Burkina Faso
| | - Richard Njouom
- Virology Unit, Virology Service, Centre Pasteur of Cameroon, 451 Rue 2005, BP 1274, Yaounde, Cameroon
| |
Collapse
|
44
|
Exploring relationships between drought and epidemic cholera in Africa using generalised linear models. BMC Infect Dis 2021; 21:1177. [PMID: 34809609 PMCID: PMC8609751 DOI: 10.1186/s12879-021-06856-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 11/08/2021] [Indexed: 11/18/2022] Open
Abstract
Background Temperature and precipitation are known to affect Vibrio cholerae outbreaks. Despite this, the impact of drought on outbreaks has been largely understudied. Africa is both drought and cholera prone and more research is needed in Africa to understand cholera dynamics in relation to drought. Methods Here, we analyse a range of environmental and socioeconomic covariates and fit generalised linear models to publicly available national data, to test for associations with several indices of drought and make cholera outbreak projections to 2070 under three scenarios of global change, reflecting varying trajectories of CO2 emissions, socio-economic development, and population growth. Results The best-fit model implies that drought is a significant risk factor for African cholera outbreaks, alongside positive effects of population, temperature and poverty and a negative effect of freshwater withdrawal. The projections show that following stringent emissions pathways and expanding sustainable development may reduce cholera outbreak occurrence in Africa, although these changes were spatially heterogeneous. Conclusions Despite an effect of drought in explaining recent cholera outbreaks, future projections highlighted the potential for sustainable development gains to offset drought-related impacts on cholera risk. Future work should build on this research investigating the impacts of drought on cholera on a finer spatial scale and potential non-linear relationships, especially in high-burden countries which saw little cholera change in the scenario analysis. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-021-06856-4.
Collapse
|
45
|
Wozniak DM, Riesle-Sbarbaro SA, Kirchoff N, Hansen-Kant K, Wahlbrink A, Stern A, Lander A, Hartmann K, Krasemann S, Kurth A, Prescott J. Inoculation route-dependent Lassa virus dissemination and shedding dynamics in the natural reservoir - Mastomys natalensis. Emerg Microbes Infect 2021; 10:2313-2325. [PMID: 34792436 PMCID: PMC8654411 DOI: 10.1080/22221751.2021.2008773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Lassa virus (LASV), a Risk Group-4 zoonotic haemorrhagic fever virus, affects sub-Saharan African countries. Lassa fever, caused by LASV, results in thousands of annual deaths. Although decades have elapsed since the identification of the Natal multimammate mouse (Mastomys natalensis) as a natural reservoir of LASV, little effort has been made to characterize LASV infection in its reservoir. The natural route of infection and transmission of LASV within M. natalensis remains unknown, and the clinical impact of LASV in M. natalensis is mostly undescribed. Herein, using an outbred colony of M. natalensis, we investigate the replication and dissemination dynamics of LASV in this reservoir following various inoculation routes. Inoculation with LASV, regardless of route, resulted in a systemic infection and accumulation of abundant LASV-RNA in many tissues. LASV infection in the Natal multimammate mice was subclinical, however, clinical chemistry values were transiently altered and immune infiltrates were observed histologically in lungs, spleens and livers, indicating a minor disease with coordinated immune responses are elicited, controlling infection. Intranasal infection resulted in unique virus tissue dissemination dynamics and heightened LASV shedding, compared to subcutaneous inoculation. Our study provides important insights into LASV infection in its natural reservoir using a contemporary infection system, demonstrating that specific inoculation routes result in disparate dissemination outcomes, suggesting intranasal inoculation is important in the maintenance of LASV in the natural reservoir, and emphasizes that selection of the appropriate inoculation route is necessary to examine aspects of viral replication, transmission and responses to zoonotic viruses in their natural reservoirs.
Collapse
Affiliation(s)
- D M Wozniak
- ZBS5-Biosafety Level-4 Laboratory, Robert Koch-Institute, Berlin, Germany
| | | | - N Kirchoff
- ZBS5-Biosafety Level-4 Laboratory, Robert Koch-Institute, Berlin, Germany
| | - K Hansen-Kant
- ZBS5-Biosafety Level-4 Laboratory, Robert Koch-Institute, Berlin, Germany
| | - A Wahlbrink
- ZBS5-Biosafety Level-4 Laboratory, Robert Koch-Institute, Berlin, Germany
| | - A Stern
- ZBS5-Biosafety Level-4 Laboratory, Robert Koch-Institute, Berlin, Germany
| | - A Lander
- ZBS5-Biosafety Level-4 Laboratory, Robert Koch-Institute, Berlin, Germany
| | - K Hartmann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - S Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - A Kurth
- ZBS5-Biosafety Level-4 Laboratory, Robert Koch-Institute, Berlin, Germany
| | - J Prescott
- ZBS5-Biosafety Level-4 Laboratory, Robert Koch-Institute, Berlin, Germany
| |
Collapse
|
46
|
Echeverria-Londono S, Li X, Toor J, de Villiers MJ, Nayagam S, Hallett TB, Abbas K, Jit M, Klepac P, Jean K, Garske T, Ferguson NM, Gaythorpe KAM. How can the public health impact of vaccination be estimated? BMC Public Health 2021; 21:2049. [PMID: 34753437 PMCID: PMC8577012 DOI: 10.1186/s12889-021-12040-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/19/2021] [Indexed: 11/25/2022] Open
Abstract
Background Deaths due to vaccine preventable diseases cause a notable proportion of mortality worldwide. To quantify the importance of vaccination, it is necessary to estimate the burden averted through vaccination. The Vaccine Impact Modelling Consortium (VIMC) was established to estimate the health impact of vaccination. Methods We describe the methods implemented by the VIMC to estimate impact by calendar year, birth year and year of vaccination (YoV). The calendar and birth year methods estimate impact in a particular year and over the lifetime of a particular birth cohort, respectively. The YoV method estimates the impact of a particular year’s vaccination activities through the use of impact ratios which have no stratification and stratification by activity type and/or birth cohort. Furthermore, we detail an impact extrapolation (IE) method for use between coverage scenarios. We compare the methods, focusing on YoV for hepatitis B, measles and yellow fever. Results We find that the YoV methods estimate similar impact with routine vaccinations but have greater yearly variation when campaigns occur with the birth cohort stratification. The IE performs well for the YoV methods, providing a time-efficient mechanism for updates to impact estimates. Conclusions These methods provide a robust set of approaches to quantify vaccination impact; however it is vital that the area of impact estimation continues to develop in order to capture the full effect of immunisation.
Collapse
Affiliation(s)
- Susy Echeverria-Londono
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Xiang Li
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Jaspreet Toor
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Margaret J de Villiers
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Shevanthi Nayagam
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Timothy B Hallett
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Kaja Abbas
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Mark Jit
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Petra Klepac
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, WC1E 7HT, UK
| | - Kévin Jean
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK.,Laboratoire MESuRS, Conservatoire national des Arts et Métiers, Paris, France.,Unité PACRI, Institut Pasteur, Conservatoire national des Arts et Métiers, Paris, France
| | - Tini Garske
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Neil M Ferguson
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK
| | - Katy A M Gaythorpe
- MRC Centre for Global Infectious Disease Analysis, Jameel Institute, School of Public Health, Imperial College London, London, UK.
| |
Collapse
|
47
|
Perkins TA, Huber JH, Tran QM, Oidtman RJ, Walters MK, Siraj AS, Moore SM. Burden is in the eye of the beholder: Sensitivity of yellow fever disease burden estimates to modeling assumptions. SCIENCE ADVANCES 2021; 7:eabg5033. [PMID: 34644110 DOI: 10.1126/sciadv.abg5033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Estimates of disease burden are important for setting public health priorities. These estimates involve numerous modeling assumptions, whose uncertainties are not always well described. We developed a framework for estimating the burden of yellow fever in Africa and evaluated its sensitivity to modeling assumptions that are often overlooked. We found that alternative interpretations of serological data resulted in a nearly 20-fold difference in burden estimates (range of central estimates, 8.4 × 104 to 1.5 × 106 deaths in 2021–2030). Uncertainty about the vaccination status of serological study participants was the primary driver of this uncertainty. Even so, statistical uncertainty was even greater than uncertainty due to modeling assumptions, accounting for a total of 87% of variance in burden estimates. Combined with estimates that most infections go unreported (range of 95% credible intervals, 99.65 to 99.99%), our results suggest that yellow fever’s burden will remain highly uncertain without major improvements in surveillance.
Collapse
Affiliation(s)
- T Alex Perkins
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - John H Huber
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Quan M Tran
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rachel J Oidtman
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Magdalene K Walters
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Amir S Siraj
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Sean M Moore
- Department of Biological Sciences and Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
48
|
Dong HL, Wang HJ, Liu ZY, Ye Q, Qin XL, Li D, Deng YQ, Jiang T, Li XF, Qin CF. Visualization of yellow fever virus infection in mice using a bioluminescent reporter virus. Emerg Microbes Infect 2021; 10:1739-1750. [PMID: 34379047 PMCID: PMC8425728 DOI: 10.1080/22221751.2021.1967705] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Yellow fever virus (YFV) is a re-emerging flavivirus, which can lead to severe clinical manifestations and high mortality, with no specific antiviral therapies available. The live-attenuated yellow fever vaccine 17D (YF17D) has been widely used for over eighty years. However, the emergence of yellow fever vaccine-associated viscerotropic disease (YFL-AVD) and yellow fever vaccine-associated neurotropic disease (YFL-AND) raised non-negligible concerns. Additionally, the attenuation mechanism of YF17D is still unclear. Thus, the development of convenient models is crucial to understand the mechanisms behind YF17D attenuation and its adverse effects. In this work, we generated a reporter YF17D expressing nano-luciferase (NLuc). In vitro and in vivo characterization demonstrated that the NLuc-YF17D shared similar biological properties with its parental strain and the NLuc activity can reflect viral infectivity reliably. Combined with in vivo bioluminescence imaging, a series of mice models of YF17D infection was established, which will be useful for the evaluation of antiviral medicines and novel vaccine candidates. Especially, we demonstrated that intraperitoneally (i.p.) infection of NLuc-YF17D in type I interferon receptor-deficient mice A129 resulted in outcomes resembling YEL-AVD and YEL-AND, evidenced by viral replication in multiple organs and invasion of the central neuronal system. Finally, in vitro and in vivo assays based on this reporter virus were established to evaluate the antiviral activities of validated antiviral agents. In conclusion, the bioluminescent reporter virus described herein provides a powerful platform to study YF17D attenuation and vaccine-associated diseases as well as to develop novel countermeasures against YFV.
Collapse
Affiliation(s)
- Hao-Long Dong
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Hong-Jiang Wang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, People's Republic of China.,Department of Comprehensive Basic Experiment, The Chinese People's Liberation Army Strategic Support Force Characteristic Medical Center, Beijing, People's Republic of China
| | - Zhong-Yu Liu
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, People's Republic of China.,The Center for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qing Ye
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Xiao-Ling Qin
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, People's Republic of China.,Department of Medicine and Health, Guangxi Vocational and Technical Institute of industry, Nanning, People's Republic of China
| | - Dan Li
- The Center for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yong-Qiang Deng
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Tao Jiang
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, People's Republic of China
| | - Xiao-Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, People's Republic of China.,Department of Pharmacology, Chinese Academy of Medical Sciences, Beijing, Republic of China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, People's Republic of China.,Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
49
|
Hansen CA, Barrett ADT. The Present and Future of Yellow Fever Vaccines. Pharmaceuticals (Basel) 2021; 14:ph14090891. [PMID: 34577591 PMCID: PMC8468696 DOI: 10.3390/ph14090891] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 12/05/2022] Open
Abstract
The disease yellow fever (YF) is prevented by a live-attenuated vaccine, termed 17D, which has been in use since the 1930s. One dose of the vaccine is thought to give lifelong (35+ years) protective immunity, and neutralizing antibodies are the correlate of protection. Despite being a vaccine-preventable disease, YF remains a major public health burden, causing an estimated 109,000 severe infections and 51,000 deaths annually. There are issues of supply and demand for the vaccine, and outbreaks in 2016 and 2018 resulted in fractional dosing of the vaccine to meet demand. The World Health Organization (WHO) has established the “Eliminate Yellow Fever Epidemics” (EYE) initiative to reduce the burden of YF over the next 10 years. As with most vaccines, the WHO has recommendations to assure the quality, safety, and efficacy of the YF vaccine. These require the use of live 17D vaccine only produced in embryonated chicken eggs, and safety evaluated in non-human primates only. Thus, any second-generation vaccines would require modification of WHO recommendations if they were to be used in endemic countries. There are multiple second-generation YF vaccine candidates in various stages of development that must be shown to be non-inferior to the current 17D vaccine in terms of safety and immunogenicity to progress through clinical trials to potential licensing. The historic 17D vaccine continues to shape the global vaccine landscape in its use in the generation of multiple licensed recombinant chimeric live vaccines and vaccine candidates, in which its structural protein genes are replaced with those of other viruses, such as dengue and Japanese encephalitis. There is no doubt that the YF 17D live-attenuated vaccine will continue to play a role in the development of new vaccines for YF, as well as potentially for many other pathogens.
Collapse
Affiliation(s)
- Clairissa A. Hansen
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-4036, USA;
| | - Alan D. T. Barrett
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-4036, USA;
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555-4036, USA
- Correspondence:
| |
Collapse
|
50
|
Park S, Tae H, Cho NJ. Biophysical Measurement Strategies for Antiviral Drug Development: Recent Progress in Virus-Mimetic Platforms Down to the Single Particle Level. Acc Chem Res 2021; 54:3204-3214. [PMID: 34346210 DOI: 10.1021/acs.accounts.1c00300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The rapid growth in the global human population has increased the prevalence of emerging infectious diseases, which poses a major risk to public health. In search of effective clinical solutions, the acquisition of knowledge and understanding of biomolecular processes associated with viral pathogens represents a prerequisite. In this context, biophysical engineering approaches are particularly promising since they can resolve biomolecular interactions systematically by circumventing the complexities associated with experiments involving natural biological systems. The engineering approaches encompass the design and construction of biomimetic platforms that simulate the physiological system. This approach enables us to characterize, measure, and quantitatively analyze biomolecular interactions.In this Account, we summarize biophysical measurements that our group has successfully adopted to develop broad-spectrum antiviral drugs based on the lipid envelope antiviral disruption (LEAD) strategy, targeting the structural integrity of the outer viral membrane to abrogate viral infectivity. We particularly focus on the engineering aspects related to the design and construction of the tethered lipid vesicle platform, which closely mimics the viral membrane. We first outline the development of the LEAD agents screening platform that integrates soft matter design components with biomaterials and surface functionalization strategies to facilitate parallel measurements tracking peptide-induced destabilization of nanoscale, virus-mimicking vesicles with tunable size and composition. Then, we describe how this platform can be effectively employed to gain insights into the membrane curvature dependency of certain peptides. The fundamental knowledge acquired through this systematic process is crucial in the identification and subsequent development of antiviral drug candidates. In particular, we highlight the development of curvature-sensitive α-helical (AH) peptides as a broad-spectrum antiviral agent that has been demonstrated as an effective therapeutic treatment against multiple enveloped viruses. Also, we introduce a tethered cluster of vesicles to mimic clusters of enveloped viruses, exhibiting higher infectivity levels in the biological system. Then, we discuss key considerations, including experimental artifacts, namely dye leakage and imaging-related photobleaching, and corresponding corrective measures to improve the accuracy of quantitative interpretation. With the ongoing development and application of the tethered lipid vesicle platform, there is a compelling opportunity to explore fundamental biointerfacial science and develop a new class of broad-spectrum antiviral agents to prepare for the future membrane-enveloped viral pandemics.
Collapse
Affiliation(s)
- Soohyun Park
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Hyunhyuk Tae
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore
| |
Collapse
|